1
|
Qi Y, Yu CH. PI(3,4,5)P3-mediated Cdc42 activation regulates macrophage podosome assembly. Cell Mol Life Sci 2025; 82:127. [PMID: 40126693 PMCID: PMC11933580 DOI: 10.1007/s00018-025-05664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/23/2025] [Accepted: 03/16/2025] [Indexed: 03/26/2025]
Abstract
Podosomes are adhesion structures with densely-polymerized F-actin. While PI(3,4,5)P3 and Cdc42-GTP are known factors to trigger WASP-mediated actin polymerization at the macrophage podosome, their causal mechanism to activate WASP remains unclear. Here, we demonstrate that spatially elevated Cdc42-GTP is a downstream effector of local PI(3,4,5)P3 production at the podosome. We further examine the expression and distribution of 19 Cdc42 guanine exchange factors (GEFs) and identify VAV1 as the key PI(3,4,5)P3-dependent Cdc42 GEF. VAV1 is spatially enriched at the macrophage podosome, and the association of VAV1 with the membrane plays a critical role in upregulating its GEF activity. Reintroduction of wildtype VAV1, rather than the PI(3,4,5)P3-binding deficient or catalytically dead mutants restores the matrix degradation and chemotactic migration of VAV1-knockdown macrophage. Thus, the biogenesis of PI(3,4,5)P3 acts as an upstream signal to locally recruit VAV1 and in turn triggers the guanine nucleotide exchange of Cdc42. Elevated levels of Cdc42-GTP then promote WASP-mediated podosome assembly and macrophage chemotaxis.
Collapse
Affiliation(s)
- Yaoyue Qi
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Cheng-Han Yu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
2
|
Gutierrez-Ruiz OL, Johnson KM, Krueger EW, Nooren RE, Cruz-Reyes N, Heppelmann CJ, Hogenson TL, Fernandez-Zapico ME, McNiven MA, Razidlo GL. Ectopic expression of DOCK8 regulates lysosome-mediated pancreatic tumor cell invasion. Cell Rep 2023; 42:113042. [PMID: 37651233 PMCID: PMC10591794 DOI: 10.1016/j.celrep.2023.113042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 06/22/2023] [Accepted: 08/11/2023] [Indexed: 09/02/2023] Open
Abstract
Amplified lysosome activity is a hallmark of pancreatic ductal adenocarcinoma (PDAC) orchestrated by oncogenic KRAS that mediates tumor growth and metastasis, though the mechanisms underlying this phenomenon remain unclear. Using comparative proteomics, we found that oncogenic KRAS significantly enriches levels of the guanine nucleotide exchange factor (GEF) dedicator of cytokinesis 8 (DOCK8) on lysosomes. Surprisingly, DOCK8 is aberrantly expressed in a subset of PDAC, where it promotes cell invasion in vitro and in vivo. DOCK8 associates with lysosomes and regulates lysosomal morphology and motility, with loss of DOCK8 leading to increased lysosome size. DOCK8 promotes actin polymerization at the surface of lysosomes while also increasing the proteolytic activity of the lysosomal protease cathepsin B. Critically, depletion of DOCK8 significantly reduces cathepsin-dependent extracellular matrix degradation and impairs the invasive capacity of PDAC cells. These findings implicate ectopic expression of DOCK8 as a key driver of KRAS-driven lysosomal regulation and invasion in pancreatic cancer cells.
Collapse
Affiliation(s)
- Omar L Gutierrez-Ruiz
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Katherine M Johnson
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Eugene W Krueger
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Roseanne E Nooren
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicole Cruz-Reyes
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Tara L Hogenson
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Mark A McNiven
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA; Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| | - Gina L Razidlo
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA; Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
3
|
Cervantes-Villagrana RD, García-Jiménez I, Vázquez-Prado J. Guanine nucleotide exchange factors for Rho GTPases (RhoGEFs) as oncogenic effectors and strategic therapeutic targets in metastatic cancer. Cell Signal 2023; 109:110749. [PMID: 37290677 DOI: 10.1016/j.cellsig.2023.110749] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/11/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023]
Abstract
Metastatic cancer cells dynamically adjust their shape to adhere, invade, migrate, and expand to generate secondary tumors. Inherent to these processes is the constant assembly and disassembly of cytoskeletal supramolecular structures. The subcellular places where cytoskeletal polymers are built and reorganized are defined by the activation of Rho GTPases. These molecular switches directly respond to signaling cascades integrated by Rho guanine nucleotide exchange factors (RhoGEFs), which are sophisticated multidomain proteins that control morphological behavior of cancer and stromal cells in response to cell-cell interactions, tumor-secreted factors and actions of oncogenic proteins within the tumor microenvironment. Stromal cells, including fibroblasts, immune and endothelial cells, and even projections of neuronal cells, adjust their shapes and move into growing tumoral masses, building tumor-induced structures that eventually serve as metastatic routes. Here we review the role of RhoGEFs in metastatic cancer. They are highly diverse proteins with common catalytic modules that select among a variety of homologous Rho GTPases enabling them to load GTP, acquiring an active conformation that stimulates effectors controlling actin cytoskeleton remodeling. Therefore, due to their strategic position in oncogenic signaling cascades, and their structural diversity flanking common catalytic modules, RhoGEFs possess unique characteristics that make them conceptual targets of antimetastatic precision therapies. Preclinical proof of concept, demonstrating the antimetastatic effect of inhibiting either expression or activity of βPix (ARHGEF7), P-Rex1, Vav1, ARHGEF17, and Dock1, among others, is emerging.
Collapse
|
4
|
Shalom B, Salaymeh Y, Risling M, Katzav S. Unraveling the Oncogenic Potential of VAV1 in Human Cancer: Lessons from Mouse Models. Cells 2023; 12:cells12091276. [PMID: 37174676 PMCID: PMC10177506 DOI: 10.3390/cells12091276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
VAV1 is a hematopoietic signal transducer that possesses a GDP/GTP nucleotide exchange factor (GEF) that is tightly regulated by tyrosine phosphorylation, along with adapter protein domains, such as SH2 and SH3. Research on VAV1 has advanced over the years since its discovery as an in vitro activated oncogene in an NIH3T3 screen for oncogenes. Although the oncogenic form of VAV1 first identified in the screen has not been detected in human clinical tumors, its wild-type and mutant forms have been implicated in mammalian malignancies of various tissue origins, as well as those of the hematopoietic system. This review article addresses the activity of human VAV1 as an overexpressed or mutated gene and also describes the differences in the distribution of VAV1 mutations in the hematopoietic system and in other tissues. The knowledge accumulated thus far from GEMMs expressing VAV1 is described, with the conclusion that GEMMs of both wild-type VAV1 and mutant VAV1 do not form tumors, yet these will be generated when additional molecular insults, such as loss of p53 or KRAS mutation, occur.
Collapse
Affiliation(s)
- Batel Shalom
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
| | - Yaser Salaymeh
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
| | - Matan Risling
- Department of Military Medicine and "Tzameret", Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
- Medical Corps, Israel Defense Forces, Tel-Hashomer 02149, Israel
| | - Shulamit Katzav
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
| |
Collapse
|
5
|
Joshi VB, Gutierrez Ruiz OL, Razidlo GL. The Cell Biology of Metastatic Invasion in Pancreatic Cancer: Updates and Mechanistic Insights. Cancers (Basel) 2023; 15:cancers15072169. [PMID: 37046830 PMCID: PMC10093482 DOI: 10.3390/cancers15072169] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related mortality worldwide. This is largely due to the lack of routine screening protocols, an absence of symptoms in early-stage disease leading to late detection, and a paucity of effective treatment options. Critically, the majority of patients either present with metastatic disease or rapidly develop metastatic disease. Thus, there is an urgent need to deepen our understanding of metastasis in PDAC. During metastasis, tumor cells escape from the primary tumor, enter the circulation, and travel to a distant site to form a secondary tumor. In order to accomplish this relatively rare event, tumor cells develop an enhanced ability to detach from the primary tumor, migrate into the surrounding matrix, and invade across the basement membrane. In addition, cancer cells interact with the various cell types and matrix proteins that comprise the tumor microenvironment, with some of these factors working to promote metastasis and others working to suppress it. In PDAC, many of these processes are not well understood. The purpose of this review is to highlight recent advances in the cell biology of the early steps of the metastatic cascade in pancreatic cancer. Specifically, we will examine the regulation of epithelial-to-mesenchymal transition (EMT) in PDAC and its requirement for metastasis, summarize our understanding of how PDAC cells invade and degrade the surrounding matrix, and discuss how migration and adhesion dynamics are regulated in PDAC to optimize cancer cell motility. In addition, the role of the tumor microenvironment in PDAC will also be discussed for each of these invasive processes.
Collapse
Affiliation(s)
- Vidhu B Joshi
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Omar L Gutierrez Ruiz
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Gina L Razidlo
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
6
|
Agbaria M, Jbara-Agbaria D, Grad E, Ben-David-Naim M, Aizik G, Golomb G. Nanoparticles of VAV1 siRNA combined with LL37 peptide for the treatment of pancreatic cancer. J Control Release 2023; 355:312-326. [PMID: 36736910 DOI: 10.1016/j.jconrel.2023.01.084] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/10/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the leading causes of cancer-related death, and it is highly resistant to therapy owing to its unique extracellular matrix. VAV1 protein, overexpressed in several cancer diseases including pancreatic cancer (PC), increases tumor proliferation and enhances metastases formation, which are associated with decreased survival. We hypothesized that an additive anti-tumor effect could be obtained by co-encapsulating in PLGA nanoparticles (NPs), the negatively charged siRNA against VAV1 (siVAV1) with the positively charged anti-tumor LL37 peptide, as a counter-ion. Several types of NPs were formulated and were characterized for their physicochemical properties, cellular internalization, and bioactivity in vitro. NPs' biodistribution, toxicity, and bioactivity were examined in a mice PDAC model. An optimal siVAV1 formulation (siVAV1-LL37 NPs) was characterized with desirable physicochemical properties in terms of nano-size, low polydispersity index (PDI), neutral surface charge, high siVAV1 encapsulation efficiency, spherical shape, and long-term shelf-life stability. Cell assays demonstrated rapid engulfment by PC cells, a specific and significant dose-dependent proliferation inhibition, as well as knockdown of VAV1 mRNA levels and migration inhibition in VAV1+ cells. Treatment with siVAV1-LL37 NPs in the mice PDAC model revealed marked accumulation of NPs in the liver and in the tumor, resulting in an increased survival rate following suppression of tumor growth and metastases, mediated via the knockdown of both VAV1 mRNA and protein levels. This proof-of-concept study validates our hypothesis of an additive effect in the treatment of PC facilitated by co-encapsulating siVAV1 in NPs with LL37 serving a dual role as a counter ion as well as an anti-tumor agent.
Collapse
Affiliation(s)
- Majd Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Doaa Jbara-Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Etty Grad
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Meital Ben-David-Naim
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Gil Aizik
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Gershon Golomb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| |
Collapse
|
7
|
Wong DCP, Pan CQ, Er SY, Thivakar T, Rachel TZY, Seah SH, Chua PJ, Jiang T, Chew TW, Chaudhuri PK, Mukherjee S, Salim A, Aye TA, Koh CG, Lim CT, Tan PH, Bay BH, Ridley AJ, Low BC. The scaffold RhoGAP protein ARHGAP8/BPGAP1 synchronizes Rac and Rho signaling to facilitate cell migration. Mol Biol Cell 2023; 34:ar13. [PMID: 36598812 PMCID: PMC10011724 DOI: 10.1091/mbc.e21-03-0099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 12/14/2022] [Accepted: 12/21/2022] [Indexed: 01/05/2023] Open
Abstract
Rho GTPases regulate cell morphogenesis and motility under the tight control of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). However, the underlying mechanism(s) that coordinate their spatiotemporal activities, whether separately or together, remain unclear. We show that a prometastatic RhoGAP, ARHGAP8/BPGAP1, binds to inactive Rac1 and localizes to lamellipodia. BPGAP1 recruits the RacGEF Vav1 under epidermal growth factor (EGF) stimulation and activates Rac1, leading to polarized cell motility, spreading, invadopodium formation, and cell extravasation and promotes cancer cell migration. Importantly, BPGAP1 down-regulates local RhoA activity, which influences Rac1 binding to BPGAP1 and its subsequent activation by Vav1. Our results highlight the importance of BPGAP1 in recruiting Vav1 and Rac1 to promote Rac1 activation for cell motility. BPGAP1 also serves to control the timing of Rac1 activation with RhoA inactivation via its RhoGAP activity. BPGAP1, therefore, acts as a dual-function scaffold that recruits Vav1 to activate Rac1 while inactivating RhoA to synchronize both Rho and Rac signaling in cell motility. As epidermal growth factor receptor (EGFR), Vav1, RhoA, Rac1, and BPGAP1 are all associated with cancer metastasis, BPGAP1 could provide a crucial checkpoint for the EGFR-BPGAP1-Vav1-Rac1-RhoA signaling axis for cancer intervention.
Collapse
Affiliation(s)
| | | | - Shi Yin Er
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - T. Thivakar
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Tan Zi Yi Rachel
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Sock Hong Seah
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Pei Jou Chua
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, Singapore 117594
| | - Tingting Jiang
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - Ti Weng Chew
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | | | - Somsubhro Mukherjee
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Agus Salim
- Melbourne School of Population and Global Health and School of Mathematics and Statistics, The University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Thike Aye Aye
- Department of Pathology, Singapore General Hospital, Singapore 169856
| | - Cheng Gee Koh
- Division of Molecular Genetics & Cell Biology, School of Biological Sciences, Nanyang Technological University, Singapore 637551
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583
| | - Puay Hoon Tan
- Department of Pathology, Singapore General Hospital, Singapore 169856
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Health System, Singapore 117594
| | - Anne J. Ridley
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Boon Chuan Low
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558
- NUS College, National University of Singapore, Singapore 138593
| |
Collapse
|
8
|
Kreider-Letterman G, Castillo A, Mahlandt EK, Goedhart J, Rabino A, Goicoechea S, Garcia-Mata R. ARHGAP17 regulates the spatiotemporal activity of Cdc42 at invadopodia. J Cell Biol 2022; 222:213782. [PMID: 36571786 PMCID: PMC9794838 DOI: 10.1083/jcb.202207020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/18/2022] [Accepted: 11/28/2022] [Indexed: 12/27/2022] Open
Abstract
Invadopodia formation is regulated by Rho GTPases. However, the molecular mechanisms that control Rho GTPase signaling at invadopodia remain poorly understood. Here, we have identified ARHGAP17, a Cdc42-specific RhoGAP, as a key regulator of invadopodia in breast cancer cells and characterized a novel ARHGAP17-mediated signaling pathway that controls the spatiotemporal activity of Cdc42 during invadopodia turnover. Our results show that during invadopodia assembly, ARHGAP17 localizes to the invadopodia ring and restricts the activity of Cdc42 to the invadopodia core, where it promotes invadopodia growth. Invadopodia disassembly starts when ARHGAP17 translocates from the invadopodia ring to the core, in a process that is mediated by its interaction with the Cdc42 effector CIP4. Once at the core, ARHGAP17 inactivates Cdc42 to promote invadopodia disassembly. Our results in invadopodia provide new insights into the coordinated transition between the activation and inactivation of Rho GTPases.
Collapse
Affiliation(s)
| | - Abel Castillo
- https://ror.org/01pbdzh19Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Eike K. Mahlandt
- https://ror.org/04dkp9463Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Joachim Goedhart
- https://ror.org/04dkp9463Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Agustin Rabino
- https://ror.org/01pbdzh19Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Silvia Goicoechea
- https://ror.org/01pbdzh19Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Rafael Garcia-Mata
- https://ror.org/01pbdzh19Department of Biological Sciences, University of Toledo, Toledo, OH, USA,Correspondence to Rafael Garcia-Mata:
| |
Collapse
|
9
|
Abstract
Cell migration, a crucial step in numerous biological processes, is tightly regulated in space and time. Cells employ Rho GTPases, primarily Rho, Rac, and Cdc42, to regulate their motility. Like other small G proteins, Rho GTPases function as biomolecular switches in regulating cell migration by operating between GDP bound 'OFF' and GTP bound 'ON' states. Guanine nucleotide exchange factors (GEFs) catalyse the shuttling of GTPases from OFF to ON state. G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors that are involved in many signalling phenomena including cell survival and cell migration events. In this review, we summarize signalling mechanisms, involving GPCRs, leading to the activation of RhoGEFs. GPCRs exhibit diverse GEF activation modes that include the interaction of heterotrimeric G protein subunits with different domains of GEFs, phosphorylation, protein-protein interaction, protein-lipid interaction, and/or a combination of these processes.
Collapse
Affiliation(s)
- Aishwarya Omble
- Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kiran Kulkarni
- Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India,CONTACT Kiran Kulkarni Academy of Scientific and Innovative Research (Acsir), Ghaziabad 201002, India
| |
Collapse
|
10
|
Hülsemann M, Sanchez C, Verkhusha PV, Des Marais V, Mao SPH, Donnelly SK, Segall JE, Hodgson L. TC10 regulates breast cancer invasion and metastasis by controlling membrane type-1 matrix metalloproteinase at invadopodia. Commun Biol 2021; 4:1091. [PMID: 34531530 PMCID: PMC8445963 DOI: 10.1038/s42003-021-02583-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 08/23/2021] [Indexed: 01/12/2023] Open
Abstract
During breast cancer metastasis, cancer cell invasion is driven by actin-rich protrusions called invadopodia, which mediate the extracellular matrix degradation required for the success of the invasive cascade. In this study, we demonstrate that TC10, a member of a Cdc42 subfamily of p21 small GTPases, regulates the membrane type 1 matrix metalloproteinase (MT1-MMP)-driven extracellular matrix degradation at invadopodia. We show that TC10 is required for the plasma membrane surface exposure of MT1-MMP at these structures. By utilizing our Förster resonance energy transfer (FRET) biosensor, we demonstrate the p190RhoGAP-dependent regulation of spatiotemporal TC10 activity at invadopodia. We identified a pathway that regulates invadopodia-associated TC10 activity and function through the activation of p190RhoGAP and the downstream interacting effector Exo70. Our findings reveal the role of a previously unknown regulator of vesicular fusion at invadopodia, TC10 GTPase, in breast cancer invasion and metastasis.
Collapse
Affiliation(s)
- Maren Hülsemann
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Colline Sanchez
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Polina V Verkhusha
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Vera Des Marais
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Serena P H Mao
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Sara K Donnelly
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Jeffrey E Segall
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Louis Hodgson
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
11
|
Rivier P, Mubalama M, Destaing O. Small GTPases all over invadosomes. Small GTPases 2021; 12:429-439. [PMID: 33487105 PMCID: PMC8583085 DOI: 10.1080/21541248.2021.1877081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/30/2020] [Accepted: 01/10/2021] [Indexed: 12/19/2022] Open
Abstract
Cell invasion is associated with numerous patho-physiologic states including cell development and metastatic dissemination. This process couples the activation of cell motility with the capacity to degrade the extracellular matrix, thereby permitting cells to pass through basal membranes. Invasion is sustained by the actions of invadosomes, an ensemble of subcellular structures with high functional homology. Invadosomes are 3D acto-adhesive structures that can also mediate local extracellular matrix degradation through the controlled delivery of proteases. Intracellular RHO GTPases play a central role in the regulation of invadosomes where their complex interplay regulates multiple invadosome functions. This review aims to provide an overview of the synergistic activities of the small GTPases in invadosome biology. This broad-based review also reinforces the importance of the spatiotemporal regulation of small GTPases and the impact of this process on invadosome dynamics.
Collapse
Affiliation(s)
- Paul Rivier
- Team DYSAD, Dept2, Institute for Advanced Biosciences, Centre de Recherche Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Grenoble, France
| | - Michel Mubalama
- Team DYSAD, Dept2, Institute for Advanced Biosciences, Centre de Recherche Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Grenoble, France
| | - Olivier Destaing
- Team DYSAD, Dept2, Institute for Advanced Biosciences, Centre de Recherche Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Grenoble, France
| |
Collapse
|
12
|
Boudria R, Laurienté V, Oudar A, Harouna-Rachidi S, Dondi E, Le Roy C, Gardano L, Varin-Blank N, Guittat L. Regulatory interplay between Vav1, Syk and β-catenin occurs in lung cancer cells. Cell Signal 2021; 86:110079. [PMID: 34252536 DOI: 10.1016/j.cellsig.2021.110079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 01/15/2023]
Abstract
Vav1 exhibits two signal transducing properties as an adaptor protein and a regulator of cytoskeleton organization through its Guanine nucleotide Exchange Factor module. Although the expression of Vav1 is restricted to the hematopoietic lineage, its ectopic expression has been unraveled in a number of solid tumors. In this study, we show that in lung cancer cells, as such in hematopoietic cells, Vav1 interacts with the Spleen Tyrosine Kinase, Syk. Likewise, Syk interacts with β-catenin and, together with Vav1, regulates the phosphorylation status of β-catenin. Depletion of Vav1, Syk or β-catenin inhibits Rac1 activity and decreases cell migration suggesting the interplay of the three effectors to a common signaling pathway. This model is further supported by the finding that in turn, β-catenin regulates the transcription of Syk gene expression. This study highlights the elaborated connection between Vav1, Syk and β-catenin and the contribution of the trio to cell migration.
Collapse
Affiliation(s)
- Rofia Boudria
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Vanessa Laurienté
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Antonin Oudar
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Souleymane Harouna-Rachidi
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Elisabetta Dondi
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Christine Le Roy
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Laura Gardano
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France
| | - Nadine Varin-Blank
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France.
| | - Lionel Guittat
- INSERM, UMR 978, Bobigny, France; Labex Inflamex, Université Sorbonne Paris Nord, UFR SMBH, Bobigny, France.
| |
Collapse
|
13
|
Luo Y, Hu J, Liu Y, Li L, Li Y, Sun B, Kong R. Invadopodia: A potential target for pancreatic cancer therapy. Crit Rev Oncol Hematol 2021; 159:103236. [PMID: 33482351 DOI: 10.1016/j.critrevonc.2021.103236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/05/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
Dissemination of cancer cells is an intricate multistep process that represents the most deadly aspect of cancer. Cancer cells form F-actin-rich protrusions known as invadopodia to invade surrounding tissues, blood vessels and lymphatics. A number of studies have demonstrated the significant roles of invadopodia in cancer. Therefore, the specific cells and molecules involved in invadopodia activity can provide as therapeutic targets. In this review, we included a thorough overview of studies in invadopodia and discussed their functions in cancer metastasis. We then presented the specific cells and molecules involved in invadopodia activity in pancreatic cancer and analyzed their suitability to be effective therapeutic targets. Currently, drugs targeting invadopodia and relevant clinical trials are negligible. Here, we highlighted the significance of potential drugs and discussed future obstacles in implementing clinical trials. This review presents a new perspective on invadopodia-induced pancreatic cancer metastasis and may prosper the development of targeted therapeutics against pancreatic cancer.
Collapse
Affiliation(s)
- Yan Luo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
14
|
Mondal C, Di Martino JS, Bravo-Cordero JJ. Actin dynamics during tumor cell dissemination. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 360:65-98. [PMID: 33962751 PMCID: PMC8246644 DOI: 10.1016/bs.ircmb.2020.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The actin cytoskeleton is a dynamic network that regulates cellular behavior from development to disease. By rearranging the actin cytoskeleton, cells are capable of migrating and invading during developmental processes; however, many of these cellular properties are hijacked by cancer cells to escape primary tumors and disseminate to distant organs in the body. In this review article, we highlight recent work describing how cancer cells regulate the actin cytoskeleton to achieve efficient invasion and metastatic colonization. We also review new imaging technologies that are capable of revealing the complex architecture and regulation of the actin cytoskeleton during motility and invasion of tumor cells.
Collapse
Affiliation(s)
- Chandrani Mondal
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Julie S Di Martino
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jose Javier Bravo-Cordero
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
15
|
Vav1 Sustains the In Vitro Differentiation of Normal and Tumor Precursors to Insulin Producing Cells Induced by all-Trans Retinoic Acid (ATRA). Stem Cell Rev Rep 2020; 17:673-684. [PMID: 33165749 PMCID: PMC8036226 DOI: 10.1007/s12015-020-10074-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
All-trans retinoic acid (ATRA) promotes the development and the function of insulin producing cells and induces partial differentiation of pancreatic tumor cells. A number of evidences clearly indicate that the ATRA mediated signaling may have a substantial role in therapeutic approaches based on restoration of functional β-cells. Among the proteins up-regulated by ATRA, Vav1 is involved in maturation and function of haematopoietic cells and is essential for retinoids induced differentiation of tumor promyelocytes. The presence of Vav1 in solid tissues, including pancreas, is considered ectopic and no role in the differentiation of human epithelial cells has so far been described. We demonstrated here that Vav1 sustains the maturation to β-cells of the normal precursors human Biliary Tree Stem/progenitor Cells (hBTSCs) induced by a differentiation medium containing ATRA and that, in the mature normal pancreas, insulin-producing cells express variable levels of Vav1. Using pancreatic ductal adenocarcinoma (PDAC)-derived cells, we also revealed that the ATRA induced up-modulation of Vav1 is essential for the retinoid-induced trans-differentiation of neoplastic cells into insulin producing cells. The results of this study identify Vav1 as crucial molecule in ATRA induced maturation of insulin producing cells and suggest this protein as a marker for new strategies ended to restore functional β-cells. Graphical abstract ![]()
Collapse
|
16
|
The role of actin and myosin in antigen extraction by B lymphocytes. Semin Cell Dev Biol 2019; 102:90-104. [PMID: 31862219 DOI: 10.1016/j.semcdb.2019.10.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/14/2019] [Accepted: 10/31/2019] [Indexed: 12/14/2022]
Abstract
B cells must extract antigens attached to the surface of antigen presenting cells to generate high-affinity antibodies. Antigen extraction requires force, and recent studies have implicated actomyosin-dependent pulling forces generated within the B cell as the major driver of antigen extraction. These actomyosin-dependent pulling forces also serve to test the affinity of the B cell antigen receptor for antigen prior to antigen extraction. Such affinity discrimination is central to the process of antibody affinity maturation. Here we review the evidence that actomyosin-dependent pulling forces generated within the B cell promote affinity discrimination and power antigen extraction. Our take on these critical B cell functions is influenced significantly by the recent identification of formin-generated, myosin-rich, concentric actin arcs in the medial portion of the T cell immune synapse, as B cells appear to contain a similar contractile actomyosin structure.
Collapse
|
17
|
Schomberg J. Identification of Targetable Pathways in Oral Cancer Patients via Random Forest and Chemical Informatics. Cancer Inform 2019; 18:1176935119889911. [PMID: 31819345 PMCID: PMC6883365 DOI: 10.1177/1176935119889911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023] Open
Abstract
Treatment of head and neck cancer has been slow to change with epidermal growth
factor receptor (EGFR) inhibitors, PD1 inhibitors, and
taxane-/plant-alkaloid-derived chemotherapies being the only therapies approved
by the U.S. Food and Drug Administration (FDA) in the last 10 years for the
treatment of head and neck cancers. Head and neck cancer is a relatively rare
cancer compared to breast or lung cancers. However, it is possible that existing
therapies for more common solid tumors or for the treatment of other diseases
could also prove effective against oral cancers. Many therapies have molecular
targets that could be appropriate in oral cancer as well as the cancer in which
the drug gained initial FDA approval. Also, there may be targets in oral cancer
for which existing FDA-approved drugs could be applied. This study describes
informatics methods that use machine learning to identify influential gene
targets in patients receiving platinum-based chemotherapy, non-platinum-based
chemotherapy, and genes influential in both groups of patients. This analysis
yielded 6 small molecules that had a high Tanimoto similarity (>50%) to
ligands binding genes shown to be highly influential in determining treatment
response in oral cancer patients. In addition to influencing treatment response,
these genes were also found to act as gene hubs connected to more than 100 other
genes in pathways enriched with genes determined to be influential in treatment
response by a random forest classifier with 20 000 trees trying 320 variables at
each tree node. This analysis validates the use of multiple informatics methods
to identify small molecules that have a greater likelihood of efficacy in a
given cancer of interest.
Collapse
Affiliation(s)
- John Schomberg
- CHOC Children's, Orange, CA, USA.,School of Population Health Science, University of California Irvine, Irvine, CA, USA.,Afecta Pharmaceuticals, Irvine, CA, USA
| |
Collapse
|
18
|
Hammer JA, Wang JC, Saeed M, Pedrosa AT. Origin, Organization, Dynamics, and Function of Actin and Actomyosin Networks at the T Cell Immunological Synapse. Annu Rev Immunol 2019; 37:201-224. [PMID: 30576253 PMCID: PMC8343269 DOI: 10.1146/annurev-immunol-042718-041341] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The engagement of a T cell with an antigen-presenting cell (APC) or activating surface results in the formation within the T cell of several distinct actin and actomyosin networks. These networks reside largely within a narrow zone immediately under the T cell's plasma membrane at its site of contact with the APC or activating surface, i.e., at the immunological synapse. Here we review the origin, organization, dynamics, and function of these synapse-associated actin and actomyosin networks. Importantly, recent insights into the nature of these actin-based cytoskeletal structures were made possible in several cases by advances in light microscopy.
Collapse
Affiliation(s)
- John A Hammer
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Jia C Wang
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Mezida Saeed
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Antonio T Pedrosa
- Cell Biology and Physiology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| |
Collapse
|
19
|
Chen YC, Baik M, Byers JT, Chen KT, French SW, Díaz B. TKS5-positive invadopodia-like structures in human tumor surgical specimens. Exp Mol Pathol 2018; 106:17-26. [PMID: 30439350 DOI: 10.1016/j.yexmp.2018.11.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022]
Abstract
Invadopodia, cancer cell protrusions with proteolytic activity, are functionally associated with active remodeling of the extracellular matrix. Here, we show that the invadopodia-related protein TKS5 is expressed in human pancreatic adenocarcinoma lines, and demonstrate that pancreatic cancer cells depend on TKS5 for invadopodia formation and function. Immunofluorescence staining of human pancreatic cancer cells reveals that TKS5 is a marker of mature and immature invadopodia. We also analyze the co-staining patterns of TKS5 and the commonly used invadopodia marker Cortactin, and find only partial co-localization of these two proteins at invadopodia, with a large fraction of TKS5-positive invadopodia lacking detectable levels of Cortactin. Whereas compelling evidence exist on the role of invadopodia as mediators of invasive migration in cultured cells and in animal models of cancer, these structures have never been detected inside human tumors. Here, using antibodies against TKS5 and Cortactin, we describe for the first time structures strongly resembling invadopodia in various paraffin-embedded human tumor surgical specimens from pancreas and other organs. Our results strongly suggest that invadopodia are present inside human tumors, and warrants further investigation on their regulation and occurrence in surgical specimens, and on the value of TKS5 antibodies as pathological research and diagnostic tools.
Collapse
Affiliation(s)
- Yu-Chuan Chen
- Division on Medical Oncology Hematology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Matthew Baik
- Division on Medical Oncology Hematology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joshua T Byers
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kathryn T Chen
- Department of Surgery, Harbor-UCLA Medical Center, Torrance, CA, USA; David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Samuel W French
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, CA, USA; David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Begoña Díaz
- Division on Medical Oncology Hematology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA; David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Lawson CD, Ridley AJ. Rho GTPase signaling complexes in cell migration and invasion. J Cell Biol 2018; 217:447-457. [PMID: 29233866 PMCID: PMC5800797 DOI: 10.1083/jcb.201612069] [Citation(s) in RCA: 357] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/23/2017] [Accepted: 11/17/2017] [Indexed: 12/02/2022] Open
Abstract
Cell migration is dependent on the dynamic formation and disassembly of actin filament-based structures, including lamellipodia, filopodia, invadopodia, and membrane blebs, as well as on cell-cell and cell-extracellular matrix adhesions. These processes all involve Rho family small guanosine triphosphatases (GTPases), which are regulated by the opposing actions of guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Rho GTPase activity needs to be precisely tuned at distinct cellular locations to enable cells to move in response to different environments and stimuli. In this review, we focus on the ability of RhoGEFs and RhoGAPs to form complexes with diverse binding partners, and describe how this influences their ability to control localized GTPase activity in the context of migration and invasion.
Collapse
Affiliation(s)
- Campbell D Lawson
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, England, UK
| | - Anne J Ridley
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, England, UK
| |
Collapse
|
21
|
Sherwood DR, Plastino J. Invading, Leading and Navigating Cells in Caenorhabditis elegans: Insights into Cell Movement in Vivo. Genetics 2018; 208:53-78. [PMID: 29301948 PMCID: PMC5753875 DOI: 10.1534/genetics.117.300082] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 10/26/2017] [Indexed: 12/30/2022] Open
Abstract
Highly regulated cell migration events are crucial during animal tissue formation and the trafficking of cells to sites of infection and injury. Misregulation of cell movement underlies numerous human diseases, including cancer. Although originally studied primarily in two-dimensional in vitro assays, most cell migrations in vivo occur in complex three-dimensional tissue environments that are difficult to recapitulate in cell culture or ex vivo Further, it is now known that cells can mobilize a diverse repertoire of migration modes and subcellular structures to move through and around tissues. This review provides an overview of three distinct cellular movement events in Caenorhabditis elegans-cell invasion through basement membrane, leader cell migration during organ formation, and individual cell migration around tissues-which together illustrate powerful experimental models of diverse modes of movement in vivo We discuss new insights into migration that are emerging from these in vivo studies and important future directions toward understanding the remarkable and assorted ways that cells move in animals.
Collapse
Affiliation(s)
- David R Sherwood
- Department of Biology, Regeneration Next, Duke University, Durham, North Carolina 27705
| | - Julie Plastino
- Institut Curie, PSL Research University, CNRS, UMR 168, F-75005 Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, UMR 168, F-75005 Paris, France
| |
Collapse
|
22
|
Li M, Wang J, Mo B, Zeng J, Yao D, Chen F, Jiang M, Rao L, Du Y. Total alkaloids of Corydalis saxicola bunting inhibits migration of A549 cells by suppressing Cdc42 or Vav1. Oncol Lett 2017; 15:475-482. [PMID: 29285198 DOI: 10.3892/ol.2017.7273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/15/2017] [Indexed: 12/19/2022] Open
Abstract
Cell division cycle 42 (Cdc42) is a critical regulator, which functions in cancer metastasis. Numerous previous studies have demonstrated that vav guanine nucleotide exchange factor 1 (Vav1) is ectopically expressed in numerous types of human malignancies and have suggested that Vav1 may efficiently promote the formation of invadopodia and matrix degradation by regulating the activation of Cdc42. Total alkaloids of Corydalis saxicola bunting (TAOCSB), a type of alkaloid compound extracted from the root of C. saxicola bunting, has been revealed to have anticancer properties. However, there is no available information to address the effects of TAOCSB on the metastasis of human lung cancer. In the present study, the anticancer effect on A549 non-small cell lung cancer cells induced by TAOCSB was investigated, as well as its underlying mechanisms. The results demonstrated that a low dose of TAOCSB exhibited anti-metastatic potential in suppressing the invasion and migration of A549 cells, and this action may be involved in TAOCSB-mediated inhibition of Cdc42 expression at the level of mRNA and protein in parallel with TAOCSB-mediated inhibition of matrix metalloproteinase (MMP)-2 and MMP-9 protein expression levels. Although the present study did not reveal the expression level of Vav1 protein in A549 cells, the expression level of Vav1 mRNA was investigated. The effect of Vav1 expression in A549 cells requires further study. Overall, the results of the present study revealed that TAOCSB may provide more information regarding lung cancer treatment.
Collapse
Affiliation(s)
- Mimi Li
- Institute of Respiratory Diseases, Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Jiying Wang
- Department of Respiratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Biwen Mo
- Department of Respiratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Jinrong Zeng
- Department of Respiratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Dong Yao
- Department of Respiratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Feng Chen
- Department of Respiratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Ming Jiang
- Department of Respiratory, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Lizong Rao
- Institute of Respiratory Diseases, Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Yinjun Du
- Institute of Respiratory Diseases, Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| |
Collapse
|
23
|
Morris DC, Popp JL, Tang LK, Gibbs HC, Schmitt E, Chaki SP, Bywaters BC, Yeh AT, Porter WW, Burghardt RC, Barhoumi R, Rivera GM. Nck deficiency is associated with delayed breast carcinoma progression and reduced metastasis. Mol Biol Cell 2017; 28:3500-3516. [PMID: 28954862 PMCID: PMC5683761 DOI: 10.1091/mbc.e17-02-0106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 09/15/2017] [Accepted: 09/20/2017] [Indexed: 12/16/2022] Open
Abstract
Nck promotes breast carcinoma progression and metastasis by directing the polarized interaction of carcinoma cells with collagen fibrils, decreasing actin turnover, and enhancing the localization and activity of MMP14 at the cell surface through modulation of the spatiotemporal activation of Cdc42 and RhoA. Although it is known that noncatalytic region of tyrosine kinase (Nck) regulates cell adhesion and migration by bridging tyrosine phosphorylation with cytoskeletal remodeling, the role of Nck in tumorigenesis and metastasis has remained undetermined. Here we report that Nck is required for the growth and vascularization of primary tumors and lung metastases in a breast cancer xenograft model as well as extravasation following injection of carcinoma cells into the tail vein. We provide evidence that Nck directs the polarization of cell–matrix interactions for efficient migration in three-dimensional microenvironments. We show that Nck advances breast carcinoma cell invasion by regulating actin dynamics at invadopodia and enhancing focalized extracellular matrix proteolysis by directing the delivery and accumulation of MMP14 at the cell surface. We find that Nck-dependent cytoskeletal changes are mechanistically linked to enhanced RhoA but restricted spatiotemporal activation of Cdc42. Using a combination of protein silencing and forced expression of wild-type/constitutively active variants, we provide evidence that Nck is an upstream regulator of RhoA-dependent, MMP14-mediated breast carcinoma cell invasion. By identifying Nck as an important driver of breast carcinoma progression and metastasis, these results lay the groundwork for future studies assessing the therapeutic potential of targeting Nck in aggressive cancers.
Collapse
Affiliation(s)
- David C Morris
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Julia L Popp
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Leung K Tang
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Holly C Gibbs
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-4467
| | - Emily Schmitt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Sankar P Chaki
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Briana C Bywaters
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| | - Alvin T Yeh
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-4467
| | - Weston W Porter
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Rola Barhoumi
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas 77843-4467
| | - Gonzalo M Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas 77843-4467
| |
Collapse
|
24
|
Dagliyan O, Tarnawski M, Chu PH, Shirvanyants D, Schlichting I, Dokholyan NV, Hahn KM. Engineering extrinsic disorder to control protein activity in living cells. Science 2017; 354:1441-1444. [PMID: 27980211 DOI: 10.1126/science.aah3404] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 11/16/2016] [Indexed: 11/03/2022]
Abstract
Optogenetic and chemogenetic control of proteins has revealed otherwise inaccessible facets of signaling dynamics. Here, we use light- or ligand-sensitive domains to modulate the structural disorder of diverse proteins, thereby generating robust allosteric switches. Sensory domains were inserted into nonconserved, surface-exposed loops that were tight and identified computationally as allosterically coupled to active sites. Allosteric switches introduced into motility signaling proteins (kinases, guanosine triphosphatases, and guanine exchange factors) controlled conversion between conformations closely resembling natural active and inactive states, as well as modulated the morphodynamics of living cells. Our results illustrate a broadly applicable approach to design physiological protein switches.
Collapse
Affiliation(s)
- Onur Dagliyan
- Program in Molecular and Cellular Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Miroslaw Tarnawski
- Department of Biomolecular Mechanisms, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Pei-Hsuan Chu
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David Shirvanyants
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ilme Schlichting
- Department of Biomolecular Mechanisms, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Nikolay V Dokholyan
- Program in Molecular and Cellular Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. .,Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Klaus M Hahn
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
25
|
Rosenberg BJ, Gil-Henn H, Mader CC, Halo T, Yin T, Condeelis J, Machida K, Wu YI, Koleske AJ. Phosphorylated cortactin recruits Vav2 guanine nucleotide exchange factor to activate Rac3 and promote invadopodial function in invasive breast cancer cells. Mol Biol Cell 2017; 28:1347-1360. [PMID: 28356423 PMCID: PMC5426849 DOI: 10.1091/mbc.e16-12-0885] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/20/2017] [Accepted: 03/22/2017] [Indexed: 12/12/2022] Open
Abstract
Phosphorylation of cortactin downstream of the EGF receptor–Src-Arg kinase cascade triggers maturation of invadopodia, actin-rich protrusions that breast cancer cells use to invade the extracellular matrix. Phosphocortactin recruits Vav2 to invadopodia to activate Rac3 and support actin polymerization, matrix degradation, and invasion. Breast carcinoma cells use specialized, actin-rich protrusions called invadopodia to degrade and invade through the extracellular matrix. Phosphorylation of the actin nucleation–promoting factor and actin-stabilizing protein cortactin downstream of the epidermal growth factor receptor–Src-Arg kinase cascade is known to be a critical trigger for invadopodium maturation and subsequent cell invasion in breast cancer cells. The functions of cortactin phosphorylation in this process, however, are not completely understood. We identify the Rho-family guanine nucleotide exchange factor Vav2 in a comprehensive screen for human SH2 domains that bind selectively to phosphorylated cortactin. We demonstrate that the Vav2 SH2 domain binds selectively to phosphotyrosine-containing peptides corresponding to cortactin tyrosines Y421 and Y466 but not to Y482. Mutation of the Vav2 SH2 domain disrupts its recruitment to invadopodia, and an SH2-domain mutant form of Vav2 cannot support efficient matrix degradation in invasive MDA-MB-231 breast cancer cells. We show that Vav2 function is required for promoting invadopodium maturation and consequent actin polymerization, matrix degradation, and invasive migratory behavior. Using biochemical assays and a novel Rac3 biosensor, we show that Vav2 promotes Rac3 activation at invadopodia. Rac3 knockdown reduces matrix degradation by invadopodia, whereas a constitutively active Rac3 can rescue the deficits in invadopodium function in Vav2-knockdown cells. Together these data indicate that phosphorylated cortactin recruits Vav2 to activate Rac3 and promote invadopodial maturation in invasive breast cancer cells.
Collapse
Affiliation(s)
| | - Hava Gil-Henn
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311520, Israel
| | | | - Tiffany Halo
- Department of Chemistry, Yale University, New Haven, CT 06520
| | - Taofei Yin
- Raymond and Beverly Sackler Laboratory of Genetics and Molecular Medicine, Department of Genetics and Genome Sciences and Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
| | - John Condeelis
- Department of Anatomy and Structural Biology and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Kazuya Machida
- Raymond and Beverly Sackler Laboratory of Genetics and Molecular Medicine, Department of Genetics and Genome Sciences and Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Yi I Wu
- Raymond and Beverly Sackler Laboratory of Genetics and Molecular Medicine, Department of Genetics and Genome Sciences and Center for Cell Analysis and Modeling, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520 .,Department of Neuroscience, Yale University, New Haven, CT 06520
| |
Collapse
|
26
|
Zhu X, Jin H, Xia Z, Wu X, Yang M, Zhang H, Shang X, Cheng R, Zhan Z, Yu Z. Vav1 expression is increased in esophageal squamous cell carcinoma and indicates poor prognosis. Biochem Biophys Res Commun 2017; 486:571-576. [PMID: 28336434 DOI: 10.1016/j.bbrc.2017.03.091] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/18/2017] [Indexed: 01/16/2023]
Abstract
Recently, Vav1 has been suggested to play an essential role in the progression of human cancers. However, the correlation between Vav1 expression and prognosis of esophageal squamous cell carcinoma (ESCC) is still unknown. The aim of this study was to investigate Vav1 expression and its prognostic value in ESCC. The expression of Vav1 was detected by real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blotting in ESCC tissues and matched nontumorous tissues. Immunohistochemistry (IHC) was carried out to detect Vav1 expression in paraffin samples from 112 primary ESCC patients. Survival analysis was performed using Kaplan-Meier method. Univariate and multivariate Cox regression analyses were performed to evaluate the correlation of Vav1 expression with prognosis of ESCC patients. The expression levels of Vav1 mRNA and protein in ESCC tissues were both significantly higher than those in adjacent nontumorous tissues. High Vav1 expression was significantly correlated with larger tumor size (P = 0.015), depth of tumor invasion (P = 0.023), lymph node metastasis (P = 0.008) and TNM stage (P < 0.001). The rate of overall survival (OS) was significantly lower in patients with high Vav1 expression than those with low Vav1 expression (P = 0.014). Multivariate Cox analysis indicated that Vav1 expression is an independent prognostic factor for OS (HR = 1.660, 95%CI = 1.058-2.607, P = 0.028). In summary, our findings demonstrate that Vav1 may be a candidate molecular prognostic marker for patients with ESCC.
Collapse
Affiliation(s)
- Xiaolei Zhu
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin 300060, China; Department of Thoracic Surgery, Second Hospital of Tianjin Medical University, Tianjin 300060, China
| | - Huan Jin
- Department of Nursing, Second Hospital of Tianjin Medical University, Tianjin 300060, China
| | - Ziwei Xia
- Department of Thoracic Surgery, Second Hospital of Tianjin Medical University, Tianjin 300060, China
| | - Xianxian Wu
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin 300060, China
| | - Mingjian Yang
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin 300060, China
| | - Hongdian Zhang
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin 300060, China
| | - Xiaobin Shang
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin 300060, China
| | - Runfen Cheng
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin 300060, China
| | - Zhongli Zhan
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin 300060, China
| | - Zhentao Yu
- Department of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin City, Tianjin 300060, China.
| |
Collapse
|
27
|
Du L, Ning Z, Zhang H, Liu F. Corepressor metastasis-associated protein 3 modulates epithelial-to-mesenchymal transition and metastasis. CHINESE JOURNAL OF CANCER 2017; 36:28. [PMID: 28279208 PMCID: PMC5345190 DOI: 10.1186/s40880-017-0193-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 02/22/2017] [Indexed: 02/05/2023]
Abstract
Worldwide, metastasis is the leading cause of more than 90% of cancer-related deaths. Currently, no specific therapies effectively impede metastasis. Metastatic processes are controlled by complex regulatory networks and transcriptional hierarchy. Corepressor metastasis-associated protein 3 (MTA3) has been confirmed as a novel component of nucleosome remodeling and histone deacetylation (NuRD). Increasing evidence supports the theory that, in the recruitment of transcription factors, coregulators function as master regulators rather than passive passengers. As a master regulator, MTA3 governs the target selection for NuRD and functions as a transcriptional repressor. MTA3 dysregulation is associated with tumor progression, invasion, and metastasis in various cancers. MTA3 is also a key regulator of E-cadherin expression and epithelial-to-mesenchymal transition. Elucidating the functions of MTA3 might help to find additional therapeutic approaches for targeting components of NuRD.
Collapse
Affiliation(s)
- Liang Du
- Cancer Research Center, Shantou University Medical College, Shantou, 515031 Guangdong P. R. China
| | - Zhifeng Ning
- Basic Medicine College, Hubei University of Science and Technology, Xianning, 437100 Hubei P. R. China
| | - Hao Zhang
- Cancer Research Center, Shantou University Medical College, Shantou, 515031 Guangdong P. R. China
- Department of Biotherapy, Affiliated Cancer Hospital of Shantou University Medical College, Shantou, 515031 Guangdong P. R. China
| | - Fuxing Liu
- Basic Medicine College, Hubei University of Science and Technology, Xianning, 437100 Hubei P. R. China
| |
Collapse
|
28
|
Vav1: A Dr. Jekyll and Mr. Hyde protein--good for the hematopoietic system, bad for cancer. Oncotarget 2016; 6:28731-42. [PMID: 26353933 PMCID: PMC4745688 DOI: 10.18632/oncotarget.5086] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 08/07/2015] [Indexed: 01/10/2023] Open
Abstract
Many deregulated signal transducer proteins are involved in various cancers at numerous stages of tumor development. One of these, Vav1, is normally expressed exclusively in the hematopoietic system, where it functions as a specific GDP/GTP nucleotide exchange factor (GEF), strictly regulated by tyrosine phosphorylation. Vav was first identified in an NIH3T3 screen for oncogenes. Although the oncogenic form of Vav1 identified in the screen has not been detected in clinical human tumors, its wild-type form has recently been implicated in mammalian malignancies, including neuroblastoma, melanoma, pancreatic, lung and breast cancers, and B-cell chronic lymphocytic leukemia. In addition, it was recently identified as a mutated gene in human cancers of various origins. However, the activity and contribution to cancer of these Vav1 mutants is still unclear. This review addresses the physiological function of wild-type Vav1 and its activity as an oncogene in human cancer. It also discusses the novel mutations identified in Vav1 in various cancers and their potential contribution to cancer development as oncogenes or tumor suppressor genes.
Collapse
|
29
|
Castro-Castro A, Marchesin V, Monteiro P, Lodillinsky C, Rossé C, Chavrier P. Cellular and Molecular Mechanisms of MT1-MMP-Dependent Cancer Cell Invasion. Annu Rev Cell Dev Biol 2016; 32:555-576. [PMID: 27501444 DOI: 10.1146/annurev-cellbio-111315-125227] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metastasis is responsible for most cancer-associated deaths. Accumulating evidence based on 3D migration models has revealed a diversity of invasive migratory schemes reflecting the plasticity of tumor cells to switch between proteolytic and nonproteolytic modes of invasion. Yet, initial stages of localized regional tumor dissemination require proteolytic remodeling of the extracellular matrix to overcome tissue barriers. Recent data indicate that surface-exposed membrane type 1-matrix metalloproteinase (MT1-MMP), belonging to a group of membrane-anchored MMPs, plays a central role in pericellular matrix degradation during basement membrane and interstitial tissue transmigration programs. In addition, a large body of work indicates that MT1-MMP is targeted to specialized actin-rich cell protrusions termed invadopodia, which are responsible for matrix degradation. This review describes the multistep assembly of actin-based invadopodia in molecular details. Mechanisms underlying MT1-MMP traffic to invadopodia through endocytosis/recycling cycles, which are key to the invasive program of carcinoma cells, are discussed.
Collapse
Affiliation(s)
| | | | - Pedro Monteiro
- Barts Cancer Institute, University of London John Vane Science Centre, London EC1M 6BQ, United Kingdom
| | - Catalina Lodillinsky
- Instituto de Oncologia Ángel H. Roffo, Research Area, Buenos Aires, C1417DTB, Argentina
| | - Carine Rossé
- Institut Curie, Paris, F-75248 France; .,PSL Research University, Paris, F-75005 France.,CNRS, UMR 144, Paris, F-75248 France
| | - Philippe Chavrier
- Institut Curie, Paris, F-75248 France; .,PSL Research University, Paris, F-75005 France.,CNRS, UMR 144, Paris, F-75248 France
| |
Collapse
|
30
|
Dhar Dwivedi SK, Mustafi SB, Mangala LS, Jiang D, Pradeep S, Rodriguez-Aguayo C, Ling H, Ivan C, Mukherjee P, Calin GA, Lopez-Berestein G, Sood AK, Bhattacharya R. Therapeutic evaluation of microRNA-15a and microRNA-16 in ovarian cancer. Oncotarget 2016; 7:15093-104. [PMID: 26918603 PMCID: PMC4924772 DOI: 10.18632/oncotarget.7618] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/23/2016] [Indexed: 12/23/2022] Open
Abstract
Treatment of chemo-resistant ovarian cancer (OvCa) remains clinically challenging and there is a pressing need to identify novel therapeutic strategies. Here we report that multiple mechanisms that promote OvCa progression and chemo-resistance could be inhibited by ectopic expression of miR-15a and miR-16. Significant correlations between low expression of miR-16, high expression of BMI1 and shortened overall survival (OS) were noted in high grade serous (HGS) OvCa patients upon analysis of The Cancer Genome Atlas (TCGA). Targeting BMI1, in vitro with either microRNA reduced clonal growth of OvCa cells. Additionally, epithelial to mesenchymal transition (EMT) as well as expression of the cisplatin transporter ATP7B were inhibited by miR-15a and miR-16 resulting in decreased degradation of the extra-cellular matrix and enhanced sensitization of OvCa cells to cisplatin. Nanoliposomal delivery of the miR-15a and miR-16 combination, in a pre-clinical chemo-resistant orthotopic mouse model of OvCa, demonstrated striking reduction in tumor burden compared to cisplatin alone. Thus, with the advent of miR replacement therapy some of which are in Phase 2 clinical trials, miR-15a and miR-16 represent novel ammunition in the anti-OvCa arsenal.
Collapse
Affiliation(s)
- Shailendra Kumar Dhar Dwivedi
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Soumyajit Banerjee Mustafi
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Lingegowda S. Mangala
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dahai Jiang
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunila Pradeep
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristian Rodriguez-Aguayo
- The Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Ling
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristina Ivan
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyabrata Mukherjee
- Department of Pathology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - George A. Calin
- The Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Lopez-Berestein
- The Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma College of Medicine, Oklahoma City, OK, USA
| |
Collapse
|
31
|
Lohmer LL, Clay MR, Naegeli KM, Chi Q, Ziel JW, Hagedorn EJ, Park JE, Jayadev R, Sherwood DR. A Sensitized Screen for Genes Promoting Invadopodia Function In Vivo: CDC-42 and Rab GDI-1 Direct Distinct Aspects of Invadopodia Formation. PLoS Genet 2016; 12:e1005786. [PMID: 26765257 PMCID: PMC4713207 DOI: 10.1371/journal.pgen.1005786] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/12/2015] [Indexed: 12/12/2022] Open
Abstract
Invadopodia are specialized membrane protrusions composed of F-actin, actin regulators, signaling proteins, and a dynamically trafficked invadopodial membrane that drive cell invasion through basement membrane (BM) barriers in development and cancer. Due to the challenges of studying invasion in vivo, mechanisms controlling invadopodia formation in their native environments remain poorly understood. We performed a sensitized genome-wide RNAi screen and identified 13 potential regulators of invadopodia during anchor cell (AC) invasion into the vulval epithelium in C. elegans. Confirming the specificity of this screen, we identified the Rho GTPase cdc-42, which mediates invadopodia formation in many cancer cell lines. Using live-cell imaging, we show that CDC-42 localizes to the AC-BM interface and is activated by an unidentified vulval signal(s) that induces invasion. CDC-42 is required for the invasive membrane localization of WSP-1 (N-WASP), a CDC-42 effector that promotes polymerization of F-actin. Loss of CDC-42 or WSP-1 resulted in fewer invadopodia and delayed BM breaching. We also characterized a novel invadopodia regulator, gdi-1 (Rab GDP dissociation inhibitor), which mediates membrane trafficking. We show that GDI-1 functions in the AC to promote invadopodia formation. In the absence of GDI-1, the specialized invadopodial membrane was no longer trafficked normally to the invasive membrane, and instead was distributed to plasma membrane throughout the cell. Surprisingly, the pro-invasive signal(s) from the vulval cells also controls GDI-1 activity and invadopodial membrane trafficking. These studies represent the first in vivo screen for genes regulating invadopodia and demonstrate that invadopodia formation requires the integration of distinct cellular processes that are coordinated by an extracellular cue. During animal development specialized cells acquire the ability move and invade into other tissues to form complex organs and structures. Understanding this cellular behavior is important medically, as cancer cells can hijack the developmental program of invasion to metastasize throughout the body. One of the most formidable barriers invasive cells face is basement membrane–-a thin, dense, sheet-like assembly of proteins and carbohydrates that surrounds most tissues. Cells deploy small, protrusive, membrane associated structures called invadopodia (invasive feet) to breach basement membranes. How invadopodia are formed and controlled during invasion has been challenging to understand, as it is difficult to examine these dynamic structures in live animals. Using the nematode worm Caenorhabditis elegans, we have conducted the first large-scale screen to isolate genes that control invadopodia in live animals. Our screen isolated 13 genes and we confirmed two are key invadopodia regulators: the Rho GTPase CDC-42 that promotes F-actin polymerization at invadopodia to generate the force to breach basement membranes, and the Rab GDI-1 that promotes membrane addition at invadopodia that may allow invadopodia to extend through basement membranes. This work provides new insights into invadopodia construction and identifies potential novel targets for anti-metastasis therapies.
Collapse
Affiliation(s)
- Lauren L. Lohmer
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Matthew R. Clay
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Kaleb M. Naegeli
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Qiuyi Chi
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Joshua W. Ziel
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Elliott J. Hagedorn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jieun E. Park
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Ranjay Jayadev
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - David R. Sherwood
- Department of Biology, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
32
|
Baruzzi A, Remelli S, Lorenzetto E, Sega M, Chignola R, Berton G. Sos1 Regulates Macrophage Podosome Assembly and Macrophage Invasive Capacity. THE JOURNAL OF IMMUNOLOGY 2015; 195:4900-12. [PMID: 26447228 DOI: 10.4049/jimmunol.1500579] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/03/2015] [Indexed: 12/29/2022]
Abstract
Podosomes are protrusive structures implicated in macrophage extracellular matrix degradation and three-dimensional migration through cell barriers and the interstitium. Podosome formation and assembly are regulated by cytoskeleton remodeling requiring cytoplasmic tyrosine kinases of the Src and the Abl families. Considering that Abl has been reported to phosphorylate the guanine nucleotide exchange factor Sos1, eliciting its Rac-guanine nucleotide exchange factor activity, and Rac regulates podosome formation in myeloid cells and invadopodia formation in cancer cells, we addressed whether Sos1 is implicated in podosome formation and function in macrophages. We found that ectopically expressed Abl or the Src kinase Fgr phosphorylate Sos1, and the Src kinases Hck and Fgr are required for Abl and Sos1 phosphorylation and Abl/Sos1 interaction in macrophages. Sos1 localizes to podosomes in both murine and human macrophages, and its silencing by small interfering RNA results in disassembly of murine macrophage podosomes and a marked reduction of GTP loading on Rac. Matrix degradative capacity, three-dimensional migration through Matrigel, and transmigration through an endothelial cell monolayer of Sos1-silenced macrophages were inhibited. In addition, Sos1- or Abl-silenced macrophages, or macrophages treated with the selective Abl inhibitor imatinib mesylate had a reduced capability to migrate into breast tumor spheroids, the majority of cells remaining at the margin and the outer layers of the spheroid itself. Because of the established role of Src and Abl kinases to regulate also invadopodia formation in cancer cells, our findings suggest that targeting the Src/Abl/Sos1/Rac pathway may represent a double-edged sword to control both cancer-invasive capacities and cancer-related inflammation.
Collapse
Affiliation(s)
- Anna Baruzzi
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, 37134 Verona, Italy
| | - Sabrina Remelli
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, 37134 Verona, Italy
| | - Erika Lorenzetto
- Department of Neurological and Movement Sciences, University of Verona, 37134 Verona, Italy; and
| | - Michela Sega
- Department of Biotechnology, University of Verona, 37134 Verona, Italy
| | - Roberto Chignola
- Department of Biotechnology, University of Verona, 37134 Verona, Italy
| | - Giorgio Berton
- Department of Pathology and Diagnostics, Section of General Pathology, University of Verona, 37134 Verona, Italy;
| |
Collapse
|
33
|
Mulens-Arias V, Rojas JM, Pérez-Yagüe S, Morales MDP, Barber DF. Polyethylenimine-coated SPION exhibits potential intrinsic anti-metastatic properties inhibiting migration and invasion of pancreatic tumor cells. J Control Release 2015; 216:78-92. [DOI: 10.1016/j.jconrel.2015.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/28/2015] [Accepted: 08/04/2015] [Indexed: 12/12/2022]
|
34
|
Brown GT, Murray GI. Current mechanistic insights into the roles of matrix metalloproteinases in tumour invasion and metastasis. J Pathol 2015; 237:273-81. [PMID: 26174849 DOI: 10.1002/path.4586] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/03/2015] [Accepted: 07/08/2015] [Indexed: 12/12/2022]
Abstract
The purpose of this review is to highlight the recent mechanistic developments elucidating the role of matrix metalloproteinases (MMPs) in tumour invasion and metastasis. The ability of tumour cells to invade, migrate, and subsequently metastasize is a fundamental characteristic of cancer. Tumour invasion and metastasis are increasingly being characterized by the dynamic relationship between cancer cells and their microenvironment and developing a greater understanding of these basic pathological mechanisms is crucial. While MMPs have been strongly implicated in these processes as a result of extensive circumstantial evidence--for example, increased expression of individual MMPs in tumours and association of specific MMPs with prognosis--the underpinning mechanisms are only now being elucidated. Recent studies are now providing a mechanistic basis, highlighting and reinforcing the catalytic and non-catalytic roles of specific MMPs as key players in tumour invasion and metastasis.
Collapse
Affiliation(s)
- Gordon T Brown
- Pathology, Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Graeme I Murray
- Pathology, Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
35
|
Sebban S, Farago M, Rabinovich S, Lazer G, Idelchuck Y, Ilan L, Pikarsky E, Katzav S. Vav1 promotes lung cancer growth by instigating tumor-microenvironment cross-talk via growth factor secretion. Oncotarget 2015; 5:9214-26. [PMID: 25313137 PMCID: PMC4253429 DOI: 10.18632/oncotarget.2400] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vav1 is a signal transducer that functions as a scaffold protein and a regulator of cytoskeleton organization in the hematopoietic system, where it is exclusively expressed. Recently, Vav1 was shown to be involved in diverse human cancers, including lung cancer. We demonstrate that lung cancer cells that abnormally express Vav1 secrete growth factors in a Vav1-dependent manner. Transcriptome analysis demonstrated that Vav1 depletion results in a marked reduction in the expression of colony-stimulating-factor-1 (CSF1), a hematopoietic growth factor. The association between Vav1 expression and CSF1 was further supported by signal transduction experiments, supporting involvement of Vav1 in regulating lung cancer secretome. Blocking of ERK phosphorylation, led to a decrease in CSF1 transcription, thus suggesting a role for ERK, a downstream effector of Vav1, in CSF1 expression. CSF1-silenced cells exhibited reduced focus formation, proliferation abilities, and growth in NOD/SCID mice. CSF1-silenced H358 cells resulted in significantly smaller tumors, showing increased fibrosis and a decrease in tumor infiltrating macrophages. Finally, immunohistochemical analysis of primary human lung tumors revealed a positive correlation between Vav1 and CSF1 expression, which was associated with tumor grade. Additional results presented herein suggest a potential cross-talk between cancer cells and the microenvironment controlled by CSF1/Vav1 signaling pathways.
Collapse
Affiliation(s)
- Shulamit Sebban
- Departement of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School - Hebrew University, Jerusalem, Israel
| | - Marganit Farago
- Departement of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School - Hebrew University, Jerusalem, Israel
| | - Shiran Rabinovich
- Departement of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School - Hebrew University, Jerusalem, Israel
| | - Galit Lazer
- Departement of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School - Hebrew University, Jerusalem, Israel
| | - Yulia Idelchuck
- Departement of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School - Hebrew University, Jerusalem, Israel
| | - Lena Ilan
- Departement of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School - Hebrew University, Jerusalem, Israel
| | - Eli Pikarsky
- Department of Immunology and Cancer Research and Department of Pathology, Institute for Medical Research Israel-Canada, Hadassah Medical School - Hebrew University, Jerusalem, Israel
| | - Shulamit Katzav
- Departement of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hadassah Medical School - Hebrew University, Jerusalem, Israel
| |
Collapse
|
36
|
Abstract
The Vav family is a group of tyrosine phosphorylation-regulated signal transduction molecules hierarchically located downstream of protein tyrosine kinases. The main function of these proteins is to work as guanosine nucleotide exchange factors (GEFs) for members of the Rho GTPase family. In addition, they can exhibit a variety of catalysis-independent roles in specific signaling contexts. Vav proteins play essential signaling roles for both the development and/or effector functions of a large variety of cell lineages, including those belonging to the immune, nervous, and cardiovascular systems. They also contribute to pathological states such as cancer, immune-related dysfunctions, and atherosclerosis. Here, I will provide an integrated view about the evolution, regulation, and effector properties of these signaling molecules. In addition, I will discuss the pros and cons for their potential consideration as therapeutic targets.
Collapse
Key Words
- Ac, acidic
- Ahr, aryl hydrocarbon receptor
- CH, calponin homology
- CSH3, most C-terminal SH3 domain of Vav proteins
- DAG, diacylglycerol
- DH, Dbl-homology domain
- Dbl-homology
- GDP/GTP exchange factors
- GEF, guanosine nucleotide exchange factor
- HIV, human immunodeficiency virus
- IP3, inositoltriphosphate
- NFAT, nuclear factor of activated T-cells
- NSH3, most N-terminal SH3 domain of Vav proteins
- PH, plekstrin-homology domain
- PI3K, phosphatidylinositol-3 kinase
- PIP3, phosphatidylinositol (3,4,5)-triphosphate
- PKC, protein kinase C
- PKD, protein kinase D
- PLC-g, phospholipase C-g
- PRR, proline-rich region
- PTK, protein tyrosine kinase
- Phox, phagocyte oxidase
- Rho GTPases
- SH2, Src homology 2
- SH3, Src homology 3
- SNP, single nucleotide polymorphism
- TCR, T-cell receptor
- Vav
- ZF, zinc finger region
- cGMP, cyclic guanosine monophosphate
- cancer
- cardiovascular biology
- disease
- immunology
- nervous system
- signaling
- therapies
Collapse
Affiliation(s)
- Xosé R Bustelo
- a Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer ; Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca ; Campus Unamuno; Salamanca , Spain
| |
Collapse
|
37
|
Razidlo GL, Magnine C, Sletten AC, Hurley RM, Almada LL, Fernandez-Zapico ME, Ji B, McNiven MA. Targeting Pancreatic Cancer Metastasis by Inhibition of Vav1, a Driver of Tumor Cell Invasion. Cancer Res 2015; 75:2907-15. [PMID: 25977335 PMCID: PMC4506209 DOI: 10.1158/0008-5472.can-14-3103] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 04/19/2015] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer, one of the most lethal forms of human cancer, is largely resistant to many conventional chemotherapeutic agents. Although many therapeutic approaches focus on tumor growth, metastasis is a primary factor contributing to lethality. Therefore, novel therapies to target metastatic invasion could prevent tumor spread and recurrence resulting from local and distant metastasis. The protein Vav1 is aberrantly expressed in more than half of pancreatic cancers. Its expression promotes activation of Rac and Cdc42 and leads to enhanced invasion and migration, as well as increased tumor cell survival and proliferation, suggesting that Vav1 could be a potent therapeutic target for pancreatic cancer. The purine analogue azathioprine, well known for its function as an anti-inflammatory compound, was recently shown to function by inhibiting Vav1 signaling in immune cells. We therefore hypothesized that azathioprine could also inhibit Vav1 in pancreatic tumor cells to reduce its proinvasive functions. Indeed, we have found that treatment of cultured pancreatic tumor cells with azathioprine inhibited Vav1-dependent invasive cell migration and matrix degradation, through inhibition of Rac and Cdc42 signaling. Furthermore, azathioprine treatment decreased metastasis in both xenograft and genetic mouse models of pancreatic cancer. Strikingly, metastasis was dramatically reduced in Vav1-expressing tumors arising from p48(Cre/+), Kras(G12D/+), p53(F/+) mice. These inhibitory effects were mediated through Vav1, as Vav1-negative cell lines and tumors were largely resistant to azathioprine treatment. These findings demonstrate that azathioprine and related compounds could be potent antimetastatic agents for Vav1-positive pancreatic tumors.
Collapse
Affiliation(s)
- Gina L Razidlo
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota. Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Christopher Magnine
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Arthur C Sletten
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota
| | | | - Baoan Ji
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Mark A McNiven
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota. Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
38
|
Lohmer LL, Kelley LC, Hagedorn EJ, Sherwood DR. Invadopodia and basement membrane invasion in vivo. Cell Adh Migr 2015; 8:246-55. [PMID: 24717190 DOI: 10.4161/cam.28406] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Over 20 years ago, protrusive, F-actin-based membrane structures, termed invadopodia, were identified in highly metastatic cancer cell lines. Invadopodia penetrate artificial or explanted extracellular matrices in 2D culture conditions and have been hypothesized to facilitate the migration of cancer cells through basement membrane, a thin, dense, barrier-like matrix surrounding most tissues. Despite intensive study, the identification of invadopodia in vivo has remained elusive and until now their possible roles during invasion or even existence have remained unclear. Studies in remarkably different cellular contexts-mouse tumor models, zebrafish intestinal epithelia, and C. elegans organogenesis-have recently identified invadopodia structures associated with basement membrane invasion. These studies are providing the first in vivo insight into the regulation, function, and role of these fascinating subcellular devices with critical importance to both development and human disease.
Collapse
|
39
|
Di Martino J, Paysan L, Gest C, Lagrée V, Juin A, Saltel F, Moreau V. Cdc42 and Tks5: a minimal and universal molecular signature for functional invadosomes. Cell Adh Migr 2015; 8:280-92. [PMID: 24840388 DOI: 10.4161/cam.28833] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Invadosomes are actin-based structures involved in extracellular-matrix degradation. Invadosomes, either known as podosomes or invadopodia, are found in an increasing number of cell types. Moreover, their overall organization and molecular composition may vary from one cell type to the other. Some are constitutive such as podosomes in hematopoietic cells whereas others are inducible. However, they share the same feature, their ability to interact and to degrade the extracellular matrix. Based on the literature and our own experiments, the aim of this study was to establish a minimal molecular definition of active invadosomes. We first highlighted that Cdc42 is the key RhoGTPase involved in invadosome formation in all described models. Using different cellular models, such as NIH-3T3, HeLa, and endothelial cells, we demonstrated that overexpression of an active form of Cdc42 is sufficient to form invadosome actin cores. Therefore, active Cdc42 must be considered not only as an inducer of filopodia, but also as an inducer of invadosomes. Depending on the expression level of Tks5, these Cdc42-dependent actin cores were endowed or not with a proteolytic activity. In fact, Tks5 overexpression rescued this activity in Tks5 low expressing cells. We thus described the adaptor protein Tks5 as a major actor of the invadosome degradation function. Surprisingly, we found that Src kinases are not always required for invadosome formation and function. These data suggest that even if Src family members are the principal kinases involved in the majority of invadosomes, it cannot be considered as a common element for all invadosome structures. We thus define a minimal and universal molecular signature of invadosome that includes Cdc42 activity and Tks5 presence in order to drive the actin machinery and the proteolytic activity of these invasive structures.
Collapse
Affiliation(s)
- Julie Di Martino
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Lisa Paysan
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Caroline Gest
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Valérie Lagrée
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Amélie Juin
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Frédéric Saltel
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Violaine Moreau
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| |
Collapse
|
40
|
Qi Y, Kong FM, Deng Q, Li JY, Cui R, Pu YD, Zhai QL, Jia YJ, Li YM. Clinical significance and prognostic value of Vav1 expression in Non-small cell lung cancer. Am J Cancer Res 2015; 5:2491-2497. [PMID: 26396925 PMCID: PMC4568785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 07/07/2015] [Indexed: 06/05/2023] Open
Abstract
Vav1 has been reported to be involved in human cancers, however, the expression and clinical significance of Vav1 in NSCLC are not fully understood. In the present study, we examined the expression of Vav1 in 170 NSCLC patients who underwent radical resection by the immunohistochemical analyses. The association between the Vav1 expression and clinicopathological variables was analyzed. The multivariate Cox proportional hazards model was conducted to determine the prognostic value of Vav1 on the long-term survival. The results showed that the elevated Vav1 expression was correlated positively with lymph node metastasis (P<0.001), T stage (P<0.001) and poor histological differentiation (P<0.001). Patients with negative or low Vav1 expression had better prognoses than those with high Vav1 expression (P<0.001). Multivariate analysis indicated that Vav1 was independent prognostic factor for overall survival (OS) (HR 2.079, 95% CI 1.564 to 2.762, P<0.001) and disease-free survival (DFS) (HR 1.810, 95% CI 1.391 to 2.356, P<0.001). Our findings showed that overexpressed Vav1 was correlated with aggressive tumor behavior. Val1 was an independent factor for NSCLC prognosis, which may serve as a novel prognostic factor and potential target to improve the long-term outcome of NSCLC.
Collapse
Affiliation(s)
- Yao Qi
- Department of Hematology, Tianjin First Center HospitalFukang Road, Nankai District, Tianjin 300192, China
| | - Fan-Ming Kong
- Department of Oncology, First Teaching Hospital of Tianjin University of TCMAnshanxi Road, Nankai District, Tianjin 300193, China
| | - Qi Deng
- Department of Hematology, Tianjin First Center HospitalFukang Road, Nankai District, Tianjin 300192, China
| | - Jing-Yi Li
- Department of Hematology, Tianjin First Center HospitalFukang Road, Nankai District, Tianjin 300192, China
| | - Rui Cui
- Department of Hematology, Tianjin First Center HospitalFukang Road, Nankai District, Tianjin 300192, China
| | - Ye-Di Pu
- Department of Hematology, Tianjin First Center HospitalFukang Road, Nankai District, Tianjin 300192, China
| | - Qiong-Li Zhai
- Department of Pathology, Tianjin Medical University Cancer Institute and HospitalTiyuanbei Road, Hexi District, Tianjin 300060, China
| | - Ying-Jie Jia
- Department of Oncology, First Teaching Hospital of Tianjin University of TCMAnshanxi Road, Nankai District, Tianjin 300193, China
| | - Yu-Ming Li
- Department of Hematology, Tianjin First Center HospitalFukang Road, Nankai District, Tianjin 300192, China
| |
Collapse
|
41
|
Cao H, Eppinga RD, Razidlo GL, Krueger EW, Chen J, Qiang L, McNiven MA. Stromal fibroblasts facilitate cancer cell invasion by a novel invadopodia-independent matrix degradation process. Oncogene 2015; 35:1099-1110. [PMID: 25982272 PMCID: PMC4651864 DOI: 10.1038/onc.2015.163] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 02/25/2015] [Accepted: 03/20/2015] [Indexed: 12/13/2022]
Abstract
Metastatic invasion of tumors into peripheral tissues is known to rely upon protease-mediated degradation of the surrounding stroma. This remodeling process uses complex, actin-based, specializations of the plasma membrane termed invadopodia that act both to sequester and release matrix metalloproteinases. Here we report that cells of mesenchymal origin, including tumor-associated fibroblasts, degrade substantial amounts of surrounding matrix by a mechanism independent of conventional invadopodia. These degradative sites lack the punctate shape of conventional invadopodia to spread along the cell base and are reticular and/or fibrous in character. In marked contrast to invadopodia, this degradation does not require the action of Src kinase, Cdc42 or Dyn2. Rather, inhibition of Dyn2 causes a marked upregulation of stromal matrix degradation. Further, expression and activity of matrix metalloproteinases are differentially regulated between tumor cells and stromal fibroblasts. This matrix remodeling by fibroblasts increases the invasive capacity of tumor cells, thereby illustrating how the tumor microenvironment can contribute to metastasis. These findings provide evidence for a novel matrix remodeling process conducted by stromal fibroblasts that is substantially more effective than conventional invadopodia, distinct in structural organization and regulated by disparate molecular mechanisms.
Collapse
Affiliation(s)
- Hong Cao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota USA
| | - Robbin D Eppinga
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota USA
| | - Gina L Razidlo
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota USA
| | - Eugene W Krueger
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota USA
| | - Jing Chen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota USA
| | - Li Qiang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota USA
| | - Mark A McNiven
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota USA
| |
Collapse
|
42
|
Krakhmal NV, Zavyalova MV, Denisov EV, Vtorushin SV, Perelmuter VM. Cancer Invasion: Patterns and Mechanisms. Acta Naturae 2015; 7:17-28. [PMID: 26085941 PMCID: PMC4463409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Cancer invasion and the ability of malignant tumor cells for directed migration and metastasis have remained a focus of research for many years. Numerous studies have confirmed the existence of two main patterns of cancer cell invasion: collective cell migration and individual cell migration, by which tumor cells overcome barriers of the extracellular matrix and spread into surrounding tissues. Each pattern of cell migration displays specific morphological features and the biochemical/molecular genetic mechanisms underlying cell migration. Two types of migrating tumor cells, mesenchymal (fibroblast-like) and amoeboid, are observed in each pattern of cancer cell invasion. This review describes the key differences between the variants of cancer cell migration, the role of epithelial-mesenchymal, collective-amoeboid, mesenchymal-amoeboid, and amoeboid- mesenchymal transitions, as well as the significance of different tumor factors and stromal molecules in tumor invasion. The data and facts collected are essential to the understanding of how the patterns of cancer cell invasion are related to cancer progression and therapy efficacy. Convincing evidence is provided that morphological manifestations of the invasion patterns are characterized by a variety of tissue (tumor) structures. The results of our own studies are presented to show the association of breast cancer progression with intratumoral morphological heterogeneity, which most likely reflects the types of cancer cell migration and results from different activities of cell adhesion molecules in tumor cells of distinct morphological structures.
Collapse
Affiliation(s)
- N. V. Krakhmal
- Siberian State Medical University, Moskovskiy Trakt, 2, 634050, Tomsk, Russia
| | - M. V. Zavyalova
- Siberian State Medical University, Moskovskiy Trakt, 2, 634050, Tomsk, Russia
- Tomsk Cancer Research Institute, Kooperativny Pereulok, 5, 634050, Tomsk, Russia
- Tomsk State University, Prosp. Lenina, 36, 634050, Tomsk, Russia
| | - E. V. Denisov
- Tomsk Cancer Research Institute, Kooperativny Pereulok, 5, 634050, Tomsk, Russia
- Tomsk State University, Prosp. Lenina, 36, 634050, Tomsk, Russia
| | - S. V. Vtorushin
- Siberian State Medical University, Moskovskiy Trakt, 2, 634050, Tomsk, Russia
- Tomsk Cancer Research Institute, Kooperativny Pereulok, 5, 634050, Tomsk, Russia
| | - V. M. Perelmuter
- Siberian State Medical University, Moskovskiy Trakt, 2, 634050, Tomsk, Russia
- Tomsk Cancer Research Institute, Kooperativny Pereulok, 5, 634050, Tomsk, Russia
| |
Collapse
|
43
|
Artym VV, Swatkoski S, Matsumoto K, Campbell CB, Petrie RJ, Dimitriadis EK, Li X, Mueller SC, Bugge TH, Gucek M, Yamada KM. Dense fibrillar collagen is a potent inducer of invadopodia via a specific signaling network. ACTA ACUST UNITED AC 2015; 208:331-50. [PMID: 25646088 PMCID: PMC4315243 DOI: 10.1083/jcb.201405099] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
High-density fibrillar collagen matrix induces invadopodia formation in both fibroblasts and carcinoma cell lines through a kindlin2-dependent mechanism that drives local ECM remodeling. Cell interactions with the extracellular matrix (ECM) can regulate multiple cellular activities and the matrix itself in dynamic, bidirectional processes. One such process is local proteolytic modification of the ECM. Invadopodia of tumor cells are actin-rich proteolytic protrusions that locally degrade matrix molecules and mediate invasion. We report that a novel high-density fibrillar collagen (HDFC) matrix is a potent inducer of invadopodia, both in carcinoma cell lines and in primary human fibroblasts. In carcinoma cells, HDFC matrix induced formation of invadopodia via a specific integrin signaling pathway that did not require growth factors or even altered gene and protein expression. In contrast, phosphoproteomics identified major changes in a complex phosphosignaling network with kindlin2 serine phosphorylation as a key regulatory element. This kindlin2-dependent signal transduction network was required for efficient induction of invadopodia on dense fibrillar collagen and for local degradation of collagen. This novel phosphosignaling mechanism regulates cell surface invadopodia via kindlin2 for local proteolytic remodeling of the ECM.
Collapse
Affiliation(s)
- Vira V Artym
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892 Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School; and Department of Biostatistics, Bioinformatics, and Biomathematics; Georgetown University, Washington, DC 20057
| | - Stephen Swatkoski
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Kazue Matsumoto
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Catherine B Campbell
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Ryan J Petrie
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Emilios K Dimitriadis
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Xin Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School; and Department of Biostatistics, Bioinformatics, and Biomathematics; Georgetown University, Washington, DC 20057
| | - Susette C Mueller
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School; and Department of Biostatistics, Bioinformatics, and Biomathematics; Georgetown University, Washington, DC 20057
| | - Thomas H Bugge
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Marjan Gucek
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Kenneth M Yamada
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
44
|
Juin A, Di Martino J, Leitinger B, Henriet E, Gary AS, Paysan L, Bomo J, Baffet G, Gauthier-Rouvière C, Rosenbaum J, Moreau V, Saltel F. Discoidin domain receptor 1 controls linear invadosome formation via a Cdc42-Tuba pathway. ACTA ACUST UNITED AC 2015; 207:517-33. [PMID: 25422375 PMCID: PMC4242841 DOI: 10.1083/jcb.201404079] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In tumor cells, the collagen receptor DDR1 collaborates with Cdc42 and its guanine exchange factor Tuba to promote linear invadosome formation and increase their matrix-invading activity. Accumulation of type I collagen fibrils in tumors is associated with an increased risk of metastasis. Invadosomes are F-actin structures able to degrade the extracellular matrix. We previously found that collagen I fibrils induced the formation of peculiar linear invadosomes in an unexpected integrin-independent manner. Here, we show that Discoidin Domain Receptor 1 (DDR1), a collagen receptor overexpressed in cancer, colocalizes with linear invadosomes in tumor cells and is required for their formation and matrix degradation ability. Unexpectedly, DDR1 kinase activity is not required for invadosome formation or activity, nor is Src tyrosine kinase. We show that the RhoGTPase Cdc42 is activated on collagen in a DDR1-dependent manner. Cdc42 and its specific guanine nucleotide-exchange factor (GEF), Tuba, localize to linear invadosomes, and both are required for linear invadosome formation. Finally, DDR1 depletion blocked cell invasion in a collagen gel. Altogether, our data uncover an important role for DDR1, acting through Tuba and Cdc42, in proteolysis-based cell invasion in a collagen-rich environment.
Collapse
Affiliation(s)
- Amélie Juin
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Julie Di Martino
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Birgit Leitinger
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, England, UK
| | - Elodie Henriet
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Anne-Sophie Gary
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Lisa Paysan
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Jeremy Bomo
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche sur la Santé l'Environnement et le Travail (IRSET), Université de Rennes 1, 35043 Rennes, France
| | - Georges Baffet
- Institut National de la Santé et de la Recherche Médicale, U1085, Institut de Recherche sur la Santé l'Environnement et le Travail (IRSET), Université de Rennes 1, 35043 Rennes, France
| | - Cécile Gauthier-Rouvière
- Universités Montpellier 2 et 1, Centre de Recherche de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique, UMR 5237, 34293 Montpellier, France
| | - Jean Rosenbaum
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Violaine Moreau
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| | - Frédéric Saltel
- Institut National de la Santé et de la Recherche Médicale, U1053, F-33076 Bordeaux, France Université de Bordeaux, F-33076 Bordeaux, France
| |
Collapse
|
45
|
Beaty BT, Condeelis J. Digging a little deeper: the stages of invadopodium formation and maturation. Eur J Cell Biol 2014; 93:438-44. [PMID: 25113547 DOI: 10.1016/j.ejcb.2014.07.003] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/10/2014] [Accepted: 07/10/2014] [Indexed: 01/09/2023] Open
Abstract
Invadopodia are actin-rich protrusions that degrade the extracellular matrix and are required for penetration through the basement membrane, stromal invasion and intravasation. Invadopodia are enriched in actin regulators, such as cortactin, cofilin, N-WASp, Arp2/3 and fascin. Much of the work to date has centered around identifying the proteins involved in regulating actin polymerization and matrix degradation. Recently, there have been significant advances in characterization of the very early stages of invadopodium precursor assembly and the role of adhesion proteins, such as β1 integrin, talin, FAK and Hic-5, in promoting invadopodium maturation. This review summarizes these findings in the context of our current model of invadopodial function and highlights some of the important unanswered questions in the field.
Collapse
Affiliation(s)
- Brian T Beaty
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| | - John Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, United States; Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| |
Collapse
|
46
|
Abstract
Intratumoral hypoxia is a common feature of solid tumors. Recent advances in cancer biology indicate that hypoxia is not only a consequence of unrestrained tumor growth, but also plays an active role in promoting tumor progression, malignancy, and resistance to therapy. Hypoxia signaling is mediated by the hypoxia-inducible factors (HIFs), which are not only stabilized under hypoxia, but also by activated oncogenes or inactivated tumor suppressors under normoxia. Hypoxia is a prominent feature of the tumor microenvironment of pancreatic tumors, also characterized by the presence of a fibrotic reaction that promotes, and is also modulated by, hypoxia. As the mechanisms by which hypoxia signaling impacts invasion and metastasis in pancreatic cancer are being elucidated, hypoxia is emerging as a key determinant of pancreatic cancer malignancy as well as an important target for therapy. Herein we present an overview of recent advances in the understanding of the impact that hypoxia has in pancreatic cancer invasion and metastasis.
Collapse
Affiliation(s)
- Angela Yuen
- Tumor Microenvironment and Metastasis Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Begoña Díaz
- Tumor Microenvironment and Metastasis Program, Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| |
Collapse
|