Network


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

Hotspot


Dive into the research topics where Aaron S. Meyer is active.

Publication


Featured researches published by Aaron S. Meyer.


Science Signaling | 2013

The Receptor AXL Diversifies EGFR Signaling and Limits the Response to EGFR-Targeted Inhibitors in Triple-Negative Breast Cancer Cells

Aaron S. Meyer; Miles A. Miller; Frank B. Gertler; Douglas A. Lauffenburger

Amplification of downstream signaling by AXL receptor tyrosine kinase alters the migratory response of breast cancer cells. Transactivating Resistance Resistance to receptor tyrosine kinase (RTK) inhibitors in cancer is often mediated by the activation of alternate receptors that provide compensatory signaling. Meyer et al. showed that the RTK AXL predicts resistance to EGFR (epidermal growth factor receptor) inhibitors in cancer cell lines. Through a ligand-independent mode of transactivation by EGFR, AXL mediated the downstream activation of proteins in a distinct combination, thereby augmenting EGFR signaling and contributing unique output. Diversified, AXL-mediated signaling was required for EGF (epidermal growth factor)–induced migration of TNBC (triple-negative breast cancer) cells. Expression profiles of AXL and EGFR in cancer cell lines predicted resistance to EGFR inhibitors, and treatment with an AXL inhibitor reduced EGFR-elicited proliferation and migration, suggesting that targeting AXL may improve EGFR-targeted drug therapy in TNBC. The relationship between drug resistance, changes in signaling, and emergence of an invasive phenotype is well appreciated, but the underlying mechanisms are not well understood. Using machine learning analysis applied to the Cancer Cell Line Encyclopedia database, we identified expression of AXL, the gene that encodes the epithelial-to-mesenchymal transition (EMT)–associated receptor tyrosine kinase (RTK) AXL, as exceptionally predictive of lack of response to ErbB family receptor–targeted inhibitors. Activation of EGFR (epidermal growth factor receptor) transactivated AXL, and this ligand-independent AXL activity diversified EGFR-induced signaling into additional downstream pathways beyond those triggered by EGFR alone. AXL-mediated signaling diversification was required for EGF (epidermal growth factor)–elicited motility responses in AXL-positive TNBC (triple-negative breast cancer) cells. Using cross-linking coimmunoprecipitation assays, we determined that AXL associated with EGFR, other ErbB receptor family members, MET (hepatocyte growth factor receptor), and PDGFR (platelet-derived growth factor receptor) but not IGF1R (insulin-like growth factor 1 receptor) or INSR (insulin receptor). From these AXL interaction data, we predicted AXL-mediated signaling synergy for additional RTKs and validated these predictions in cells. This alternative mechanism of receptor activation limits the use of ligand-blocking therapies and indicates against therapy withdrawal after acquired resistance. Further, subadditive interaction between EGFR- and AXL-targeted inhibitors across all AXL-positive TNBC cell lines may indicate that increased abundance of EGFR is principally a means to transactivation-mediated signaling.


Journal of Cell Biology | 2012

2D protrusion but not motility predicts growth factor-induced cancer cell migration in 3D collagen.

Aaron S. Meyer; Shannon K. Hughes-Alford; Jennifer Elizabeth Kay; Amalchi Castillo; Alan Wells; Frank B. Gertler; Douglas A. Lauffenburger

In breast cancer cells, growth factor stimulation of membrane protrusion was a better predictor of 3D migration than 2D motility, cognate receptor expression, or receptor activation.


Proceedings of the National Academy of Sciences of the United States of America | 2013

ADAM-10 and -17 regulate endometriotic cell migration via concerted ligand and receptor shedding feedback on kinase signaling

Miles A. Miller; Aaron S. Meyer; Michael T. Beste; Zainab Lasisi; Sonika Reddy; Karen W. Jeng; Chia-Hung Chen; Jongyoon Han; Keith B. Isaacson; Linda G. Griffith; Douglas A. Lauffenburger

Significance Regulated cell-surface proteolysis underpins processes of cellular migration in both physiological and pathological contexts. However, comprehending how multiple proteolytic events cohesively integrate to yield context-dependent cellular behavior remains a challenge. Here we present an experimental/computational paradigm for analyzing networks of protease activities that interface with signaling pathways to influence cellular migration in the invasive disease of endometriosis. We find that induced cellular migration is a quantitative consequence of positive feedback through ligand release and negative feedback through receptor shedding, which furthermore drives rapid resistance to kinase inhibitor treatment. Targeted clinical proteomics confirms dysregulated proteolysis in endometriosis. A Disintegrin and Metalloproteinases (ADAMs) are the principal enzymes for shedding receptor tyrosine kinase (RTK) ectodomains and ligands from the cell surface. Multiple layers of activity regulation, feedback, and catalytic promiscuity impede our understanding of context-dependent ADAM “sheddase” function and our ability to predictably target that function in disease. This study uses combined measurement and computational modeling to examine how various growth factor environments influence sheddase activity and cell migration in the invasive disease of endometriosis. We find that ADAM-10 and -17 dynamically integrate numerous signaling pathways to direct cell motility. Data-driven modeling reveals that induced cell migration is a quantitative function of positive feedback through EGF ligand release and negative feedback through RTK shedding. Although sheddase inhibition prevents autocrine ligand shedding and resultant EGF receptor transactivation, it also leads to an accumulation of phosphorylated receptors (HER2, HER4, and MET) on the cell surface, which subsequently enhances Jnk/p38 signaling. Jnk/p38 inhibition reduces cell migration by blocking sheddase activity while additionally preventing the compensatory signaling from accumulated RTKs. In contrast, Mek inhibition reduces ADAM-10 and -17 activities but fails to inhibit compensatory signaling from accumulated RTKs, which actually enhances cell motility in some contexts. Thus, here we present a sheddase-based mechanism of rapidly acquired resistance to Mek inhibition through reduced RTK shedding that can be overcome with rationally directed combination inhibitor treatment. We investigate the clinical relevance of these findings using targeted proteomics of peritoneal fluid from endometriosis patients and find growth-factor–driven ADAM-10 activity and MET shedding are jointly dysregulated with disease.


Biotechnology and Bioengineering | 2009

Concentration of mammalian genomic DNA using two-phase aqueous micellar systems.

Foad Mashayekhi; Aaron S. Meyer; Stacey A. Shiigi; Vu Nguyen; Daniel T. Kamei

The concentration of biomarkers, such as DNA, prior to a subsequent detection step may facilitate the early detection of cancer, which could significantly increase chances for survival. In this study, the partitioning behavior of mammalian genomic DNA fragments in a two‐phase aqueous micellar system was investigated using both experiment and theory. The micellar system was generated using the nonionic surfactant Triton X‐114 and phosphate‐buffered saline (PBS). Partition coefficients were measured under a variety of conditions and compared with our theoretical predictions. With this comparison, we demonstrated that the partitioning behavior of DNA fragments in this system is primarily driven by repulsive, steric, excluded‐volume interactions that operate between the micelles and the DNA fragments, but is limited by the entrainment of micelle‐poor, DNA‐rich domains in the macroscopic micelle‐rich phase. Furthermore, the volume ratio, that is, the volume of the top, micelle‐poor phase divided by that of the bottom, micelle‐rich phase, was manipulated to concentrate DNA fragments in the top phase. Specifically, by decreasing the volume ratio from 1 to 1/10, we demonstrated proof‐of‐principle that the concentration of DNA fragments in the top phase could be increased two‐ to nine‐fold in a predictive manner. Biotechnol. Bioeng. 2009;102: 1613–1623.


Molecular & Cellular Proteomics | 2011

Signaling Network State Predicts Twist-Mediated Effects on Breast Cell Migration Across Diverse Growth Factor Contexts

Hyung-Do Kim; Aaron S. Meyer; Joel P. Wagner; Shannon K. Alford; Alan Wells; Frank B. Gertler; Douglas A. Lauffenburger

Epithelial-mesenchymal transition (EMT), whether in developmental morphogenesis or malignant transformation, prominently involves modified cell motility behavior. Although major advances have transpired in understanding the molecular pathways regulating the process of EMT induction per se by certain environmental stimuli, an important outstanding question is how the activities of signaling pathways governing motility yield the diverse movement behaviors characteristic of pre-induction versus postinduction states across a broad landscape of growth factor contexts. For the particular case of EMT induction in human mammary cells by ectopic expression of the transcription factor Twist, we found the migration responses to a panel of growth factors (EGF, HRG, IGF, HGF) dramatically disparate between confluent pre-Twist epithelial cells and sparsely distributed post-Twist mesenchymal cells—but that a computational model quantitatively integrating multiple key signaling node activities could nonetheless account for this full range of behavior. Moreover, motility in both conditions was successfully predicted a priori for an additional growth factor (PDGF) treatment. Although this signaling network state model could comprehend motility behavior globally, modulation of the network interactions underlying the altered pathway activities was identified by ascertaining differences in quantitative topological influences among the nodes between the two conditions.


Scientific Reports | 2015

Targeting autocrine HB-EGF signaling with specific ADAM12 inhibition using recombinant ADAM12 prodomain.

Miles A. Miller; Marcia L. Moss; Gary Powell; Robert M. Petrovich; Lori L. Edwards; Aaron S. Meyer; Linda G. Griffith; Douglas A. Lauffenburger

Dysregulation of ErbB-family signaling underlies numerous pathologies and has been therapeutically targeted through inhibiting ErbB-receptors themselves or their cognate ligands. For the latter, “decoy” antibodies have been developed to sequester ligands including heparin-binding epidermal growth factor (HB-EGF); however, demonstrating sufficient efficacy has been difficult. Here, we hypothesized that this strategy depends on properties such as ligand-receptor binding affinity, which varies widely across the known ErbB-family ligands. Guided by computational modeling, we found that high-affinity ligands such as HB-EGF are more difficult to target with decoy antibodies compared to low-affinity ligands such as amphiregulin (AREG). To address this issue, we developed an alternative method for inhibiting HB-EGF activity by targeting its cleavage from the cell surface. In a model of the invasive disease endometriosis, we identified A Disintegrin and Metalloproteinase 12 (ADAM12) as a protease implicated in HB-EGF shedding. We designed a specific inhibitor of ADAM12 based on its recombinant prodomain (PA12), which selectively inhibits ADAM12 but not ADAM10 or ADAM17. In endometriotic cells, PA12 significantly reduced HB-EGF shedding and resultant cellular migration. Overall, specific inhibition of ligand shedding represents a possible alternative to decoy antibodies, especially for ligands such as HB-EGF that exhibit high binding affinity and localized signaling.


Journal of Cell Biology | 2016

A requirement for filopodia extension toward Slit during Robo-mediated axon repulsion

Russell E. McConnell; J. Edward van Veen; Marina Vidaki; Adam V. Kwiatkowski; Aaron S. Meyer; Frank B. Gertler

McConnell et al. find that gradients of the repulsive guidance factor Slit induce filopodia formation and elongation toward the cue. This activity is required for axonal repulsion and is driven by ligand-induced binding of Ena/VASP proteins to the Robo receptor.


Bioscience Reports | 2015

Selectivity in subunit composition of Ena/VASP tetramers

Daisy Riquelme; Aaron S. Meyer; Melanie Barzik; Amy E. Keating; Frank B. Gertler

Ena/VASP tetramer composition was analysed and mixed oligomerization of Mena with EVL was found to be unfavourable, while other paralogue combinations formed without apparent bias. The tetramerization domain of Ena/VASP proteins is responsible for their selective tetramer formation.


Cancer Research | 2016

JNK Pathway Activation Modulates Acquired Resistance to EGFR/HER2–Targeted Therapies

Simin Manole; Edward Richards; Aaron S. Meyer

Resistance limits the effectiveness of receptor tyrosine kinase (RTK)-targeted therapies. Combination therapies targeting resistance mechanisms can considerably improve response, but will require an improved understanding of when particular combinations will be effective. One common form of resistance is bypass signaling, wherein RTKs not targeted by an inhibitor can direct reactivation of pathways essential for survival. Although this mechanism of resistance is well appreciated, it is unclear which downstream signaling events are responsible. Here, we apply a combined experimental- and statistical modeling-based approach to identify a set of pathway reactivation essential for RTK-mediated bypass resistance. Differences in the downstream pathway activation provided by particular RTKs lead to qualitative differences in the capacity of each receptor to drive therapeutic resistance. We identify and validate that the JNK pathway is activated during and strongly modulates bypass resistance. These results identify effective therapeutic combinations that block bypass-mediated resistance and provide a basic understanding of this network-level change in kinase dependence that will inform the design of prognostic assays for identifying effective therapeutic combinations in individual patients. Cancer Res; 76(18); 5219-28. ©2016 AACR.


Cancer Research | 2016

Systems Approaches to Cancer Biology

Tenley C. Archer; Elana J. Fertig; Sara J.C. Gosline; Marc Hafner; Shannon K. Hughes; Brian A. Joughin; Aaron S. Meyer; Stephen R. Piccolo; Ayesha N. Shajahan-Haq

Cancer systems biology aims to understand cancer as an integrated system of genes, proteins, networks, and interactions rather than an entity of isolated molecular and cellular components. The inaugural Systems Approaches to Cancer Biology Conference, cosponsored by the Association of Early Career Cancer Systems Biologists and the National Cancer Institute of the NIH, focused on the interdisciplinary field of cancer systems biology and the challenging cancer questions that are best addressed through the combination of experimental and computational analyses. Attendees found that elucidating the many molecular features of cancer inevitably reveals new forms of complexity and concluded that ensuring the reproducibility and impact of cancer systems biology studies will require widespread method and data sharing and, ultimately, the translation of important findings to the clinic. Cancer Res; 76(23); 6774-7. ©2016 AACR.

Collaboration


Dive into the Aaron S. Meyer's collaboration.

Top Co-Authors

Avatar

Douglas A. Lauffenburger

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Frank B. Gertler

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Linda G. Griffith

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Alan Wells

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Edward Richards

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Annelien Zweemer

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Attya Omer

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge