Network


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

Hotspot


Dive into the research topics where Madeleine J. Oudin is active.

Publication


Featured researches published by Madeleine J. Oudin.


Cancer Discovery | 2016

Tumor Cell–Driven Extracellular Matrix Remodeling Drives Haptotaxis during Metastatic Progression

Madeleine J. Oudin; Oliver Jonas; Tatsiana Kosciuk; Liliane C. Broye; Bruna Candido Guido; Jeffrey Wyckoff; Daisy Riquelme; John M. Lamar; Sreeja B. Asokan; Charles A. Whittaker; Duanduan Ma; Robert Langer; Michael J. Cima; Kari B. Wisinski; Richard O. Hynes; Douglas A. Lauffenburger; Patricia J. Keely; James E. Bear; Frank B. Gertler

UNLABELLED Fibronectin (FN) is a major component of the tumor microenvironment, but its role in promoting metastasis is incompletely understood. Here, we show that FN gradients elicit directional movement of breast cancer cells, in vitro and in vivo Haptotaxis on FN gradients requires direct interaction between α5β1 integrin and MENA, an actin regulator, and involves increases in focal complex signaling and tumor cell-mediated extracellular matrix (ECM) remodeling. Compared with MENA, higher levels of the prometastatic MENA(INV) isoform associate with α5, which enables 3-D haptotaxis of tumor cells toward the high FN concentrations typically present in perivascular space and in the periphery of breast tumor tissue. MENA(INV) and FN levels were correlated in two breast cancer cohorts, and high levels of MENA(INV) were significantly associated with increased tumor recurrence as well as decreased patient survival. Our results identify a novel tumor cell-intrinsic mechanism that promotes metastasis through ECM remodeling and ECM-guided directional migration. SIGNIFICANCE Here, we provide new insight into how tumor cell:ECM interactions generate signals and structures that promote directed tumor cell migration, a critical component of metastasis. Our results identify a tumor cell-intrinsic mechanism driven by the actin regulatory protein MENA that promotes ECM remodeling and haptotaxis along FN gradients. Cancer Discov; 6(5); 516-31. ©2016 AACR.See related commentary by Santiago-Medina and Yang, p. 474This article is highlighted in the In This Issue feature, p. 461.


Genes & Development | 2015

Foxa2 and Cdx2 cooperate with Nkx2-1 to inhibit lung adenocarcinoma metastasis

Carman Man-Chung Li; Vasilena Gocheva; Madeleine J. Oudin; Arjun Bhutkar; Shi Yun Wang; Saya R. Date; Sheng Rong Ng; Charles A. Whittaker; Roderick T. Bronson; Eric L. Snyder; Frank B. Gertler; Tyler Jacks

Despite the fact that the majority of lung cancer deaths are due to metastasis, the molecular mechanisms driving metastatic progression are poorly understood. Here, we present evidence that loss of Foxa2 and Cdx2 synergizes with loss of Nkx2-1 to fully activate the metastatic program. These three lineage-specific transcription factors are consistently down-regulated in metastatic cells compared with nonmetastatic cells. Knockdown of these three factors acts synergistically and is sufficient to promote the metastatic potential of nonmetastatic cells to that of naturally arising metastatic cells in vivo. Furthermore, silencing of these three transcription factors is sufficient to account for a significant fraction of the gene expression differences between the nonmetastatic and metastatic states in lung adenocarcinoma, including up-regulated expression of the invadopodia component Tks5long, the embryonal proto-oncogene Hmga2, and the epithelial-to-mesenchymal mediator Snail. Finally, analyses of tumors from a genetically engineered mouse model and patients show that low expression of Nkx2-1, Foxa2, and Cdx2 strongly correlates with more advanced tumors and worse survival. Our findings reveal that a large part of the complex transcriptional network in metastasis can be controlled by a small number of regulatory nodes that function redundantly, and loss of multiple nodes is required to fully activate the metastatic program.


Molecular Biology of the Cell | 2015

PTP1B-dependent regulation of receptor tyrosine kinase signaling by the actin-binding protein Mena.

Shannon K. Hughes; Madeleine J. Oudin; Jenny Tadros; Jason R. Neil; Amanda M. Del Rosario; Brian A. Joughin; Laila Ritsma; Jeff Wyckoff; Eliza Vasile; Robert J. Eddy; Ulrike Philippar; Alisha Lussiez; John Condeelis; Jacco van Rheenen; Forest M. White; Douglas A. Lauffenburger; Frank B. Gertler

The actin-binding protein Mena regulates RTK signaling after growth factor stimulation in tumor cells by a novel mechanism. The alternatively spliced MenaINV isoform disrupts this attenuation to drive sensitivity to growth factors, resistance to targeted inhibitors, and ultimately tumor invasion and metastasis.


Clinical & Experimental Metastasis | 2016

Characterization of the expression of the pro-metastatic Mena INV isoform during breast tumor progression

Madeleine J. Oudin; Shannon K. Hughes; Nazanin Rohani; Mira N. Moufarrej; Joan G. Jones; John Condeelis; Douglas A. Lauffenburger; Frank B. Gertler

Several functionally distinct isoforms of the actin regulatory Mena are produced by alternative splicing during tumor progression. Forced expression of the MenaINV isoform drives invasion, intravasation and metastasis. However, the abundance and distribution of endogenously expressed MenaINV within primary tumors during progression remain unknown, as most studies to date have only assessed relative mRNA levels from dissociated tumor samples. We have developed a MenaINV isoform-specific monoclonal antibody and used it to examine MenaINV expression patterns in mouse mammary and human breast tumors. MenaINV expression increases during tumor progression and to examine the relationship between MenaINV expression and markers for epithelial or mesenchymal status, stemness, stromal cell types and hypoxic regions. Further, while MenaINV robustly expressed in vascularized areas of the tumor, it is not confined to cells adjacent to blood vessels. Altogether, these data demonstrate the specificity and utility of the anti-MenaINV-isoform specific antibody, and provide the first description of endogenous MenaINV protein expression in mouse and human tumors.


Cold Spring Harbor Symposia on Quantitative Biology | 2016

Physical and Chemical Gradients in the Tumor Microenvironment Regulate Tumor Cell Invasion, Migration, and Metastasis

Madeleine J. Oudin; Valerie M. Weaver

Cancer metastasis requires the invasion of tumor cells into the stroma and the directed migration of tumor cells through the stroma toward the vasculature and lymphatics where they can disseminate and colonize secondary organs. Physical and biochemical gradients that form within the primary tumor tissue promote tumor cell invasion and drive persistent migration toward blood vessels and the lymphatics to facilitate tumor cell dissemination. These microenvironment cues include hypoxia and pH gradients, gradients of soluble cues that induce chemotaxis, and ions that facilitate galvanotaxis, as well as modifications to the concentration, organization, and stiffness of the extracellular matrix that produce haptotactic, alignotactic, and durotactic gradients. These gradients form through dynamic interactions between the tumor cells and the resident fibroblasts, adipocytes, nerves, endothelial cells, infiltrating immune cells, and mesenchymal stem cells. Malignant progression results from the integrated response of the tumor to these extrinsic physical and chemical cues. Here, we first describe how these physical and chemical gradients develop, and we discuss their role in tumor progression. We then review assays to study these gradients. We conclude with a discussion of clinical strategies used to detect and inhibit these gradients in tumors and of new intervention opportunities. Clarifying the role of these gradients in tumor evolution offers a unique approach to target metastasis.


Molecular Cancer Therapeutics | 2017

MENA Confers Resistance to Paclitaxel in Triple-Negative Breast Cancer

Madeleine J. Oudin; Lucie Barbier; Claudia Schäfer; Tatsiana Kosciuk; Miles A. Miller; Sangyoon J. Han; Oliver Jonas; Douglas A. Lauffenburger; Frank B. Gertler

Taxane therapy remains the standard of care for triple-negative breast cancer. However, high frequencies of recurrence and progression in treated patients indicate that metastatic breast cancer cells can acquire resistance to this drug. The actin regulatory protein MENA and particularly its invasive isoform, MENAINV, are established drivers of metastasis. MENAINV expression is significantly correlated with metastasis and poor outcome in human patients with breast cancer. We investigated whether MENA isoforms might play a role in driving resistance to chemotherapeutics. We find that both MENA and MENAINV confer resistance to the taxane paclitaxel, but not to the widely used DNA-damaging agents doxorubicin or cisplatin. Furthermore, paclitaxel treatment does not attenuate growth of MENAINV-driven metastatic lesions. Mechanistically, MENA isoform expression alters the ratio of dynamic and stable microtubule populations in paclitaxel-treated cells. MENA expression also increases MAPK signaling in response to paclitaxel treatment. Decreasing ERK phosphorylation by co-treatment with MEK inhibitor restored paclitaxel sensitivity by driving microtubule stabilization in MENA isoform–expressing cells. Our results reveal a novel mechanism of taxane resistance in highly metastatic breast cancer cells and identify a combination therapy to overcome such resistance. Mol Cancer Ther; 16(1); 143–55. ©2016 AACR.


Clinical Cancer Research | 2016

Parallel in-vivo assessment of drug phenotypes at various time points during systemic BRAF inhibition reveals tumor adaptation and altered treatment vulnerabilities

Oliver Jonas; Madeleine J. Oudin; Tatsiana Kosciuk; Matthew Whitman; Frank B. Gertler; Michael J. Cima; Keith T. Flaherty; Robert Langer

Purpose: Treatment of BRAF-mutated melanoma tumors with BRAF inhibitor–based therapy produces high response rates, but of limited duration in the vast majority of patients. Published investigations of resistance mechanisms suggest numerous examples of tumor adaptation and signal transduction bypass mechanisms, but without insight into biomarkers that would predict which mechanism will predominate. Monitoring phenotypic response of multiple adaptive mechanisms simultaneously within the same tumor as it adapts during treatment has been elusive. Experimental Design: This study reports on a method to provide a more complete understanding of adaptive tumor responses. We simultaneously measured in vivo antitumor activity of 12 classes of inhibitors, which are suspected of enabling adaptive escape mechanisms, at various time points during systemic BRAF inhibition. We used implantable microdevices to release multiple compounds into distinct regions of a tumor to measure the efficacy of each compound independently and repeated these measurements as tumors progressed on systemic BRAF treatment. Results: We observed varying phenotypic responses to specific inhibitors before, during, and after prolonged systemic treatment with BRAF inhibitors. Our results specifically identify PI3K, PDGFR, EGFR, and HDAC inhibitors as becoming significantly more efficacious during systemic BRAF inhibition. The sensitivity to other targeted inhibitors remained mostly unchanged, whereas local incremental sensitivity to PLX4720 declined sharply. Conclusions: These findings suggest redundancy of several resistance mechanisms and may help identify optimal constituents of more effective combination therapy in BRAF-mutant melanoma. They also represent a new paradigm for dynamic measurement of adaptive signaling mechanisms within the same tumor during therapy. Clin Cancer Res; 22(24); 6031–8. ©2016 AACR.


Molecular Biology of the Cell | 2016

MenaINV mediates synergistic cross-talk between signaling pathways driving chemotaxis and haptotaxis

Madeleine J. Oudin; Miles A. Miller; Joelle Klazen; Tatsiana Kosciuk; Alisha Lussiez; Shannon K. Hughes; Jenny Tadros; James E. Bear; Douglas A. Lauffenburger; Frank B. Gertler

MenaINV, an actin-regulatory protein known to promote metastasis, has roles in driving cross-talk between extracellular matrix, growth factor cues, and their downstream pathways during tumor cell invasion. MenaINV is a shared component of the signaling pathway driving both EGF chemotaxis and fibronectin haptotaxis.


Trends in cancer | 2017

Signatures of Breast Cancer Metastasis: aMENAble to Interpretation?

Madeleine J. Oudin; Frank B. Gertler

MenaINV, an isoform of the motility regulator protein Mena, contributes to prometastatic phenotypes. Tumor microenvironment of metastasis (TMEM), a three-cell structure associated with intravasation, contains a stationary Mena-expressing tumor cell. TMEM density and MenaINV expression both correlate with poor clinical outcome in breast cancer patients. However, is MenaINV involved in TMEM assembly and function?


Cancer Research | 2016

Abstract PR15: Haptotaxis and direct remodeling of the extracellular matrix by tumor cells is important for metastasis

Madeleine J. Oudin; Oliver Jonas; Tatiana Kosciuk; Liliane C. Broye; Jeff Wyckoff; Joelle Klazen; John M. Lamar; Sreeja B. Asokan; Charlie A. Whittaker; Robert Langer; Michael J. Cima; Kari B. Wisinski; Richard O. Hynes; Douglas A. Lauffenburger; Patricia J. Keely; James E. Bear; Frank B. Gertler

The tissue microenvironment, composed of stromal cells and extracellular matrix (ECM), is known to contribute to tumor progression, by providing both the structure and signals that promote tumor cell proliferation, survival and invasion. Fibronectin and collagen are major components of the tumor microenvironment, however, how tumor cells gain the ability to respond to these substrate-bound cues and how ECM sensing contributes to promoting metastasis remain poorly understood. Here, using a combination of in vitro and in vivo imaging, we show that gradients of FN can promote directional cell motility of breast cancer cells, a process dependent upon the actin regulatory Mena. Expression of the pro-metastatic isoform Mena INV allows cells to haptotax towards very high concentrations of FN, found near blood vessels and tumor periphery. Haptotaxis requires the direct interaction between α5β1 integrin and Mena/ Mena INV and is driven by outside-in signaling at focal complexes, crosstalk between α5β1 and EGFR, and inside-out tumor cell dependent ECM remodeling. These findings are clinically relevant as patients with high levels of Mena INV protein and FN have increased recurrence and decreased survival in two breast cancer cohorts. Our results identify a novel tumor cell-intrinsic mechanism that promotes ECM remodeling and directed migration, ultimately affecting metastasis. Citation Format: Madeleine J. Oudin, Oliver Jonas, Tatiana Kosciuk, Liliane Broye, Jeff Wyckoff, Joelle Klazen, John Lamar, Sreeja Asokan, Charlie Whittaker, Robert Langer, Michael Cima, Kari Wisinski, Richard Hynes, Douglas Lauffenburger, Patricia Keely, James Bear, Frank Gertler. Haptotaxis and direct remodeling of the extracellular matrix by tumor cells is important for metastasis. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Metastasis; 2015 Nov 30-Dec 3; Austin, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(7 Suppl):Abstract nr PR15.

Collaboration


Dive into the Madeleine J. Oudin's collaboration.

Top Co-Authors

Avatar

Frank B. Gertler

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Douglas A. Lauffenburger

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James E. Bear

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Tatsiana Kosciuk

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Alisha Lussiez

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Shannon K. Hughes

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jenny Tadros

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Joelle Klazen

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Oliver Jonas

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge