Network


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

Hotspot


Dive into the research topics where Judith A. Varner is active.

Publication


Featured researches published by Judith A. Varner.


Science | 1995

Definition of Two Angiogenic Pathways by Distinct αv Integrins

Martin Friedlander; Peter C. Brooks; Robert W. Shaffer; Christine M. Kincaid; Judith A. Varner; David A. Cheresh

Angiogenesis depends on cytokines and vascular cell adhesion events. Two cytokine-dependent pathways of angiogenesis were shown to exist and were defined by their dependency on distinct vascular cell integrins. In vivo angiogenesis in corneal or chorioallantoic membrane models induced by basic fibroblast growth factor or by tumor necrosis factor-α depended on αvβ3, whereas angiogenesis initiated by vascular endothelial growth factor, transforming growth factor-α, or phorbol ester depended on αvβ5. Antibody to each integrin selectively blocked one of these pathways, and a cyclic peptide antagonist of both integrins blocked angiogenesis stimulated by each cytokine tested. These pathways are further distinguished by their sensitivity to calphostin C, an inhibitor of protein kinase C that blocked angiogenesis potentiated by αvβ5 but not by αvβ3.


Nature Reviews Cancer | 2008

Integrins in angiogenesis and lymphangiogenesis

Christie J. Avraamides; Barbara Garmy-Susini; Judith A. Varner

Blood vessels promote tumour growth, and both blood and lymphatic vessels facilitate tumour metastasis by serving as conduits for the transport of tumour cells to new sites. Angiogenesis and lymphangiogenesis are regulated by integrins, which are members of a family of cell surface receptors whose ligands are extracellular matrix proteins and immunoglobulin superfamily molecules. Select integrins promote endothelial cell migration and survival during angiogenesis and lymphangiogenesis, whereas other integrins promote pro-angiogenic macrophage trafficking to tumours. Several integrin-targeted therapeutic agents are currently in clinical trials for cancer therapy. Here, we review the evidence implicating integrins as a family of fundamental regulators of angiogenesis and lymphangiogenesis.


American Journal of Pathology | 2000

Regulation of Angiogenesis in Vivo by Ligation of Integrin α5β1 with the Central Cell-Binding Domain of Fibronectin

Semi Kim; Kelly Bell; Shaker A. Mousa; Judith A. Varner

Angiogenesis depends on the cooperation of growth factors and cell adhesion events. Although αv integrins have been shown to play critical roles in angiogenesis, recent studies in αv-null mice suggest that other adhesion receptors and their ligands also regulate this process. Evidence is now provided that the integrin α5β1 and its ligand fibronectin are coordinately up-regulated on blood vessels in human tumor biopsies and play critical roles in angiogenesis, resulting in tumor growth in vivo. Angiogenesis induced by multiple growth factors in chick embryos was blocked by monoclonal antibodies to the cell-binding domain of fibronectin. Furthermore, application of fibronectin or a proteolytic fragment of fibronectin containing the central cell-binding domain to the chick chorioallantoic membrane enhanced angiogenesis in an integrin α5β1-dependent manner. Importantly, antibody, peptide, and novel nonpeptide antagonists of integrin α5β1 blocked angiogenesis induced by several growth factors but had little effect on angiogenesis induced by vascular endothelial growth factor (VEGF) in both chick embryo and murine models. In fact, these α5β1 antagonists inhibited tumor angiogenesis, thereby causing regression of human tumors in animal models. Thus, fibronectin and integrin α5β1, like integrin αvβ3, contribute to an angiogenesis pathway that is distinct from VEGF-mediated angiogenesis, yet important for the growth of tumors.


Current Opinion in Cell Biology | 1996

Integrins and cancer

Judith A. Varner; David A. Cheresh

The past year or two has seen great advances in the elucidation of significant roles for integrins in cancer cells. These include roles in signal transduction, gene expression, proliferation, apoptosis regulation, invasion and metastasis, and angiogenesis. In particular, integrin alphavbeta3 has been implicated in the neovascularization of tumors. In addition, this integrin has been shown to contribute to the survival, proliferation and metastatic phenotype of human melanoma.


Journal of Biological Chemistry | 2000

Regulation of Integrin αvβ3-mediated Endothelial Cell Migration and Angiogenesis by Integrin α5β1 and Protein Kinase A

Semi Kim; Marina Harris; Judith A. Varner

Recent studies indicate that angiogenesis depends, in part, on ligation of integrin α5β1 by fibronectin. Evidence is now provided that integrin α5β1 regulates the function of integrin αvβ3 on endothelial cells during their migration in vitro or angiogenesisin vivo. Secretion of fibronectin by endothelial cells leads to the ligation of integrin α5β1, which potentiates αvβ3-mediated migration on vitronectin without influencing αvβ3-mediated cell adhesion. Endothelial cell attachment to vitronectin suppresses protein kinase A (PKA) activity, while addition of soluble anti-α5β1 restores this activity. Moreover, agents that activate intracellular PKA, such as forskolin, dibutyryl cAMP or α5β1 antagonists, suppress endothelial cell migration on vitronectin in vitro or angiogenesis in vivo. In contrast, inhibitors of PKA reverse the anti-migratory or anti-angiogenic effects mediated by α5β1 antagonists. Therefore, αvβ3-mediated endothelial cell migration and angiogenesis can be regulated by PKA activity, which depends on the ligation state of integrin α5β1.


Cancer Cell | 2011

Receptor tyrosine kinases and TLR/IL1Rs unexpectedly activate myeloid cell PI3Kγ, a single convergent point promoting tumor inflammation and progression

Michael C. Schmid; Christie J. Avraamides; Holly C. Dippold; Irene Franco; Philippe Foubert; Lesley G. Ellies; Lissette M. Acevedo; Joan R.E. Manglicmot; Xiaodan Song; Wolfgang Wrasidlo; Sara L. Blair; Mark H. Ginsberg; David A. Cheresh; Emilio Hirsch; Seth J. Field; Judith A. Varner

Tumor inflammation promotes angiogenesis, immunosuppression, and tumor growth, but the mechanisms controlling inflammatory cell recruitment to tumors are not well understood. We found that a range of chemoattractants activating G protein-coupled receptors (GPCRs), receptor tyrosine kinases (RTKs) and Toll-like/IL-1 receptors (TLR/IL1Rs) unexpectedly initiate tumor inflammation by activating the PI3-kinase isoform p110γ in Gr1+CD11b+ myeloid cells. Whereas GPCRs activate p110γ in a Ras/p101-dependent manner, RTKs and TLR/IL1Rs directly activate p110γ in a Ras/p87-dependent manner. Once activated, p110γ promotes inside-out activation of a single integrin, α4β1, causing myeloid cell invasion into tumors. Pharmacological or genetic blockade of p110γ suppressed inflammation, growth, and metastasis of implanted and spontaneous tumors, revealing an important therapeutic target in oncology.


Journal of Biological Chemistry | 1999

Del1 Induces Integrin Signaling and Angiogenesis by Ligation of αVβ3

Kalyani Penta; Judith A. Varner; Lucy Liaw; Chiaki Hidai; Randy Schatzman; Thomas Quertermous

Del1 is a novel extracellular matrix protein encoding three Notch-like epidermal growth factor repeats, an RGD motif, and two discoidin domains. Del1 is expressed in an endothelial cell-restricted pattern during early development. In studies reported here, recombinant baculovirus Del1 protein was shown to promote αvβ3-dependent endothelial cell attachment and migration. Attachment of endothelial cells to Del1 was associated with clustering of αvβ3, the formation of focal complexes, and recruitment of talin and vinculin into these complexes. These events were shown to be associated with phosphorylation of proteins in the focal complexes, including the time-dependent phosphorylation of p125FAK, MAPK, and Shc. When recombinant Del1 was evaluated in an in ovo chick chorioallantoic membrane assay, it was found to have potent angiogenic activity. This angiogenic activity was inhibited by a monoclonal antibody directed against αvβ3, and an RAD mutant Del1 protein was inactive. Thus Del1 provides a unique autocrine angiogenic pathway for the embryonic endothelium, and this function is mediated in part by productive ligation of integrin αvβ3.


Journal of Clinical Investigation | 2002

Inhibition of endothelial cell survival and angiogenesis by protein kinase A

Semi Kim; Manjiri Bakre; Hong Yin; Judith A. Varner

Receptors for the provisional ECM are important regulators of angiogenesis. One of these receptors, integrin alpha5beta1, plays a critical role in tumor- and growth factor-induced angiogenesis, because antagonists of this integrin potently inhibit angiogenesis and tumor growth. Here we show that the integrin alpha5beta1 promotes endothelial cell survival during angiogenesis in vivo by suppressing the activity of protein kinase A (PKA). Antagonists of integrin alpha5beta1 activate PKA, which then leads to the activation of caspase-8 and induction of apoptosis. Direct activation of PKA by cAMP or by expression of the PKA catalytic subunit also induces endothelial cell apoptosis, resulting in angiogenesis inhibition in vivo. Our studies indicate that ligation of integrin alpha5beta1 during angiogenesis suppresses an apoptotic program that is dependent on PKA. These studies also indicate that induction of endothelial cell apoptosis in vivo by genetic or pharmacological activation of PKA may be a useful strategy to inhibit angiogenesis.


Nature | 2016

PI3Kγ is a molecular switch that controls immune suppression

Megan M. Kaneda; Karen Messer; Natacha Ralainirina; Hongying Li; Christopher J. Leem; Sara Gorjestani; Gyunghwi Woo; Abraham V. Nguyen; Camila C. Figueiredo; Philippe Foubert; Michael C. Schmid; Melissa Pink; David G. Winkler; Matthew Rausch; Vito J. Palombella; Jeffery L. Kutok; Karen McGovern; Kelly A. Frazer; Xuefeng Wu; Michael Karin; Roman Sasik; Ezra E.W. Cohen; Judith A. Varner

Macrophages play critical, but opposite, roles in acute and chronic inflammation and cancer. In response to pathogens or injury, inflammatory macrophages express cytokines that stimulate cytotoxic T cells, whereas macrophages in neoplastic and parasitic diseases express anti-inflammatory cytokines that induce immune suppression and may promote resistance to T cell checkpoint inhibitors. Here we show that macrophage PI 3-kinase γ controls a critical switch between immune stimulation and suppression during inflammation and cancer. PI3Kγ signalling through Akt and mTor inhibits NFκB activation while stimulating C/EBPβ activation, thereby inducing a transcriptional program that promotes immune suppression during inflammation and tumour growth. By contrast, selective inactivation of macrophage PI3Kγ stimulates and prolongs NFκB activation and inhibits C/EBPβ activation, thus promoting an immunostimulatory transcriptional program that restores CD8+ T cell activation and cytotoxicity. PI3Kγ synergizes with checkpoint inhibitor therapy to promote tumour regression and increased survival in mouse models of cancer. In addition, PI3Kγ-directed, anti-inflammatory gene expression can predict survival probability in cancer patients. Our work thus demonstrates that therapeutic targeting of intracellular signalling pathways that regulate the switch between macrophage polarization states can control immune suppression in cancer and other disorders.


Cancer Research | 2010

Integrin α4β1 Signaling Is Required for Lymphangiogenesis and Tumor Metastasis

Barbara Garmy-Susini; Christie J. Avraamides; Michael C. Schmid; Philippe Foubert; Lesley G. Ellies; Leo Barnes; Chloé C. Féral; Thalia Papayannopoulou; Andrew M. Lowy; Sarah L. Blair; David A. Cheresh; Mark H. Ginsberg; Judith A. Varner

Recent studies have shown that lymphangiogenesis or the growth of lymphatic vessels at the periphery of tumors promotes tumor metastasis to lymph nodes. We show here that the fibronectin-binding integrin alpha4beta1 and its ligand fibronectin are novel functional markers of proliferative lymphatic endothelium. Tumors and lymphangiogenic growth factors, such as vascular endothelial growth factor-C (VEGF-C) and VEGF-A, induce lymphatic vessel expression of integrin alpha4beta1. Integrin alpha4beta1 then promotes growth factor and tumor-induced lymphangiogenesis, as genetic loss of integrin alpha4beta1 expression in Tie2Cre+ alpha4(loxp/loxp) mice or genetic loss of alpha4 signaling in alpha4Y991A knock-in mice blocks growth factor and tumor-induced lymphangiogenesis, as well as tumor metastasis to lymph nodes. In addition, antagonists of integrin alpha4beta1 suppress lymphangiogenesis and tumor metastasis. Our studies show that integrin alpha4beta1 and the signals it transduces regulate the adhesion, migration, invasion, and survival of proliferating lymphatic endothelial cells. As suppression of alpha4beta1 expression, signal transduction, or function in tumor lymphatic endothelium not only inhibits tumor lymphangiogenesis but also prevents metastatic disease, these results show that integrin alpha4beta1-mediated tumor lymphangiogenesis promotes metastasis and is a useful target for the suppression of metastatic disease.

Collaboration


Dive into the Judith A. Varner's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew M. Lowy

University of California

View shared research outputs
Top Co-Authors

Avatar

Michael Bouvet

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge