Network


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

Hotspot


Dive into the research topics where Amanda L. Willis is active.

Publication


Featured researches published by Amanda L. Willis.


Journal of Cell Biology | 2013

Physical limits of cell migration: Control by ECM space and nuclear deformation and tuning by proteolysis and traction force

Katarina Wolf; Mariska te Lindert; Marina Krause; Stephanie Alexander; Joost te Riet; Amanda L. Willis; Robert M. Hoffman; Carl G. Figdor; Stephen J. Weiss; Peter Friedl

The physical limits of cell migration in dense porous environments are dependent upon the available space and the deformability of the nucleus and are modulated by matrix metalloproteinases, integrins and actomyosin function.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Estrogen induces apoptosis in estrogen deprivation-resistant breast cancer through stress responses as identified by global gene expression across time

Eric A. Ariazi; Heather E. Cunliffe; Joan S. Lewis-Wambi; Michael Slifker; Amanda L. Willis; Pilar Ramos; Coya Tapia; Helen R. Kim; Smitha Yerrum; Emmanuelle Nicolas; Yoganand Balagurunathan; Eric A. Ross; V. Craig Jordan

In laboratory studies, acquired resistance to long-term antihormonal therapy in breast cancer evolves through two phases over 5 y. Phase I develops within 1 y, and tumor growth occurs with either 17β-estradiol (E2) or tamoxifen. Phase II resistance develops after 5 y of therapy, and tamoxifen still stimulates growth; however, E2 paradoxically induces apoptosis. This finding is the basis for the clinical use of estrogen to treat advanced antihormone-resistant breast cancer. We interrogated E2-induced apoptosis by analysis of gene expression across time (2–96 h) in MCF-7 cell variants that were estrogen-dependent (WS8) or resistant to estrogen deprivation and refractory (2A) or sensitive (5C) to E2-induced apoptosis. We developed a method termed differential area under the curve analysis that identified genes uniquely regulated by E2 in 5C cells compared with both WS8 and 2A cells and hence, were associated with E2-induced apoptosis. Estrogen signaling, endoplasmic reticulum stress (ERS), and inflammatory response genes were overrepresented among the 5C-specific genes. The identified ERS genes indicated that E2 inhibited protein folding, translation, and fatty acid synthesis. Meanwhile, the ERS-associated apoptotic genes Bcl-2 interacting mediator of cell death (BIM; BCL2L11) and caspase-4 (CASP4), among others, were induced. Evaluation of a caspase peptide inhibitor panel showed that the CASP4 inhibitor z-LEVD-fmk was the most active at blocking E2-induced apoptosis. Furthermore, z-LEVD-fmk completely prevented poly (ADP-ribose) polymerase (PARP) cleavage, E2-inhibited growth, and apoptotic morphology. The up-regulated proinflammatory genes included IL, IFN, and arachidonic acid-related genes. Functional testing showed that arachidonic acid and E2 interacted to superadditively induce apoptosis. Therefore, these data indicate that E2 induced apoptosis through ERS and inflammatory responses in advanced antihormone-resistant breast cancer.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Canonical Wnt suppressor, Axin2, promotes colon carcinoma oncogenic activity

Zhao Qiu Wu; Thomas Brabletz; Eric R. Fearon; Amanda L. Willis; Casey Yuexian Hu; Xiao Yan Li; Stephen J. Weiss

Aberrant activation of canonical Wingless-type MMTV integration site family (Wnt) signaling is pathognomonic of colorectal cancers (CRC) harboring functional mutations in either adenomatous polyposis coli or β-catenin. Coincident with Wnt cascade activation, CRCs also up-regulate the expression of Wnt pathway feedback inhibitors, particularly the putative tumor suppressor, Axin2. Because Axin2 serves as a negative regulator of canonical Wnt signaling in normal cells, recent attention has focused on the utility of increasing Axin2 levels in CRCs as a means to slow tumor progression. However, rather than functioning as a tumor suppressor, we demonstrate that Axin2 acts as a potent promoter of carcinoma behavior by up-regulating the activity of the transcriptional repressor, Snail1, inducing a functional epithelial-mesenchymal transition (EMT) program and driving metastatic activity. Silencing Axin2 expression decreases Snail1 activity, reverses EMT, and inhibits CRC invasive and metastatic activities in concert with global effects on the Wnt-regulated cancer cell transcriptome. The further identification of Axin2 and nuclear Snail1 proteins at the invasive front of human CRCs supports a revised model wherein Axin2 acts as a potent tumor promoter in vivo.


Molecular Cancer Research | 2008

The Fibroblast Growth Factor–Inducible 14 Receptor Is Highly Expressed in HER2-Positive Breast Tumors and Regulates Breast Cancer Cell Invasive Capacity

Amanda L. Willis; Nhan L. Tran; Julie M. Chatigny; Nichole Charlton; Hong Vu; Sharron A.N. Brown; Michael A. Black; Wendy S. McDonough; Shannon P. Fortin; Joshua R. Niska; Jeffrey A. Winkles; Heather E. Cunliffe

Genomic characterization is beginning to define a molecular taxonomy for breast cancer; however, the molecular basis of invasion and metastasis remains poorly understood. We report a pivotal role for the fibroblast growth factor–inducible 14 (Fn14) receptor in this process. We examined whether Fn14 and its ligand tumor necrosis factor–like weak inducer of apoptosis (TWEAK) were expressed in breast tumors and whether deregulation of Fn14 levels affected malignant behavior of breast cancer cell lines. Analysis of TWEAK and Fn14 in publicly available gene expression data indicated that high Fn14 expression levels significantly correlated with several poor prognostic indicators (P < 0.05). Fn14 expression was highest in the HER2-positive/estrogen receptor–negative (HER2+/ER−) intrinsic subtype (P = 0.0008). An association between Fn14 and HER2 expression in breast tumors was confirmed by immunohistochemistry. Fn14 levels were elevated in invasive, ER− breast cancer cell lines. Overexpression of Fn14 in weakly invasive MCF7 and T47D cells resulted in a marked induction of invasion and activation of nuclear factor-κB (NF-κB) signaling. Ectopic expression of Fn14tCT, a Fn14 deletion mutant that cannot activate NF-κB signaling, was not able to induce invasion. Moreover, ectopic expression of Fn14tCT in highly invasive MDA-MB-231 cells reduced their invasive capability. RNA interference–mediated inhibition of Fn14 expression in both MDA-MB-231 and MDA-MB-436 cells reduced invasion. Expression profiling of the Fn14-depleted cells revealed deregulation of NF-κB activity. Our findings support a role for Fn14-mediated NF-κB pathway activation in breast tumor invasion and metastasis. (Mol Cancer Res 2008;6(5):725–34)


European Journal of Cancer | 2008

Overexpression of CEACAM6 promotes migration and invasion of oestrogen-deprived breast cancer cells

Joan S. Lewis-Wambi; Heather E. Cunliffe; Helen R. Kim; Amanda L. Willis; V. Craig Jordan

Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) is an intercellular adhesion molecule that is overexpressed in a wide variety of human cancers, including colon, breast and lung and is associated with tumourigenesis, tumour cell adhesion, invasion and metastasis. In this study, we showed that CEACAM6 was overexpressed in a panel of oestrogen receptor (ERalpha)-positive human breast cancer cell lines (MCF-7:5C and MCF-7:2A) that have acquired resistance to oestrogen deprivation, and this overexpression was associated with a more aggressive invasive phenotype in vitro. Expression array analysis revealed that MCF-7:5C and MCF-7:2A cells overexpressed CEACAM6 mRNA by 27-fold and 12-fold, respectively, and were 6-15-times more invasive compared to non-invasive wild-type MCF-7 cells which expressed low levels of CEACAM6. Suppression of CEACAM6 expression using small interfering RNA (siRNA) completely reversed migration and invasion of MCF-7:5C and MCF-7:2A cells and it significantly reduced phosphorylated Akt and c-Src expression in these cells. In conclusion, our findings establish CEACAM6 as a unique mediator of migration and invasion of drug resistant oestrogen-deprived breast cancer cells and suggest that this protein could be an important biomarker of metastasis.


Nature Communications | 2014

Snail1-dependent control of embryonic stem cell pluripotency and lineage commitment

Yongshun Lin; Xiao Yan Li; Amanda L. Willis; Chengyu Liu; Guokai Chen; Stephen J. Weiss

Embryonic stem cells (ESCs) exhibit the dual properties of self-renewal and pluripotency as well as the ability to undergo differentiation that gives rise to all three germ layers. Wnt family members can both promote ESC maintenance and trigger differentiation while also controlling the expression of Snail1, a zinc-finger transcriptional repressor. Snail1 has been linked to events ranging from cell cycle regulation and cell survival to epithelial-mesenchymal transition (EMT) and gastrulation, but its role in self-renewal, pluripotency or lineage commitment in ESCs remains undefined. Here we demonstrate using isogenic pairs of conditional knockout mouse ESCs, that Snail1 exerts Wnt- and EMT independent control over the stem cell transcriptome without affecting self-renewal or pluripotency-associated functions. By contrast, during ESC differentiation, an endogenous Wnt-mediated burst in Snail1 expression regulates neuroectodermal fate while playing a required role in epiblast stem cell exit and the consequent lineage fate decisions that define mesoderm commitment.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2011

Pulmonary fibroblasts mobilize the membrane-tethered matrix metalloprotease, MT1-MMP, to destructively remodel and invade interstitial type I collagen barriers

R. Grant Rowe; Daniel Keena; Farideh Sabeh; Amanda L. Willis; Stephen J. Weiss

In acute and chronic lung disease, widespread disruption of tissue architecture underlies compromised pulmonary function. Pulmonary fibroblasts have been implicated as critical effectors of tissue-destructive extracellular matrix (ECM) remodeling by mobilizing a spectrum of proteolytic enzymes. Although efforts to date have focused on the catabolism of type I collagen, the predominant component of the lung interstitial matrix, the key collagenolytic enzymes employed by pulmonary fibroblasts remain unidentified. Herein, membrane type-1 matrix metalloprotease (MT1-MMP) is identified as the dominant and direct-acting protease responsible for the type I collagenolytic activity mediated by both mouse and human pulmonary fibroblasts. Furthermore, MT1-MMP is shown to be essential for pulmonary fibroblast migration within three-dimensional (3-D) hydrogels of cross-linked type I collagen that recapitulate ECM barriers encountered in the in vivo environment. Together, these findings demonstrate that MT1-MMP serves as a key effector of type I collagenolytic activity in pulmonary fibroblasts and earmark this pericellular collagenase as a potential target for therapeutic intervention.


Clinical Cancer Research | 2013

Inactivation or loss of TTP promotes invasion in head and neck cancer via transcript stabilization and secretion of MMP9, MMP2 and IL-6

Elizabeth Van Tubergen; Rajat Banerjee; Min Liu; Robert Vander Broek; Emily Light; Shiuhyang Kuo; Stephen E. Feinberg; Amanda L. Willis; Gregory T. Wolf; Thomas E. Carey; Carol R. Bradford; Mark E. Prince; Francis P. Worden; Keith L. Kirkwood; Nisha J. D'Silva

Purpose: Invasion is the critical step in progression of a precancerous lesion to squamous cell carcinoma of the head and neck (HNSCC). Invasion is regulated by multiple proinflammatory mediators. Tristetraprolin (TTP) is an mRNA-degrading protein that regulates multiple proinflammatory mediators. TTP may serve as an excellent treatment target. Rap1 is a ras-like oncoprotein that induces critical signaling pathways. In this study, the role of rap1 in TTP-mediated invasion was investigated. Experimental Design: Using complementary approaches, we modulated TTP and altered expression of interleukin (IL)-6 and matrix metalloproteinase (MMP) 2/9, which were quantified by ELISA and zymogram. Invasion was evaluated in vitro using the oral-cancer-equivalent (OCE) three-dimensional model and in vivo in the chick chorioallantoic membrane (CAM). The role of rap1 and p38 were established using knockdown strategies. Results: Downregulation of TTP significantly increased invasion via secretion of MMP9/2 and IL-6. In the novel OCE and CAM invasion models of HNSCC, cells with downregulated TTP destroyed the basement membrane to invade the underlying connective tissue. Rap1 induces p38 mitogen-activated protein kinase (p38)-mediated inactivation of TTP. Inactive TTP enhances transcript stability via binding to the 3′-untranslated region (UTR). High IL-6 and MMP9 are prognostic for poor clinical outcomes in patients with HNSCC. Conclusions: Targeting the rap1-p38-TTP cascade is an attractive novel treatment strategy in HNSCC to concurrently suppress multiple mediators of invasion. Clin Cancer Res; 19(5); 1169–79. ©2012 AACR.


Journal of the American Chemical Society | 2013

Coupling protein engineering with probe design to inhibit and image matrix metalloproteinases with controlled specificity.

Montse Morell; Thinh Nguyen Duc; Amanda L. Willis; Salahuddin Syed; Jiyoun Lee; Edgar Deu; Yang Deng; Junpeng Xiao; Benjamin E. Turk; Jason R. Jessen; Stephen J. Weiss; Matthew Bogyo

Matrix metalloproteinases (MMPs) are zinc endopeptidases that play roles in numerous pathophysiological processes and therefore are promising drug targets. However, the large size of this family and a lack of highly selective compounds that can be used for imaging or inhibition of specific MMPs members has limited efforts to better define their biological function. Here we describe a protein engineering strategy coupled with small-molecule probe design to selectively target individual members of the MMP family. Specifically, we introduce a cysteine residue near the active-site of a selected protease that does not alter its overall activity or function but allows direct covalent modification by a small-molecule probe containing a reactive electrophile. This specific engineered interaction between the probe and the target protease provides a means to both image and inhibit the modified protease with absolute specificity. Here we demonstrate the feasibility of the approach for two distinct MMP proteases, MMP-12 and MT1-MMP (or MMP-14).


Cancer Research | 2010

Abstract 1174: Mechanisms of Fn14 overexpression in invasive breast tumors

Joshua R. Niska; Amanda L. Willis; Pilar Ramos; Catherine M. Mancini; Julie E. Getz; Heather E. Cunliffe

The fibroblast growth factor-inducible 14 (Fn14) signaling axis plays an important role in the regulation of breast cancer cell invasion. Breast tumors express Fn14 whereas normal mammary epithelium does not, and Fn14 expression correlates with poor prognostic indicators. The molecular mechanisms driving aberrant Fn14 expression in breast cancer have not yet been determined. This investigation aims to elucidate the mechanisms underlying Fn14 over-expression. Soluble tumor necrosis factor-like weak inducer of apoptosis (TWEAK), the cognate ligand for Fn14, was found to induce Fn14 expression in some breast cancer cell lines via a proposed autoregulatory mechanism. We also provide evidence to support the likelihood that membrane-bound TWEAK can stimulate Fn14 expression through adjacent tumor-cell interaction. EGF and a variety of other receptor tyrosine kinase ligands known to play a role in breast tumor progression stimulated de novo Fn14 expression in a panel of breast cancer cell lines. We therefore hypothesized that Fn14 expression may be induced by aberrant activation of the MAPK signaling pathway. Using an MCF7 model of TPA-mediated MAPK pathway activation and in vitro invasion using Matrigel-coated Transwell Boyden chambers, we observed TPA-mediated expression of Fn14 concomitant with a dramatic increase in cell invasion. Fn14 expression and invasion in this model were abrogated in the presence of U0126, a specific inhibitor of Mek1/2, implicating a role for MAPK signaling in regulation of Fn14 expression. From previously published microarray-based surveys of genomic aberrations in breast cancer, we also hypothesized that constitutive Fn14 expression may be due to Fn14 gene amplification. We performed fluorescence in situ hybridization analysis of 53 breast tumors and identified 15/53 (28%) tumors with additional copies of Fn14 relative to centromere 16. Fn14 was not amplified in normal breast epithelium or in Fn14-negative cell lines. We conclude that deregulation of Fn14 expression can occur through diverse molecular mechanisms associated with breast tumor progression. A more complete definition of the spectrum of molecular aberrations leading to induction of Fn14 expression in breast cancer requires further studies. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1174.

Collaboration


Dive into the Amanda L. Willis's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pilar Ramos

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Helen R. Kim

Fox Chase Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Catherine M. Mancini

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Coya Tapia

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric A. Ross

Fox Chase Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge