136 results on '"Gregory D Longmore"'
Search Results
2. Data from Randomly Distributed K14+ Breast Tumor Cells Polarize to the Leading Edge and Guide Collective Migration in Response to Chemical and Mechanical Environmental Cues
- Author
-
Steven C. George, Gregory D. Longmore, Ashley C. King, Audrey Brenot, and Priscilla Y. Hwang
- Abstract
Collective cell migration is an adaptive, coordinated interactive process involving cell–cell and cell–extracellular matrix (ECM) microenvironmental interactions. A critical aspect of collective migration is the sensing and establishment of directional movement. It has been proposed that a subgroup of cells known as leader cells localize at the front edge of a collectively migrating cluster and are responsible for directing migration. However, it is unknown how and when leader cells arrive at the front edge and what environmental cues dictate leader cell development and behavior. Here, we addressed these questions by combining a microfluidic device design that mimics multiple tumor microenvironmental cues concurrently with biologically relevant primary, heterogeneous tumor cell organoids. Prior to migration, breast tumor leader cells (K14+) were present throughout a tumor organoid and migrated (polarized) to the leading edge in response to biochemical and biomechanical cues. Impairment of either CXCR4 (biochemical responsive) or the collagen receptor DDR2 (biomechanical responsive) abrogated polarization of leader cells and directed collective migration. This work demonstrates that K14+ leader cells utilize both chemical and mechanical cues from the microenvironment to polarize to the leading edge of collectively migrating tumors.Significance:These findings demonstrate that pre-existing, randomly distributed leader cells within primary tumor organoids use CXCR4 and DDR2 to polarize to the leading edge and direct migration.
- Published
- 2023
- Full Text
- View/download PDF
3. Movie 2 from Randomly Distributed K14+ Breast Tumor Cells Polarize to the Leading Edge and Guide Collective Migration in Response to Chemical and Mechanical Environmental Cues
- Author
-
Steven C. George, Gregory D. Longmore, Ashley C. King, Audrey Brenot, and Priscilla Y. Hwang
- Abstract
Movie demonstrating K14-GFP migration
- Published
- 2023
- Full Text
- View/download PDF
4. Movie 3 from Randomly Distributed K14+ Breast Tumor Cells Polarize to the Leading Edge and Guide Collective Migration in Response to Chemical and Mechanical Environmental Cues
- Author
-
Steven C. George, Gregory D. Longmore, Ashley C. King, Audrey Brenot, and Priscilla Y. Hwang
- Abstract
Movie demonstrating DDR2-/- cells migration
- Published
- 2023
- Full Text
- View/download PDF
5. Movie 1 from Randomly Distributed K14+ Breast Tumor Cells Polarize to the Leading Edge and Guide Collective Migration in Response to Chemical and Mechanical Environmental Cues
- Author
-
Steven C. George, Gregory D. Longmore, Ashley C. King, Audrey Brenot, and Priscilla Y. Hwang
- Abstract
Movie demonstrating collective migration
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figure 3 from p38δ Mitogen-Activated Protein Kinase Is Essential for Skin Tumor Development in Mice
- Author
-
Tatiana Efimova, J. Simon C. Arthur, Grace E. Kissling, Gregory D. Longmore, Robert J. Owen, Meei-Hua Lin, Yan Yin, Carole J. Burns, Erin L. Gribben, Anna Hindes, and Eva M. Schindler
- Abstract
Supplementary Figure 3 from p38δ Mitogen-Activated Protein Kinase Is Essential for Skin Tumor Development in Mice
- Published
- 2023
- Full Text
- View/download PDF
7. Supplemental Figure S5 from Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
David D. Tran, Gregory D. Longmore, Kun Zhang, Michael Kim, Krishna Luitel, and Hung D. Tran
- Abstract
Supplemental Figure S5. Related to Figure 4C-E: Primary breast tumors in Early SNAIL1 KO exhibit loss of EMT phenotype and metastatic potential
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figure S1 from Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
David D. Tran, Gregory D. Longmore, Kun Zhang, Michael Kim, Krishna Luitel, and Hung D. Tran
- Abstract
Supplemental Figure S1. Related to Figure 1: Generation and characterization of the SNAIL1-CBR mouse
- Published
- 2023
- Full Text
- View/download PDF
9. Supplemental Material and Methods from Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
David D. Tran, Gregory D. Longmore, Kun Zhang, Michael Kim, Krishna Luitel, and Hung D. Tran
- Abstract
Supplemental Material and Methods. Additional description of materials and methods used
- Published
- 2023
- Full Text
- View/download PDF
10. Supplemental Figure S4 from Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
David D. Tran, Gregory D. Longmore, Kun Zhang, Michael Kim, Krishna Luitel, and Hung D. Tran
- Abstract
Supplemental Figure S4. Related to Figure 3: SNAIL1 expression in primary breast tumors precedes detection of DTCs, but SNAIL1 is not expressed by DTCs and lung metastases in MMTV-PyMT mice
- Published
- 2023
- Full Text
- View/download PDF
11. Supplemental Figure S2 from Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
David D. Tran, Gregory D. Longmore, Kun Zhang, Michael Kim, Krishna Luitel, and Hung D. Tran
- Abstract
Supplemental Figure S2. Related to Figure 2A-E: SNAIL1 expression in primary breast tumors strongly correlates with increased metastasis in MMTV-neuNT mice
- Published
- 2023
- Full Text
- View/download PDF
12. Supplemental Figure S6 from Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
David D. Tran, Gregory D. Longmore, Kun Zhang, Michael Kim, Krishna Luitel, and Hung D. Tran
- Abstract
Supplemental Figure S6. Related to Figure 5D: SNAIL1 is not expressed by lung metastases in mice overexpressing SNAIL1 in primary breast tumors
- Published
- 2023
- Full Text
- View/download PDF
13. Supplemental Table S1 from Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
David D. Tran, Gregory D. Longmore, Kun Zhang, Michael Kim, Krishna Luitel, and Hung D. Tran
- Abstract
Supplemental Table S1. Spatiotemporal expression of SNAIL1 in MMTV-PyMT mice
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure 1 from p38δ Mitogen-Activated Protein Kinase Is Essential for Skin Tumor Development in Mice
- Author
-
Tatiana Efimova, J. Simon C. Arthur, Grace E. Kissling, Gregory D. Longmore, Robert J. Owen, Meei-Hua Lin, Yan Yin, Carole J. Burns, Erin L. Gribben, Anna Hindes, and Eva M. Schindler
- Abstract
Supplementary Figure 1 from p38δ Mitogen-Activated Protein Kinase Is Essential for Skin Tumor Development in Mice
- Published
- 2023
- Full Text
- View/download PDF
15. Supplemental Figure S3 from Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
David D. Tran, Gregory D. Longmore, Kun Zhang, Michael Kim, Krishna Luitel, and Hung D. Tran
- Abstract
Supplemental Figure S3. Related to Figure 2F-J: SNAIL1 expression in primary breast tumors correlates with increased invasion
- Published
- 2023
- Full Text
- View/download PDF
16. Supplemental Table and Figure Legends from Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
David D. Tran, Gregory D. Longmore, Kun Zhang, Michael Kim, Krishna Luitel, and Hung D. Tran
- Abstract
Supplemental Table and Figure Legends. Legends to the supplemental table and figures
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Methods, Figure Legends 1-3 from p38δ Mitogen-Activated Protein Kinase Is Essential for Skin Tumor Development in Mice
- Author
-
Tatiana Efimova, J. Simon C. Arthur, Grace E. Kissling, Gregory D. Longmore, Robert J. Owen, Meei-Hua Lin, Yan Yin, Carole J. Burns, Erin L. Gribben, Anna Hindes, and Eva M. Schindler
- Abstract
Supplementary Methods, Figure Legends 1-3 from p38δ Mitogen-Activated Protein Kinase Is Essential for Skin Tumor Development in Mice
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure 2 from p38δ Mitogen-Activated Protein Kinase Is Essential for Skin Tumor Development in Mice
- Author
-
Tatiana Efimova, J. Simon C. Arthur, Grace E. Kissling, Gregory D. Longmore, Robert J. Owen, Meei-Hua Lin, Yan Yin, Carole J. Burns, Erin L. Gribben, Anna Hindes, and Eva M. Schindler
- Abstract
Supplementary Figure 2 from p38δ Mitogen-Activated Protein Kinase Is Essential for Skin Tumor Development in Mice
- Published
- 2023
- Full Text
- View/download PDF
19. A Cdh3-Lam332 signaling axis in a leader cell subpopulation controls protrusion dynamics and tumor organoid collective migration
- Author
-
Priscilla Y Hwang, Jairaj Mathur, Yanyang Cao, Jose Almeida, Daphne Cornish, Maria Clarke, Amit Pathak, and Gregory D Longmore
- Abstract
Carcinoma dissemination can occur when heterogeneous tumor and tumor stromal cells clusters migrate together via collective migration. Cells at the front lead and direct collective migration, yet how these leader cells form and interact with the microenvironment to direct migration are not fully appreciated. From live videos of primary mouse and human breast tumor organoids in a 3D microfluidic system that mimics the native breast tumor microenvironment, we developed 3D computational models which hypothesize that leader cells generate high protrusive forces and overcome extracellular matrix (ECM) resistance. Using single cell sequencing, we reveal leader cells are heterogeneous, and identify and isolate a unique Cadherin-3 (Cdh3) positive leader cell subpopulation that is necessary and sufficient to lead migration. Cdh3 controls leader cell protrusion dynamics through the local production of Laminin-332 which is required for integrin/focal adhesion function. Our findings highlight how a subset of leader cells interact with the microenvironment to direct collective migration.TeaserHigher protrusions of Cdh3+ leader cells polarize tumor organoids that then invade collagen via Lam332 adhesion feedback.
- Published
- 2022
- Full Text
- View/download PDF
20. A Cdh3-β-catenin-laminin signaling axis in a subset of breast tumor leader cells control leader cell polarization and directional collective migration
- Author
-
Priscilla Y. Hwang, Jairaj Mathur, Yanyang Cao, Jose Almeida, Jiayu Ye, Vasilios Morikis, Daphne Cornish, Maria Clarke, Sheila A. Stewart, Amit Pathak, and Gregory D. Longmore
- Subjects
Cell Biology ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology ,Developmental Biology - Published
- 2023
- Full Text
- View/download PDF
21. Tyrosine kinase-independent actions of DDR2 in tumor cells and cancer-associated fibroblasts influence tumor invasion, migration and metastasis
- Author
-
Audrey Brenot, Maria Clarke, Patrick Pence, Priscilla Y. Hwang, Craig E. Barcus, Gregory D. Longmore, and Vasilios Morikis
- Subjects
Stromal cell ,medicine.drug_class ,Breast Neoplasms ,Tyrosine-kinase inhibitor ,Receptor tyrosine kinase ,Metastasis ,Mice ,Discoidin Domain Receptor 2 ,Collective Cell Migration ,Cancer-Associated Fibroblasts ,Paracrine actions ,Cell Movement ,Tumor Microenvironment ,medicine ,DDR2 ,Animals ,Humans ,Phosphorylation ,Tyrosine ,Kinase independent ,Tumor microenvironment ,biology ,Cell Biology ,Cell Biology and Disease ,Fibroblasts ,medicine.disease ,Metastatic breast cancer ,biology.protein ,Cancer research ,Female ,Tyrosine kinase ,Research Article - Abstract
Both tumor cell-intrinsic signals and tumor cell-extrinsic signals from cells within the tumor microenvironment influence tumor cell dissemination and metastasis. The fibrillar collagen receptor tyrosine kinase (RTK) discoidin domain receptor 2 (DDR2) is essential for breast cancer metastasis in mouse models, and high expression of DDR2 in tumor and tumor stromal cells is strongly associated with poorer clinical outcomes. DDR2 tyrosine kinase activity has been hypothesized to be required for the metastatic activity of DDR2; however, inhibition of DDR2 tyrosine kinase activity, along with that of other RTKs, has failed to provide clinically relevant responses in metastatic patients. Here, we show that tyrosine kinase activity-independent action of DDR2 in tumor cells can support Matrigel invasion and in vivo metastasis. Paracrine actions of DDR2 in tumor cells and cancer-associated fibroblasts (CAFs) also support tumor invasion, migration and lung colonization in vivo. These data suggest that tyrosine kinase-independent functions of DDR2 could explain failures of tyrosine kinase inhibitor treatment in metastatic breast cancer patients and highlight the need for alternative therapeutic strategies that inhibit both tyrosine kinase-dependent and -independent actions of RTKs in the treatment of breast cancer. This article has an associated First Person interview with the first author of the paper., Summary: The collagen receptor DDR2, a receptor tyrosine kinase, exhibits tyrosine kinase-independent functions that influence paracrine regulation of breast tumor cell invasion and breast cancer metastasis.
- Published
- 2021
- Full Text
- View/download PDF
22. Collagen Linearization within Tumors
- Author
-
Gregory D. Longmore and Craig E. Barcus
- Subjects
Cancer Research ,Lung Neoplasms ,Mice, Nude ,Apoptosis ,Breast Neoplasms ,Mice, SCID ,Collagen Type I ,Article ,Metastasis ,CCN Intercellular Signaling Proteins ,Extracellular matrix ,Mice ,Immune system ,Cell Movement ,Mice, Inbred NOD ,Proto-Oncogene Proteins ,Neoplasms ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Neoplasm Invasiveness ,Tumor growth ,Cell Proliferation ,Mice, Inbred BALB C ,Tumor microenvironment ,Chemistry ,Tumor Cell Invasion ,Cancer ,Endothelial Cells ,Prognosis ,medicine.disease ,Xenograft Model Antitumor Assays ,Extracellular Matrix ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,Survival Rate ,Oncology ,Cancer research ,Female ,sense organs ,Collagen ,Function (biology) ,Signal Transduction - Abstract
It is now well appreciated that the tumor microenvironment (TME) surrounding primary tumors impacts tumor growth, progression (invasion and migration), and response to therapy. Broadly speaking, the TME is composed of cells (immune cells, activated fibroblasts, adipocytes, endothelial cells), acellular extracellular matrix (ECM), and cytokines or growth factors, some of which are bound or tethered to the ECM proteins. All these compartments undergo significant changes during tumor development and progression. Changes to the ECM, in particular, can dramatically influence cancer biology. This has stimulated the development of therapies that directly reverse or prevent the structural changes in the TME ECM that facilitate cancer progression. But to do so, in a rational manner, we need to understand how structural changes to tumor ECM arise, are remodeled, and function to facilitate tumor cell invasion and migration that give rise to metastatic disease, which is the main cause of cancer-related deaths. In this issue of Cancer Research, Janjanam and colleagues show that the ratio of WISP1/WISP2 in tumors is critical for ECM collagen fiber linearization and important for metastasis. WISP2 binds ECM collagen directly and can inhibit WISP1-mediated collagen linearization. These new results offer a new approach for targeting the altered collagen ECM in tumors by preventing or reversing collagen linearization. See related article by Janjanam et al., p. 5666
- Published
- 2021
23. LATS1/2 kinases trigger self-renewal of cancer stem cells in aggressive oral cancer
- Author
-
Yoko Naito, Norikazu Yabuta, Towa Sasakura, Kohshiro Fukushima, Moe Fukuzawa, Ayumi Okamoto, Satomi Mukai, Masami Nozaki, Gregory D. Longmore, and Hiroshi Nojima
- Subjects
0301 basic medicine ,Hippo signaling pathway ,cancer stem cells (CSCs) ,biology ,Cell division ,SNAIL ,Kinase ,Hippo pathway ,LATS ,Cancer ,Snail ,Tumor initiation ,oral cancer ,Self renewal ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Cancer stem cell ,030220 oncology & carcinogenesis ,biology.animal ,Cancer research ,medicine ,Priority Research Paper - Abstract
Cancer stem cells (CSCs), which play important roles in tumor initiation and progression, are resistant to many types of therapies. However, the regulatory mechanisms underlying CSC-specific properties, including self-renewal, are poorly understood. Here, we found that LATS1/2, the core Hippo pathway-kinases, were highly expressed in the oral squamous cell carcinoma line SAS, which exhibits high capacity of CSCs, and that depletion of these kinases prevented SAS cells from forming spheres under serum-free conditions. Detailed examination of the expression and activation of LATS kinases and related proteins over a time course of sphere formation revealed that LATS1/2 were more highly expressed and markedly activated before initiation of self-renewal. Moreover, TAZ, SNAIL, CHK1/2, and Aurora-A were expressed in hierarchical, oscillating patterns during sphere formation, suggesting that the process consists of four sequential steps. Our results indicate that LATS1/2 trigger self-renewal of CSCs by regulating the Hippo pathway, the EMT, and cell division.
- Published
- 2019
- Full Text
- View/download PDF
24. Micro-strains in the extracellular matrix induce angiogenesis
- Author
-
Joshua B. Katz, Steven C. George, M. K. Sewell-Loftin, and Gregory D. Longmore
- Subjects
Cellular activity ,Angiogenesis ,Microfluidics ,Biomedical Engineering ,Neovascularization, Physiologic ,Bioengineering ,Biochemistry ,Article ,Extracellular matrix ,Neovascularization ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Neoplasms ,medicine ,Tumor Microenvironment ,Humans ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,Neovascularization, Pathologic ,Chemistry ,General Chemistry ,In vitro ,Cell biology ,Extracellular Matrix ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine.symptom ,Blood vessel - Abstract
An improved understanding of biomechanical factors that control tumor development, including angiogenesis, could explain why few of the promising treatment strategies discovered via in vitro models translate well into in vivo or clinical studies. The ability to manipulate and in real-time study the multiple independent biomechanical properties on cellular activity has been limited, primarily due to limitations in traditional in vitro platforms or the inability to manipulate such factors in vivo. We present a novel microfluidic platform that mimics the vascularized tumor microenvironment with independent control of interstitial flow and mechanical strain. The microtissue platform design isolates mechanically-stimulated angiogenesis in the tumor microenvironment, by manipulating interstitial flow to eliminate soluble factors that could drive blood vessel growth. Our studies demonstrate that enhanced mechanical strain induced by cancer-associated fibroblasts (CAFs) promotes angiogenesis in microvasculature models, even when preventing diffusion of soluble factors to the growing vasculature. Moreover, small but significant decreases in micro-strains induced by inhibited CAFs were sufficient to reduce angiogenesis. Ultimately, we believe this platform represents a significant advancement in the ability to investigate biomechanical signals while controlling for biochemical signals, with a potential to be utilized in fields beyond cancer research.
- Published
- 2020
25. TWIST1 induces expression of discoidin domain receptor 2 to promote ovarian cancer metastasis
- Author
-
A. Lohrey, E.H. Meller, Laura M. Divine, Riva A. Desai, Whitney R. Grither, Katherine Fuh, Daniel Wilke, Andrew J. Loza, Peinan Zhao, and Gregory D. Longmore
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Snail1 ,Mice, Nude ,Article ,Receptor tyrosine kinase ,Metastasis ,Collagen receptor ,Mice ,03 medical and health sciences ,Ovarian tumor ,Discoidin Domain Receptor 2 ,0302 clinical medicine ,Cell Movement ,fibronectin ,Biomarkers, Tumor ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Receptor ,Molecular Biology ,Cells, Cultured ,collagen receptor ,Neoplasm Staging ,Ovarian Neoplasms ,Mice, Inbred BALB C ,biology ,Twist-Related Protein 1 ,Nuclear Proteins ,medicine.disease ,Up-Regulation ,3. Good health ,Gene Expression Regulation, Neoplastic ,Fibronectin ,030104 developmental biology ,030220 oncology & carcinogenesis ,receptor tyrosine kinase ,Cancer research ,biology.protein ,Female ,Ovarian cancer ,Mesothelial cell clearance ,Discoidin domain - Abstract
The mesenchymal gene program has been shown to promote the metastatic progression of ovarian cancer; however, specific proteins induced by this program that lead to these metastatic behaviors have not been identified. Using patient derived tumor cells and established human ovarian tumor cell lines, we find that the Epithelial-to –Mesenchymal Transition inducing factor TWIST1 drives expression of Discoidin Domain Receptor 2 (DDR2), a receptor tyrosine kinase (RTK) that recognizes fibrillar collagen as ligand. The expression and action of DDR2 was critical for mesothelial cell clearance, invasion and migration in ovarian tumor cells. It does so, in part, by upregulating expression and activity of matrix remodeling enzymes that lead to increased cleavage of fibronectin and spreading of tumor cells. Additionally, DDR2 stabilizes SNAIL1, allowing for sustained mesenchymal phenotype. In patient derived ovarian cancer specimens, DDR2 expression correlated with enhanced invasiveness. DDR2 expression was associated with advanced stage ovarian tumors and metastases. In vivo studies demonstrated that the presence of DDR2 is critical for ovarian cancer metastasis. These findings indicate that the collagen receptor DDR2 is critical for multiple steps of ovarian cancer progression to metastasis, and thus, identifies DDR2 as a potential new target for the treatment of metastatic ovarian cancer.
- Published
- 2018
- Full Text
- View/download PDF
26. Author Correction: Guidelines and definitions for research on epithelial–mesenchymal transition
- Author
-
Jing Yang, Geert Berx, Elizabeth D. Williams, Sendurai A. Mani, Gregory J. Goodall, David R. McClay, Binhua P. Zhou, Parker B. Antin, Raghu Kalluri, Pierre Savagner, Erik W. Thompson, Roberto Mayor, Amparo Cano, Jinsong Liu, Robert A. Weinberg, Masatoshi Takeichi, Thomas Brabletz, Chaya Kalcheim, Donald F. Newgreen, Shoukat Dedhar, Kyra Campbell, Herbert Levine, Raymond B. Runyan, Jordi Casanova, Rik Derynck, Anna-Katerina Hadjantonakis, M. Angela Nieto, Marc P. Stemmler, Yeesim Khew-Goodall, Jean Paul Thiery, Heide L. Ford, Marianne E. Bronner, Yibin Kang, Antonio García de Herreros, Ben Z. Stanger, Cédric Blanpain, Keith E. Mostov, Jonas Fuxe, Eric Theveneau, Yoshiko Takahashi, Alain Puisieux, Ruby Yun-Ju Huang, Gerhard Christofori, Guojun Sheng, Joan Massagué, Gregory D. Longmore, and Jianhua Xing
- Subjects
Biomedical Research ,Consensus ,Epithelial-Mesenchymal Transition ,business.industry ,Published Erratum ,Cell Plasticity ,MEDLINE ,Cell Biology ,Cell Movement ,Neoplasms ,Terminology as Topic ,Cancer research ,Animals ,Humans ,Medicine ,Epithelial–mesenchymal transition ,Author Correction ,business ,Molecular Biology ,Developmental Biology - Abstract
Epithelial-mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by 'the EMT International Association' (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT.
- Published
- 2021
- Full Text
- View/download PDF
27. Wnt5a influences the directional displacement of ovarian cancer cells
- Author
-
Breanna Baker, Vasilios Morikis, David G. Mutch, Katherine Fuh, Whitney R. Grither, Premal H. Thaker, Matthew A. Powell, Lindsay M. Kuroki, Gregory D. Longmore, Andrea R. Hagemann, and Dineo Khabele
- Subjects
biology ,business.industry ,Wnt signaling pathway ,Obstetrics and Gynecology ,Motility ,ROR2 ,Cell migration ,medicine.disease ,Metastasis ,body regions ,Fibronectin ,Oncology ,embryonic structures ,Cancer research ,biology.protein ,Medicine ,sense organs ,business ,Ovarian cancer ,Receptor - Abstract
Objectives: The noncanonical Wnt ligand Wnt5a is found in high concentrations in ovarian cancer ascites. This microenvironmental Wnt5a has recently been shown to potentiate ovarian cancer metastasis. We sought to identify whether Wnt5a modulates ovarian cancer cell motility. Methods: We engineered and validated a microfluidic device that establishes stable gradients of soluble ligands. After a Wnt5a gradient was established, ovarian cancer cells (ES2 or OVCAR5) were introduced and allowed to adhere to a 2-dimensional fibronectin substrate. Real time imaging was carried out over 7 hours and cellular migration was analyzed with respect to speed, displacement, and directionality. In a subset of experiments, ROR2, a noncanonical Wnt5a receptor involved in planar cell polarity (PCP) pathways, was depleted with siRNA to assess contribution of ROR2-dependent PCP to Wnt5a mediated behaviors. Results: In the presence of Wnt5a, ES2 and OVCAR5 ovarian cancer cells demonstrated an increase in both mean and instantaneous velocity of migration when compared to absence of ligand (P Conclusions: Wnt5a promotes the motility of ovarian cancer cells by influencing both directionality of motion as well as degree of displacement. Furthermore, the Wnt5a receptor ROR2 was shown to be critical in mediating these behaviors, suggesting a role for planar cell polarity pathways in ovarian cancer metastasis. As Wnt5a is produced at high levels by peritoneal mesothelial cells and omental adipose tissue in the metastatic microenvironment of ovarian cancer, these data suggest a potential mechanism by which ovarian cancer cells ‘home’ to their metastatic niche. ROR2 represents an attractive target for disrupting this tumor cell/microenvironment interaction and represents a possible new therapeutic target.
- Published
- 2021
- Full Text
- View/download PDF
28. Silencing ROR2 inhibits metastatic behavior of ovarian cancer cells
- Author
-
David G. Mutch, Premal H. Thaker, Whitney R. Grither, Lindsay M. Kuroki, Dineo Khabele, Gregory D. Longmore, Andrea R. Hagemann, Katherine Fuh, Matthew A. Powell, and Breanna Baker
- Subjects
endocrine system diseases ,business.industry ,Cell ,Wnt signaling pathway ,Obstetrics and Gynecology ,medicine.disease ,Metastasis ,body regions ,Ovarian tumor ,medicine.anatomical_structure ,Oncology ,Cell culture ,Cancer research ,Medicine ,Gene silencing ,business ,Ovarian cancer ,Mesothelial Cell - Abstract
Objectives: Adhesion of migrating ovarian tumor cells to peritoneal mesothelial cells and their subsequent clearance of the mesothelial monolayer is a key early event in metastatic seeding of ovarian cancer. We sought to identify mediators of ovarian cancer adhesion and invasion contributing to this tumor-microenvironment interplay. Methods: To identify potential drivers of ovarian cancer adhesion and invasion, a multicellular assay incorporating tumor (epithelial ovarian cancer cells) and microenvironment (patient derived omental fibroblasts) was adapted to carry out a high throughput genetic screen using a human kinome siRNA library. Based on screen candidates, ROR2, a noncanonical Wnt receptor that binds Wnt5a, was identified. ROR2 expression was evaluated in primary patient derived ovarian cancer cells and in high grade ovarian cancer cell lines (ES2, A2780, OVCAR3, OVCAR5, OVCAR8, ID8Trp53-/-BRCA2-/-). Biologic roles of ROR2 as well as the therapeutic effect of ROR2 silencing were examined in vitro using a validated mesothelial cell clearance assay. We further examined the effect of ROR2 silencing on force generation by ovarian cancer cell spheroids using immunofluorescent staining of phospho-myosin light chain (pMLC). Results: ROR2 was found to be expressed at a protein level across established ovarian cancer cell lines as well as primary ovarian cancer cells from patient derived tumors. Analysis in multicellular assays demonstrated that ROR2 silencing with siRNA dramatically decreases mesothelial cell clearance by tumor cell spheroids when compared to control siNEG spheroids. This result was observed across multiple cell lines (ES2, OVCAR8, OVCAR5, and OVCAR3) (P Conclusions: ROR2 plays a critical role in mediating the ability of ovarian cancer cells to clear a mesothelial cell monolayer, a key step leading to metastasis in ovarian cancer. Mesothelial clearance has been demonstrated to be a force dependent process, and our data suggests that ROR2 may play a role in force generation by ovarian cancer spheroids. Understanding the factors driving prometastatic tumor-microenvironment interactions is critical to develop targeted therapeutics to block the spread of ovarian cancer. As ROR2 is an actionable target, further investigation of ROR2 targeting in ovarian cancer is needed as it may represent a novel treatment strategy.
- Published
- 2021
- Full Text
- View/download PDF
29. Contextual Cues from Cancer Cells Govern CAF Heterogeneity
- Author
-
Jing Wang, Russell William, Jordan S. Miller, Daniel W. Sazer, Chad J. Creighton, Xin Liu, Michael weiger, Hou-Fu Guo, Barbara Mino, Kuanwei Sheng, Neus Bota-Rabassedas, Gregory D. Longmore, Joshua J.A. Firestone, Maria Gabriela Raso, Pamela Villalobos, B. Leticia Rodriguez, Jaime Rodriguez-Canales, Yuanxin Xi, Ignacio I. Wistuba, Yichi Niu, Priyam Banerjee, Jonathan M. Kurie, Edwin Roger Parra, Wenjian Cao, Don L. Gibbons, Xiaochao Tan, Jiayi Luo, Luisa S Solis, Jiang Yu, Jacob L. Albritton, Chenghang Zong, and Harold A. Chapman
- Subjects
Tumor microenvironment ,Stromal cell ,Single cell sequencing ,Cancer cell ,medicine ,Cancer-Associated Fibroblasts ,Adenocarcinoma ,Biology ,medicine.disease ,Reprogramming ,Phenotype ,Cell biology - Abstract
Cancer cells function as primary architects of the tumor microenvironment. Yet, the molecular features of cancer cells that govern stromal cell phenotypes remain unclear. Here, we show that cancer-associated fibroblasts (CAFs) are distinguishable on the basis of gene expression signatures they acquire in co-culture with epithelial- or mesenchymal-like lung adenocarcinoma (LUAD) cells. High expression of the EMT activator ZEB1 endows LUAD cells with the capacity to activate a soluble factor exchange that leads to CAF reprogramming, to generate CAF-led invasive projections in multicellular aggregates, and to respond to pro-metastatic signals from CAFs in mice. Thus, ZEB1-expressing LUAD cells are positioned at the apex of a signaling hierarchy in the tumor microenvironment.
- Published
- 2020
- Full Text
- View/download PDF
30. DDR2 controls breast tumor stiffness and metastasis by regulating integrin mediated mechanotransduction in CAFs
- Author
-
Whitney R. Grither, Christopher Walter, Amit Pathak, Samantha Van Hove Bayer, Melanie Ernst, Gregory D. Longmore, Audrey Brenot, Craig E. Barcus, Patrick Pence, and Priscilla Y. Hwang
- Subjects
collagen ,0301 basic medicine ,Integrins ,Mouse ,QH301-705.5 ,Science ,Integrin ,Breast Neoplasms ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Collagen receptor ,Mice ,03 medical and health sciences ,Discoidin Domain Receptor 2 ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,In vivo ,Tumor Microenvironment ,medicine ,Animals ,Humans ,DDR2 ,metastasis ,Biology (General) ,Neoplasm Metastasis ,Mechanotransduction ,Cancer Biology ,mechanotransduction ,Tumor microenvironment ,Lung ,General Immunology and Microbiology ,biology ,business.industry ,General Neuroscience ,Cell Biology ,General Medicine ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Medicine ,business ,Research Article - Abstract
Biomechanical changes in the tumor microenvironment influence tumor progression and metastases. Collagen content and fiber organization within the tumor stroma are major contributors to biomechanical changes (e., tumor stiffness) and correlated with tumor aggressiveness and outcome. What signals and in what cells control collagen organization within the tumors, and how, is not fully understood. We show in mouse breast tumors that the action of the collagen receptor DDR2 in CAFs controls tumor stiffness by reorganizing collagen fibers specifically at the tumor-stromal boundary. These changes were associated with lung metastases. The action of DDR2 in mouse and human CAFs, and tumors in vivo, was found to influence mechanotransduction by controlling full collagen-binding integrin activation via Rap1-mediated Talin1 and Kindlin2 recruitment. The action of DDR2 in tumor CAFs is thus critical for remodeling collagen fibers at the tumor-stromal boundary to generate a physically permissive tumor microenvironment for tumor cell invasion and metastases.
- Published
- 2019
- Full Text
- View/download PDF
31. Author response: DDR2 controls breast tumor stiffness and metastasis by regulating integrin mediated mechanotransduction in CAFs
- Author
-
Gregory D. Longmore, Audrey Brenot, Amit Pathak, Christopher Walter, Whitney R. Grither, Samantha Vh Bayer, Patrick Pence, Melanie Ernst, Craig E. Barcus, and Priscilla Y. Hwang
- Subjects
biology ,business.industry ,Integrin ,medicine ,Cancer research ,biology.protein ,Mechanotransduction ,medicine.disease ,business ,Metastasis ,Breast tumor - Published
- 2019
- Full Text
- View/download PDF
32. Correction: Mechanical signals regulate and activate SNAIL1 protein to control the fibrogenic response of cancer-associated fibroblasts (doi:10.1242/jcs.180539)
- Author
-
Hirak Biswas, Suzanne M. Ponik, Patricia J. Keely, Kun Zhang, Samantha Van Hove, Kevin W. Eliceiri, Gregory D. Longmore, and Whitney R. Grither
- Subjects
0303 health sciences ,Cell ,Substrate (chemistry) ,Cell Biology ,Biology ,Cell biology ,Original data ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Cancer-Associated Fibroblasts ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
There was an error in J. Cell Sci. (2016) 129, [1989-2002][1] ([doi:10.1242/jcs.180539][2]). The -YAP panels on stiff substrate in Fig. 8C were incorrectly duplicated in Fig. 6A. The journal has seen the original data for Fig. 6A and the corrected and original panels are shown below. This error
- Published
- 2019
- Full Text
- View/download PDF
33. Randomly Distributed K14+ Breast Tumor Cells Polarize to the Leading Edge and Guide Collective Migration in Response to Chemical and Mechanical Environmental Cues
- Author
-
Priscilla Y. Hwang, Audrey Brenot, Gregory D. Longmore, Ashley C. King, and Steven C. George
- Subjects
0301 basic medicine ,Cancer Research ,Leading edge ,Cell signaling ,Cellular differentiation ,Oncology and Carcinogenesis ,Bioengineering ,Cell Communication ,Biology ,Transgenic ,Collagen receptor ,Extracellular matrix ,03 medical and health sciences ,Mice ,Experimental ,0302 clinical medicine ,Discoidin Domain Receptor 2 ,Cell Movement ,Receptors ,Breast Cancer ,Tumor Microenvironment ,Animals ,Humans ,Oncology & Carcinogenesis ,Sensory cue ,Cancer ,CXCR4 ,Tumor microenvironment ,Mammary Neoplasms ,Keratin-14 ,Cell Differentiation ,Cell biology ,Extracellular Matrix ,Organoids ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Female ,Signal transduction ,Signal Transduction - Abstract
Collective cell migration is an adaptive, coordinated interactive process involving cell–cell and cell–extracellular matrix (ECM) microenvironmental interactions. A critical aspect of collective migration is the sensing and establishment of directional movement. It has been proposed that a subgroup of cells known as leader cells localize at the front edge of a collectively migrating cluster and are responsible for directing migration. However, it is unknown how and when leader cells arrive at the front edge and what environmental cues dictate leader cell development and behavior. Here, we addressed these questions by combining a microfluidic device design that mimics multiple tumor microenvironmental cues concurrently with biologically relevant primary, heterogeneous tumor cell organoids. Prior to migration, breast tumor leader cells (K14+) were present throughout a tumor organoid and migrated (polarized) to the leading edge in response to biochemical and biomechanical cues. Impairment of either CXCR4 (biochemical responsive) or the collagen receptor DDR2 (biomechanical responsive) abrogated polarization of leader cells and directed collective migration. This work demonstrates that K14+ leader cells utilize both chemical and mechanical cues from the microenvironment to polarize to the leading edge of collectively migrating tumors. Significance: These findings demonstrate that pre-existing, randomly distributed leader cells within primary tumor organoids use CXCR4 and DDR2 to polarize to the leading edge and direct migration.
- Published
- 2019
34. Randomly Distributed K14
- Author
-
Priscilla Y, Hwang, Audrey, Brenot, Ashley C, King, Gregory D, Longmore, and Steven C, George
- Subjects
Receptors, CXCR4 ,Keratin-14 ,Mammary Neoplasms, Experimental ,Cell Differentiation ,Mice, Transgenic ,Cell Communication ,Article ,Extracellular Matrix ,Organoids ,Mice ,Discoidin Domain Receptor 2 ,Cell Movement ,Tumor Microenvironment ,Animals ,Humans ,Female ,Signal Transduction - Abstract
Collective cell migration is an adaptive, coordinated interactive process involving cell-cell and cell-extracellular matrix (ECM) microenvironmental interactions. A critical aspect of collective migration is the sensing and establishment of directional movement. It has been proposed that a subgroup of cells known as leader cells localize at the front edge of a collectively migrating cluster and are responsible for directing migration. However, it is unknown how and when leader cells arrive at the front edge and what environmental cues dictate leader cell development and behavior. Here we addressed these questions by combining a microfluidic device design that mimics multiple tumor microenvironmental cues concurrently with biologically relevant primary, heterogeneous tumor cell organoids. Prior to migration, breast tumor leader cells (K14+) were present throughout a tumor organoid and migrated (polarized) to the leading edge in response to biochemical and biomechanical cues. Impairment of either CXCR4 (biochemical responsive) or the collagen receptor DDR2 (biomechanical responsive) abrogated polarization of leader cells and directed collective migration. This work demonstrates that K14+ leader cells utilize both chemical and mechanical cues from the microenvironment to polarize to the leading edge of collectively migrating tumors.
- Published
- 2018
35. Inhibition of tumor-microenvironment interaction and tumor invasion by small-molecule allosteric inhibitor of DDR2 extracellular domain
- Author
-
Gregory D. Longmore and Whitney R. Grither
- Subjects
0301 basic medicine ,Stromal cell ,Medical Sciences ,Allosteric regulation ,Antineoplastic Agents ,Breast Neoplasms ,Mice, Transgenic ,allosteric inhibition ,Receptor tyrosine kinase ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Discoidin Domain Receptor 2 ,Allosteric Regulation ,medicine ,Tumor Microenvironment ,Animals ,Humans ,DDR2 ,metastasis ,Neoplasm Invasiveness ,Receptor ,Protein Kinase Inhibitors ,Tumor microenvironment ,Mice, Inbred BALB C ,Multidisciplinary ,biology ,Chemistry ,Fibroblasts ,Biological Sciences ,medicine.disease ,Small molecule ,3. Good health ,Cell biology ,Neoplasm Proteins ,030104 developmental biology ,PNAS Plus ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Discoidin domain ,Signal Transduction - Abstract
Significance To effectively prevent cancer spread from primary tumor sites, new treatments need to target tumor cells, the cells and extracellular matrix within the tumor environment, and communicating pathways between these sites simultaneously. The collagen receptor discoidin domain receptor 2 (DDR2) has been implicated as such a target. Here, we describe the identification and characterization of a small molecule inhibitor of DDR2 that uniquely acts in an allosteric manner via the extracellular domain to selectively inhibit the action of DDR2 in tumor cells and tumor stromal cancer-associated fibroblasts. In experimental mouse models of breast cancer, WRG-28 inhibits DDR2 signaling and tumor cell invasion., The action of the collagen binding receptor tyrosine kinase (RTK) discoidin domain receptor 2 (DDR2) in both tumor and tumor stromal cells has been established as critical for breast cancer metastasis. Small molecule inhibitors that target the extracellular domain of RTKs are rare, as they have classically been regarded as too small to block binding with large polypeptide ligands. Here, we report the identification and characterization of a selective, extracellularly acting small molecule inhibitor (WRG-28) of DDR2 that uniquely inhibits receptor–ligand interactions via allosteric modulation of the receptor. By targeting DDR2, WRG-28 inhibits tumor invasion and migration, as well as tumor-supporting roles of the stroma, and inhibits metastatic breast tumor cell colonization in the lungs. These findings represent an approach to inhibiting tumor–stromal interactions and support the development of allosteric inhibitors of DDR2, such as WRG-28, as a promising approach to antimetastasis treatment.
- Published
- 2018
36. SNAIL1 action in tumor cells influences macrophage polarization and metastasis in breast cancer through altered GM-CSF secretion
- Author
-
David G. DeNardo, Brett L. Knolhoff, Audrey Brenot, and Gregory D. Longmore
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,business.industry ,Macrophage polarization ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Article ,3. Good health ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Tumor progression ,030220 oncology & carcinogenesis ,Genetic model ,Cancer research ,Medicine ,Tumor necrosis factor alpha ,skin and connective tissue diseases ,business ,Molecular Biology - Abstract
The EMT inducer SNAIL1 regulates breast cancer metastasis and its expression in human primary breast tumor predicts for poor outcomes. During tumor progression SNAIL1 has multiple effects in tumor cells that can impact metastasis. An inflammatory tumor microenvironment also impacts metastasis and recently SNAIL1 has been implicated as modulating the secretion of cytokines that can influence the tumor immune infiltrate. Using a spontaneous genetic model of breast cancer metastasis and syngeneic orthotopic transplant experiments we show that the action of SNAIL1 in primary breast tumor cells is required for breast tumor growth and metastasis. It does so, in part, by regulating production of GM-CSF, IL1α, IL-6, and TNFα by breast cancer cells. The SNAIL1-dependent tumor cell secretome modulates the primary tumor-associated macrophage (TAM) polarization. GM-CSF alone modulates TAM polarization and impacts breast cancer metastasis in vivo. This study highlights another role for breast tumor SNAIL1 in cancer progression to metastasis—modulation of the immune microenvironment of primary breast tumors.
- Published
- 2018
- Full Text
- View/download PDF
37. Past matrix stiffness primes epithelial cells and regulates their future collective migration through a mechanical memory
- Author
-
Christopher Walter, Amit Pathak, Gregory V. Schimizzi, Andrew J. Loza, Samila Nasrollahi, and Gregory D. Longmore
- Subjects
0301 basic medicine ,Materials science ,Biophysics ,Morphogenesis ,Priming (immunology) ,Motility ,Bioengineering ,02 engineering and technology ,Mechanotransduction, Cellular ,Article ,Biomaterials ,Extracellular matrix ,Focal adhesion ,03 medical and health sciences ,Cell Movement ,medicine ,Humans ,Mechanotransduction ,Cytoskeleton ,Focal Adhesions ,Stiffness ,Epithelial Cells ,021001 nanoscience & nanotechnology ,Cell biology ,Extracellular Matrix ,030104 developmental biology ,Mechanics of Materials ,Ceramics and Composites ,Mechanosensitive channels ,medicine.symptom ,0210 nano-technology ,Biomedical engineering - Abstract
During morphogenesis and cancer metastasis, grouped cells migrate through tissues of dissimilar stiffness. Although the influence of matrix stiffness on cellular mechanosensitivity and motility are well-recognized, it remains unknown whether these matrix-dependent cellular features persist after cells move to a new microenvironment. Here, we interrogate whether priming of epithelial cells by a given matrix stiffness influences their future collective migration on a different matrix – a property we refer to as the ‘mechanical memory’ of migratory cells. To prime cells on a defined matrix and track their collective migration onto an adjoining secondary matrix of dissimilar stiffness, we develop a modular polyacrylamide substrate through step-by-step polymerization of different PA compositions. We report that epithelial cells primed on a stiff matrix migrate faster, display higher actomyosin expression, form larger focal adhesions, and retain nuclear YAP even after arriving onto a soft secondary matrix, as compared to their control behavior on a homogeneously soft matrix. Priming on a soft ECM causes a reverse effect. The depletion of YAP dramatically reduces this memory-dependent migration. Our results present a previously unidentified regulation of mechanosensitive collective cell migration by past matrix stiffness, in which mechanical memory depends on YAP activity.
- Published
- 2017
38. Cancer-associated fibroblasts support vascular growth through mechanical force
- Author
-
Gregory D. Longmore, Sofia M. Joison, Samantha Van Hove Bayer, Elizabeth Crist, Steven C. George, Taylor Hughes, and M. K. Sewell-Loftin
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,Pathology ,Mechanotransduction ,lcsh:Medicine ,Cardiovascular ,Mechanotransduction, Cellular ,Neovascularization ,chemistry.chemical_compound ,Cancer-Associated Fibroblasts ,Neoplasms ,Tumor Microenvironment ,2.1 Biological and endogenous factors ,Aetiology ,lcsh:Science ,Cancer ,rho-Associated Kinases ,Multidisciplinary ,Tumor ,Neovascularization, Pathologic ,Cell biology ,Vascular endothelial growth factor ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,medicine.symptom ,Blood vessel ,medicine.medical_specialty ,Bioengineering ,Biology ,Article ,Cell Line ,03 medical and health sciences ,Vasculogenesis ,Cell Line, Tumor ,Breast Cancer ,medicine ,Humans ,Cell Proliferation ,Pathologic ,Tumor microenvironment ,Neoplastic ,lcsh:R ,030104 developmental biology ,chemistry ,Gene Expression Regulation ,Tumor progression ,lcsh:Q ,Cellular - Abstract
The role of cancer-associated fibroblasts (CAFs) as regulators of tumor progression, specifically vascular growth, has only recently been described. CAFs are thought to be more mechanically active but how this trait may alter the tumor microenvironment is poorly understood. We hypothesized that enhanced mechanical activity of CAFs, as regulated by the Rho/ROCK pathway, contributes to increased blood vessel growth. Using a 3D in vitro tissue model of vasculogenesis, we observed increased vascularization in the presence of breast cancer CAFs compared to normal breast fibroblasts. Further studies indicated this phenomenon was not simply a result of enhanced soluble signaling factors, including vascular endothelial growth factor (VEGF), and that CAFs generated significantly larger deformations in 3D gels compared to normal fibroblasts. Inhibition of the mechanotransductive pathways abrogated the ability of CAFs to deform the matrix and suppressed vascularization. Finally, utilizing magnetic microbeads to mechanically stimulate mechanically-inhibited CAFs showed partial rescue of vascularization. Our studies demonstrate enhanced mechanical activity of CAFs may play a crucial and previously unappreciated role in the formation of tumor-associated vasculature which could possibly offer potential novel targets in future anti-cancer therapies.
- Published
- 2017
39. Transient SNAIL1 Expression Is Necessary for Metastatic Competence in Breast Cancer
- Author
-
Hung D. Tran, Michael Kim, Kaihua Zhang, Gregory D. Longmore, Krishna Luitel, and David Tran
- Subjects
Oncology ,CA15-3 ,Cancer Research ,medicine.medical_specialty ,Transgene ,Breast Neoplasms ,Article ,Metastasis ,Transcriptome ,Breast cancer ,Internal medicine ,Genetic model ,Tumor Microenvironment ,medicine ,Humans ,Neoplasm Metastasis ,Cell Proliferation ,Regulation of gene expression ,Tumor microenvironment ,business.industry ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Female ,Snail Family Transcription Factors ,business ,Signal Transduction ,Transcription Factors - Abstract
SNAIL1 has been suggested to regulate breast cancer metastasis based on analyses of human breast tumor transcriptomes and experiments using cancer cell lines and xenografts. However, in vivo genetic experimental support for a role for SNAIL1 in breast cancer metastasis that develops in an immunocompetent tumor microenvironment has not been determined. To address this question, we created a genetic SNAIL1 model by coupling an endogenous SNAIL1 reporter with an inducible SNAIL1 transgene. Using multiple genetic models of breast cancer, we demonstrated that endogenous SNAIL1 expression was restricted to primary tumors that ultimately disseminate. SNAIL1 gene deletion either during the premalignant phase or after primary tumors have reached a palpable size blunted metastasis, indicating that late metastasis was the main driver of metastasis and that this was dependent on SNAIL1. Importantly, SNAIL1 expression during breast cancer metastasis was transient and forced transient, but not continuous. SNAIL1 expression in breast tumors was sufficient to increase metastasis. Cancer Res; 74(21); 6330–40. ©2014 AACR.
- Published
- 2014
- Full Text
- View/download PDF
40. β-Catenin Serves as a Clutch between Low and High Intercellular E-Cadherin Bond Strengths
- Author
-
Gregory D. Longmore, Denis Wirtz, Yunfeng Feng, and Saumendra Bajpai
- Subjects
Beta-catenin ,Biophysics ,Biology ,Adherens junction ,Extracellular matrix ,Glycogen Synthase Kinase 3 ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Cell Adhesion ,Humans ,Kinase activity ,Cell adhesion ,beta Catenin ,Sequence Deletion ,030304 developmental biology ,Extracellular Matrix Proteins ,0303 health sciences ,Glycogen Synthase Kinase 3 beta ,Base Sequence ,Cadherin ,Cadherins ,Cell biology ,Gene Expression Regulation, Neoplastic ,Cell Biophysics ,Tumor progression ,030220 oncology & carcinogenesis ,Catenin ,Colonic Neoplasms ,biology.protein ,Extracellular Space ,Protein Binding - Abstract
A wide range of invasive pathological outcomes originate from the loss of epithelial phenotype and involve either loss of function or downregulation of transmembrane adhesive receptor complexes, including Ecadherin (Ecad) and binding partners β-catenin and α-catenin at adherens junctions. Cellular pathways regulating wild-type β-catenin level, or direct mutations in β-catenin that affect the turnover of the protein have been shown to contribute to cancer development, through induction of uncontrolled proliferation of transformed tumor cells, particularly in colon cancer. Using single-molecule force spectroscopy, we show that depletion of β-catenin or the prominent cancer-related S45 deletion mutation in β-catenin present in human colon cancers both weaken tumor intercellular Ecad/Ecad bond strength and diminishes the capacity of specific extracellular matrix proteins—including collagen I, collagen IV, and laminin V—to modulate intercellular Ecad/Ecad bond strength through α-catenin and the kinase activity of glycogen synthase kinase 3 (GSK-3β). Thus, in addition to regulating tumor cell proliferation, cancer-related mutations in β-catenin can influence tumor progression by weakening the adhesion of tumor cells to one another through reduced individual Ecad/Ecad bond strength and cellular adhesion to specific components of the extracellular matrix and the basement membrane.
- Published
- 2013
- Full Text
- View/download PDF
41. The Arp2/3 complex mediates multigeneration dendritic protrusions for efficient 3‐dimensional cancer cell migration
- Author
-
Nicholaus J. Trenton, Gregory D. Longmore, Saumendra Bajpai, Hasini Jayatilaka, Denis Wirtz, and Anjil Giri
- Subjects
biology ,Actin-Related Protein 2-3 Complex ,Arp2/3 complex ,Cell migration ,macromolecular substances ,CDC42 ,Cell Surface Extension ,Fibroblasts ,Biochemistry ,Research Communications ,Cell biology ,Gene Expression Regulation, Neoplastic ,Focal adhesion ,Cell Movement ,Cell Line, Tumor ,Genetics ,biology.protein ,Humans ,Cell Surface Extensions ,RNA, Small Interfering ,Lamellipodium ,Molecular Biology ,Cortactin ,Biotechnology - Abstract
Arp2/3 is a protein complex that nucleates actin filament assembly in the lamellipodium in adherent cells crawling on planar 2-dimensional (2D) substrates. However, in physiopathological situations, cell migration typically occurs within a 3-dimensional (3D) environment, and little is known about the role of Arp2/3 and associated proteins in 3D cell migration. Using time resolved live-cell imaging and HT1080, a fibrosarcoma cell line commonly used to study cell migration, we find that the Arp2/3 complex and associated proteins N-WASP, WAVE1, cortactin, and Cdc42 regulate 3D cell migration. We report that this regulation is caused by formation of multigeneration dendritic protrusions, which mediate traction forces on the surrounding matrix and effective cell migration. The primary protrusions emanating directly from the cell body and prolonging the nucleus forms independent of Arp2/3 and dependent on focal adhesion proteins FAK, talin, and p130Cas. The Arp2/3 complex, N-WASP, WAVE1, cortactin, and Cdc42 regulate the secondary protrusions branching off from the primary protrusions. In 3D matrices, fibrosarcoma cells as well as migrating breast, pancreatic, and prostate cancer cells do not display lamellipodial structures. This study characterizes the unique topology of protrusions made by cells in a 3D matrix and show that these dendritic protrusions play a critical role in 3D cell motility and matrix deformation. The relative contribution of these proteins to 3D migration is significantly different from their role in 2D migration.—Giri, A., Bajpai, S., Trenton, N., Jayatilaka, H., Longmore, G. D., Wirtz, D. The Arp2/3 complex mediates multigeneration dendritic protrusions for efficient 3-dimensional cancer cell migration.
- Published
- 2013
- Full Text
- View/download PDF
42. The collagen receptor discoidin domain receptor 2 stabilizes SNAIL1 to facilitate breast cancer metastasis
- Author
-
Gregory D. Longmore, Callie Ann S. Corsa, Julie L. Prior, Kevin W. Eliceiri, Patricia J. Keely, Suzanne M. Ponik, Kun Zhang, and David Piwnica-Worms
- Subjects
Epithelial-Mesenchymal Transition ,Receptors, Collagen ,Breast Neoplasms ,Article ,Receptor tyrosine kinase ,Collagen receptor ,Metastasis ,Mice ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Epithelial–mesenchymal transition ,Neoplasm Metastasis ,Phosphorylation ,RNA, Small Interfering ,Discoidin Domain Receptors ,Cell Proliferation ,Mitogen-Activated Protein Kinase 1 ,Mice, Inbred BALB C ,biology ,Carcinoma, Ductal, Breast ,Receptor Protein-Tyrosine Kinases ,Cancer ,Cell migration ,Cell Biology ,Cadherins ,medicine.disease ,Cell biology ,HEK293 Cells ,Receptors, Mitogen ,biology.protein ,Cancer research ,Female ,RNA Interference ,Snail Family Transcription Factors ,Signal transduction ,Discoidin domain ,Signal Transduction ,Transcription Factors - Abstract
Increased stromal collagen deposition in human breast tumours correlates with metastases. We show that activation of the collagen I receptor DDR2 (discoidin domain receptor 2) regulates SNAIL1 stability by stimulating ERK2 activity, in a Src-dependent manner. Activated ERK2 directly phosphorylates SNAIL1, leading to SNAIL1 nuclear accumulation, reduced ubiquitylation and increased protein half-life. DDR2-mediated stabilization of SNAIL1 promotes breast cancer cell invasion and migration in vitro, and metastasis in vivo. DDR2 expression was observed in most human invasive ductal breast carcinomas studied, and was associated with nuclear SNAIL1 and absence of E-cadherin expression. We propose that DDR2 maintains SNAIL1 level and activity in tumour cells that have undergone epithelial-mesenchymal transition (EMT), thereby facilitating continued tumour cell invasion through collagen-I-rich extracellular matrices by sustaining the EMT phenotype. As such, DDR2 could be an RTK (receptor tyrosine kinase) target for the treatment of breast cancer metastasis.
- Published
- 2013
- Full Text
- View/download PDF
43. α-Actinin1 and 4 tyrosine phosphorylation is critical for stress fiber establishment, maintenance and focal adhesion maturation
- Author
-
Gregory D. Longmore, Hai Ngu, Shannon K. Alford, Frank C.P. Yin, Yunfeng Feng, and Michael E. Ward
- Subjects
Stress fiber ,PTK2 ,macromolecular substances ,Actinin ,Biology ,Article ,Actin cytoskeleton organization ,Zyxin ,Focal adhesion ,chemistry.chemical_compound ,Cell Line, Tumor ,Stress Fibers ,Cell Adhesion ,Humans ,Phosphorylation ,Paxillin ,Focal Adhesions ,Microfilament Proteins ,Cell Differentiation ,Tyrosine phosphorylation ,Cell Biology ,Protein-Tyrosine Kinases ,Phosphoproteins ,Cell biology ,HEK293 Cells ,chemistry ,Focal Adhesion Kinase 1 ,biology.protein ,Tyrosine ,Cell Adhesion Molecules - Abstract
In polarized, migrating cells, stress fibers are a highly dynamic network of contractile acto-myosin structures composed of bundles of actin filaments held together by actin cross-linking proteins such as α-actinins. As such, α-actinins influence actin cytoskeleton organization and dynamics and focal adhesion maturation. In response to environmental signals, α-actinins are tyrosine phosphorylated and this affects their binding to actin stress fibers; however, the cellular role of α-actinin tyrosine phosphorylation remains largely unknown. We found that non-muscle α-actinin1/4 are critical for the establishment of dorsal stress fibers and maintenance of transverse arc stress fibers. Analysis of cells genetically depleted of α-actinin1 and 4 reveals two distinct modes for focal adhesion maturation. An α-actinin1 or 4 dependent mode that uses dorsal stress fiber precursors as a template for establishing focal adhesions and their maturation, and an α-actinin-independent manner that uses transverse arc precursors to establish focal adhesions at both ends. Focal adhesions formed in the absence of α-actinins are delayed in their maturation, exhibit altered morphology, have decreased amounts of Zyxin and VASP, and reduced adhesiveness to extracellular matrix. Further rescue experiments demonstrate that the tyrosine phosphorylation of α-actinin1 at Y12 and α-actinin4 at Y265 is critical for dorsal stress fiber establishment, transverse arc maintenance and focal adhesion maturation.
- Published
- 2013
- Full Text
- View/download PDF
44. Stromal senescence establishes an immunosuppressive microenvironment that drives tumorigenesis
- Author
-
Aude Helene Capietto, Elise Alspach, Megan K. Ruhland, Brian A. Belt, Gregory D. Longmore, Jingqin Luo, Brett L. Knolhoff, Sheila A. Stewart, David G. DeNardo, Roberta Faccio, Andras Schaffer, Kevin C. Flanagan, Xianmin Luo, Andrew J. Loza, John R. Edwards, and Kathleen M. Leahy
- Subjects
Adult ,0301 basic medicine ,Cell signaling ,Stromal cell ,Carcinogenesis ,Science ,General Physics and Astronomy ,Inflammation ,Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Mice ,03 medical and health sciences ,Immune system ,Tumor Microenvironment ,medicine ,Animals ,Antigens, Ly ,Humans ,Immunologic Surveillance ,Cellular Senescence ,Cell Proliferation ,Skin ,Immunosuppression Therapy ,Tumor microenvironment ,CD11b Antigen ,Multidisciplinary ,Innate immune system ,Interleukin-6 ,Myeloid-Derived Suppressor Cells ,General Chemistry ,Fibroblasts ,Middle Aged ,Cell biology ,030104 developmental biology ,Myeloid-derived Suppressor Cell ,Stromal Cells ,medicine.symptom - Abstract
Age is a significant risk factor for the development of cancer. However, the mechanisms that drive age-related increases in cancer remain poorly understood. To determine if senescent stromal cells influence tumorigenesis, we develop a mouse model that mimics the aged skin microenvironment. Using this model, here we find that senescent stromal cells are sufficient to drive localized increases in suppressive myeloid cells that contributed to tumour promotion. Further, we find that the stromal-derived senescence-associated secretory phenotype factor interleukin-6 orchestrates both increases in suppressive myeloid cells and their ability to inhibit anti-tumour T-cell responses. Significantly, in aged, cancer-free individuals, we find similar increases in immune cells that also localize near senescent stromal cells. This work provides evidence that the accumulation of senescent stromal cells is sufficient to establish a tumour-permissive, chronic inflammatory microenvironment that can shelter incipient tumour cells, thus allowing them to proliferate and progress unabated by the immune system., The risk of developing cancer increases with age. Here, the authors address the contribution of age-dependent accumulation of senescent cells within the tumour stroma compartment and show that senescent cells increase the infiltration of myeloid-derived suppressor cells that inhibit cytotoxic T-cells, thus facilitating tumour outgrowth.
- Published
- 2016
- Full Text
- View/download PDF
45. Upholding a role for EMT in breast cancer metastasis
- Author
-
Jing Yang, M. Angela Nieto, Ben Z. Stanger, Thomas Brabletz, Xin Ye, Yibin Kang, Gregory D. Longmore, and Robert A. Weinberg
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Multidisciplinary ,business.industry ,MEDLINE ,Breast cancer metastasis ,Article ,03 medical and health sciences ,030104 developmental biology ,Text mining ,Internal medicine ,embryonic structures ,medicine ,business - Abstract
The cell-biological program termed the epithelial-to-mesenchymal transition (EMT) has been invoked as a critical component of the metastatic process. Contrastingly, Fischer et al.1 recently reported that in two genetically engineered mouse models of mammary tumour development, carcinoma cells could metastasize without activating EMT programs. However, as detailed below, we find their evidence that EMT programs were not expressed in these primary tumours to be insufficient. Therefore, the contribution of EMT to carcinoma metastasis could not be ruled out in their analysis. There is a Reply to this Comment by Fischer, K. R. et al. Nature 547, 10.1038/nature22817 (2017).
- Published
- 2016
46. AJUBA LIM Proteins Limit Hippo Activity in Proliferating Cells by Sequestering the Hippo Core Kinase Complex in the Cytosol
- Author
-
Kun Zhang, Gregory D. Longmore, Gregory V. Schimizzi, Norikazu Yabuta, Andrew J. Loza, Hitoshi Nojima, and Radhika Jagannathan
- Subjects
0301 basic medicine ,endocrine system ,animal structures ,Biology ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,Cytosol ,Animals ,Drosophila Proteins ,Humans ,Wings, Animal ,Hippo Signaling Pathway ,Spotlight ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Hippo signaling pathway ,Cell growth ,Kinase ,fungi ,Intracellular Signaling Peptides and Proteins ,Epithelial Cells ,YAP-Signaling Proteins ,Cell Biology ,Articles ,LIM Domain Proteins ,Phosphoproteins ,Cell biology ,body regions ,030104 developmental biology ,Drosophila melanogaster ,HEK293 Cells ,Hippo signaling ,MCF-7 Cells ,Cancer development ,sense organs ,Signal Transduction ,Transcription Factors - Abstract
The Hippo pathway controls organ growth and is implicated in cancer development. Whether and how Hippo pathway activity is limited to sustain or initiate cell growth when needed is not understood. The members of the AJUBA family of LIM proteins are negative regulators of the Hippo pathway. In mammalian epithelial cells, we found that AJUBA LIM proteins limit Hippo regulation of YAP, in proliferating cells only, by sequestering a cytosolic Hippo kinase complex in which LATS kinase is inhibited. At the plasma membranes of growth-arrested cells, AJUBA LIM proteins do not inhibit or associate with the Hippo kinase complex. The ability of AJUBA LIM proteins to inhibit YAP regulation by Hippo and to associate with the kinase complex directly correlate with their capacity to limit Hippo signaling during Drosophila wing development. AJUBA LIM proteins did not influence YAP activity in response to cell-extrinsic or cell-intrinsic mechanical signals. Thus, AJUBA LIM proteins limit Hippo pathway activity in contexts where cell proliferation is needed.
- Published
- 2016
47. Mismatch in Mechanical and Adhesive Properties Induces Pulsating Cancer Cell Migration in Epithelial Monolayer
- Author
-
Pei Hsun Wu, Meng-Horng Lee, Robert Ros, Gregory D. Longmore, Jack R. Staunton, and Denis Wirtz
- Subjects
Cell ,Biophysics ,Breast Neoplasms ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Myosin ,Cell Adhesion ,medicine ,Cellular Biophysics and Electrophysiology ,Humans ,Neoplasm Invasiveness ,Breast ,Neoplasm Metastasis ,Cell adhesion ,Mechanical Phenomena ,030304 developmental biology ,0303 health sciences ,Chemistry ,Mesenchymal stem cell ,Epithelial Cells ,Adhesion ,Biomechanical Phenomena ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Tumor progression ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Immunology ,alpha Catenin - Abstract
The mechanical and adhesive properties of cancer cells significantly change during tumor progression. Here we assess the functional consequences of mismatched stiffness and adhesive properties between neighboring normal cells on cancer cell migration in an epithelial-like cell monolayer. Using an in vitro coculture system and live-cell imaging, we find that the speed of single, mechanically soft breast carcinoma cells is dramatically enhanced by surrounding stiff nontransformed cells compared with single cells or a monolayer of carcinoma cells. Soft tumor cells undergo a mode of pulsating migration that is distinct from conventional mesenchymal and amoeboid migration, whereby long-lived episodes of slow, random migration are interlaced with short-lived episodes of extremely fast, directed migration, whereas the surrounding stiff cells show little net migration. This bursty migration is induced by the intermittent, myosin II-mediated deformation of the soft nucleus of the cancer cell, which is induced by the transient crowding of the stiff nuclei of the surrounding nontransformed cells, whose movements depend directly on the cadherin-mediated mismatched adhesion between normal and cancer cells as well as α-catenin-based intercellular adhesion of the normal cells. These results suggest that a mechanical and adhesive mismatch between transformed and nontransformed cells in a cell monolayer can trigger enhanced pulsating migration. These results shed light on the role of stiff epithelial cells that neighbor individual cancer cells in early steps of cancer dissemination.
- Published
- 2012
- Full Text
- View/download PDF
48. Temporal and Spatial Cooperation of Snail1 and Twist1 during Epithelial–Mesenchymal Transition Predicts for Human Breast Cancer Recurrence
- Author
-
Rebecca Aft, Gregory D. Longmore, Callie Ann S. Corsa, Hirak Biswas, and David Tran
- Subjects
Cancer Research ,Epithelial-Mesenchymal Transition ,animal structures ,MAP Kinase Signaling System ,Breast Neoplasms ,Biology ,Article ,Metastasis ,Twist transcription factor ,Downregulation and upregulation ,Recurrence ,Transforming Growth Factor beta ,medicine ,Humans ,Epithelial–mesenchymal transition ,Promoter Regions, Genetic ,Molecular Biology ,Regulation of gene expression ,Twist-Related Protein 1 ,Nuclear Proteins ,Epithelial Cells ,Transforming growth factor beta ,Cell Dedifferentiation ,Prognosis ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Oncology ,Gene Knockdown Techniques ,Bone marrow neoplasm ,Cancer cell ,biology.protein ,Cancer research ,Female ,RNA Interference ,Snail Family Transcription Factors ,Bone Marrow Neoplasms ,Protein Binding ,Signal Transduction ,Transcription Factors - Abstract
Epithelial–mesenchymal transition (EMT) is a normal developmental program that is considered to also play an important role in cancer metastasis. Ultimate inducers of EMT are transcriptional repressors that individually can induce experimental EMT, yet in many cells, particularly cancer cells, multiple inducers are expressed simultaneously. Why, and if, and how they interact to regulate EMT is unanswered. Using RNA interference technology to affect protein knockdown and avoid potential overexpression artifact coupled with transient TGFβ treatment to better mimic in vivo conditions we show, in both nontumorigenic and tumorigenic epithelial cancer cells, that Snail1 is uniquely required for EMT initiation, whereas Twist1 is required to maintain late EMT. Twist1, present in resting epithelial cells, is dispensable for EMT initiation. Mechanistically, in response to transient TGFβ treatment, transient Snail1 expression represses Twist1 transcription directly, which is subsequently upregulated, as Snail1 levels decrease, to sustain E-cadherin downregulation and growth arrest of EMT. Persistent Twist1 expression is associated with a p38 and extracellular signal–regulated kinase signal feedback loop that sustains growth-inhibitory signals characteristic of quiescent micrometastatic tumors. This Snail1–Twist1 temporal and spatial cooperation was also observed in vivo during human breast cancer progression to metastasis. Twist1 level, but not Snail1 level, and Twist1:Snail1 ratio in disseminated micrometastatic bone marrow tumor cells was found to correlate with survival and treatment resistance and is highly predictive of metastatic or recurrent disease. Mol Cancer Res; 9(12); 1644–57. ©2011 AACR.
- Published
- 2011
- Full Text
- View/download PDF
49. Lats2 kinase potentiates Snail1 activity by promoting nuclear retention upon phosphorylation
- Author
-
Norikazu Yabuta, Hiroshi Nojima, Kun Zhang, M. Angela Nieto, Gregory D. Longmore, Robert J. Owen, José Manuel Mingot, and Eva Rodriguez-Aznar
- Subjects
General Immunology and Microbiology ,Kinase ,General Neuroscience ,Cell ,Regulator ,Biology ,Subcellular localization ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,medicine.anatomical_structure ,embryonic structures ,medicine ,Phosphorylation ,Kinome ,Epithelial–mesenchymal transition ,Molecular Biology ,Transcription factor - Abstract
Snail1 is a central regulator of epithelial cell adhesion and movement in epithelial-to-mesenchymal transitions (EMTs) during embryo development; a process reactivated during cancer metastasis. While induction of Snail1 transcription precedes EMT induction, post-translational regulation of Snail1 is also critical for determining Snail1's protein level, subcellular localization, and capacity to induce EMT. To identify novel post-translational regulators of Snail1, we developed a live cell, bioluminescence-based screen. From a human kinome RNAi screen, we have identified Lats2 kinase as a novel regulator of Snail1 protein level, subcellular localization, and thus, activity. We show that Lats2 interacts with Snail1 and directly phosphorylates Snail1 at residue T203. This occurs in the nucleus and serves to retain Snail1 in the nucleus thereby enhancing its stability. Lats2 was found to positively influence cellular EMT and tumour cell invasion, in a Snail1-dependent manner. Indeed during TGFβ-induced EMT Lats2 is activated and Snail1 phosphorylated at T203. Analysis in mouse and zebrafish embryo development confirms that Lats2 acts as a positive modulator of Snail1 protein level and potentiates its in vivo EMT activity.
- Published
- 2011
- Full Text
- View/download PDF
50. LIM-domain proteins, LIMD1, Ajuba, and WTIP are required for microRNA-mediated gene silencing
- Author
-
Simon J. Morley, Chia-Ying Chu, Cornelia H. de Moor, Victoria James, Yi Wen Kong, Yungfeng Feng, Tyson V. Sharp, Tim Self, Daniel E. Foxler, Thomas M. Webb, Gregory D. Longmore, Martin Bushell, Tariq M. Rana, Yining Zhang, and Dimitrios Lagos
- Subjects
Cytoplasm ,F-box protein ,Mice ,chemistry.chemical_compound ,microRNA ,Animals ,Gene silencing ,Gene Silencing ,RNA, Messenger ,RNA, Small Interfering ,Gene ,LIM domain ,Genetics ,Messenger RNA ,Multidisciplinary ,biology ,EIF4G ,EIF4E ,Intracellular Signaling Peptides and Proteins ,Proteins ,LIM Domain Proteins ,Biological Sciences ,Cell biology ,MicroRNAs ,Genes ,chemistry ,biology.protein ,Carrier Proteins - Abstract
In recent years there have been major advances with respect to the identification of the protein components and mechanisms of microRNA (miRNA) mediated silencing. However, the complete and precise repertoire of components and mechanism(s) of action remain to be fully elucidated. Herein we reveal the identification of a family of three LIM domain-containing proteins, LIMD1, Ajuba and WTIP (Ajuba LIM proteins) as novel mammalian processing body (P-body) components, which highlight a novel mechanism of miRNA-mediated gene silencing. Furthermore, we reveal that LIMD1, Ajuba, and WTIP bind to Ago1/2, RCK, Dcp2, and eIF4E in vivo, that they are required for miRNA-mediated, but not siRNA-mediated gene silencing and that all three proteins bind to the mRNA 5′ m 7 GTP cap–protein complex. Mechanistically, we propose the Ajuba LIM proteins interact with the m 7 GTP cap structure via a specific interaction with eIF4E that prevents 4EBP1 and eIF4G interaction. In addition, these LIM-domain proteins facilitate miRNA-mediated gene silencing by acting as an essential molecular link between the translationally inhibited eIF4E-m 7 GTP-5 ′ cap and Ago1/2 within the miRISC complex attached to the 3′-UTR of mRNA, creating an inhibitory closed-loop complex.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.