1
|
Semeradtova A, Liegertova M, Herma R, Capkova M, Brignole C, Del Zotto G. Extracellular vesicles in cancer´s communication: messages we can read and how to answer. Mol Cancer 2025; 24:86. [PMID: 40108630 PMCID: PMC11921637 DOI: 10.1186/s12943-025-02282-1] [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: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Extracellular vesicles (EVs) are emerging as critical mediators of intercellular communication in the tumor microenvironment (TME), profoundly influencing cancer progression. These nano-sized vesicles, released by both tumor and stromal cells, carry a diverse cargo of proteins, nucleic acids, and lipids, reflecting the dynamic cellular landscape and mediating intricate interactions between cells. This review provides a comprehensive overview of the biogenesis, composition, and functional roles of EVs in cancer, highlighting their significance in both basic research and clinical applications. We discuss how cancer cells manipulate EV biogenesis pathways to produce vesicles enriched with pro-tumorigenic molecules, explore the specific contributions of EVs to key hallmarks of cancer, such as angiogenesis, metastasis, and immune evasion, emphasizing their role in shaping TME and driving therapeutic resistance. Concurrently, we submit recent knowledge on how the cargo of EVs can serve as a valuable source of biomarkers for minimally invasive liquid biopsies, and its therapeutic potential, particularly as targeted drug delivery vehicles and immunomodulatory agents, showcasing their promise for enhancing the efficacy and safety of cancer treatments. By deciphering the intricate messages carried by EVs, we can gain a deeper understanding of cancer biology and develop more effective strategies for early detection, targeted therapy, and immunotherapy, paving the way for a new era of personalized and precise cancer medicine with the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Alena Semeradtova
- Institute of Photonics and Electronics of the CAS, Chaberská 1014/57, Prague, 182 51, Czech Republic.
| | - Michaela Liegertova
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Pasteurova 3632/15, Ústí Nad Labem, 40096, Czech Republic
| | - Regina Herma
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Pasteurova 3632/15, Ústí Nad Labem, 40096, Czech Republic
| | - Magdalena Capkova
- Institute of Photonics and Electronics of the CAS, Chaberská 1014/57, Prague, 182 51, Czech Republic
| | - Chiara Brignole
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy.
| | - Genny Del Zotto
- Core Facilities, Department of Research and Diagnostics, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy.
| |
Collapse
|
2
|
Kelly H, Inada M, Itoh Y. The Diverse Pathways for Cell Surface MT1-MMP Localization in Migratory Cells. Cells 2025; 14:209. [PMID: 39937000 PMCID: PMC11816416 DOI: 10.3390/cells14030209] [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: 10/22/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
Controlled cell migration is an essential biological process in health, while uncontrolled cell migration contributes to disease progression. For cells to migrate through tissue, they must first degrade the extracellular matrix (ECM), which acts as a physical barrier to cell migration. A type I transmembrane-type matrix metalloproteinase, MT1-MMP, is the key enzyme involved in this process. It has been extensively shown that MT1-MMP promotes the migration of different cell types in tissue, including fibroblasts, epithelial cells, endothelial cells, macrophages, mesenchymal stem cells, and cancer cells. MT1-MMP is tightly regulated at different levels, and its localization to leading-edge membrane structures is an essential process for MT1-MMP to promote cellular invasion. Different cells display different motility-associated membrane structures, which contribute to their invasive ability, and there are diverse mechanisms of MT1-MMP localization to these structures. In this article, we will discuss the current understanding of MT1-MMP regulation, in particular, localization mechanisms to these different motility-associated membrane structures.
Collapse
Affiliation(s)
- Hannah Kelly
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK;
| | - Masaki Inada
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei, Tokyo 184-8588, Japan;
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei, Tokyo 184-8588, Japan
| | - Yoshifumi Itoh
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK;
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei, Tokyo 184-8588, Japan
| |
Collapse
|
3
|
Grafinger OR, Hayward JJ, Meng Y, Geddes-McAlister J, Li Y, Mar S, Sheng M, Su B, Thillainadesan G, Lipsman N, Coppolino MG, Trant JF, Jerzak KJ, Leong HS. Cancer cell extravasation requires iplectin-mediated delivery of MT1-MMP at invadopodia. Br J Cancer 2024; 131:931-943. [PMID: 38969866 PMCID: PMC11369281 DOI: 10.1038/s41416-024-02782-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/10/2024] [Accepted: 06/21/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Invadopodia facilitate cancer cell extravasation, but the molecular mechanism whereby invadopodia-specific proteases such as MT1-MMP are called to invadopodia is unclear. METHODS Mass spectrometry and immunoprecipitation were used to identify interactors of MT1-MMP in metastatic breast cancer cells. After identification, siRNA and small molecule inhibitors were used to assess the effect these interactors had on cellular invasiveness. The chicken embryo chorioallantoic membrane (CAM) model was used to assess extravasation and invadopodia formation in vivo. RESULTS In metastatic breast cancer cells, MT1-MMP was found to associate with plectin, a cytolinker and scaffolding protein. Complex formation between plectin and MT1-MMP launches invadopodia formation, a subtype we termed iplectin (i = invadopodial). iPlectin delivers MT1-MMP to invadopodia and is indispensable for regulating cell surface levels of the enzyme. Genetic depletion of plectin with siRNA reduced invadopodia formation and cell invasion in vitro. In vivo extravasation efficiency assays and intravital imaging revealed iplectin to be a key contributor to invadopodia ultrastructure and essential for extravasation. Pharmacologic inhibition of plectin using the small molecule Plecstatin-1 (PST-1) abrogated MT1-MMP delivery to invadopodia and extravasation efficiency. CONCLUSIONS Anti-metastasis therapeutic approaches that target invadopodia are possible by disrupting interactions between MT1-MMP and iplectin. CLINICAL TRIAL REGISTRATION NUMBER NCT04608357.
Collapse
Affiliation(s)
- Olivia R Grafinger
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - John J Hayward
- Department of Chemistry, University of Windsor, Windsor, ON, Canada
| | - Ying Meng
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | | | - Yan Li
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Sara Mar
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Minzhi Sheng
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Boyang Su
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Gobi Thillainadesan
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Marc G Coppolino
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - John F Trant
- Department of Chemistry, University of Windsor, Windsor, ON, Canada
| | - Katarzyna J Jerzak
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Medical Oncology, Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Hon S Leong
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada.
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Wang H, Lakshmana MK, Fields GB. Identification of binding partners that facilitate membrane-type 5 matrix metalloproteinase (MT5-MMP) processing of amyloid precursor protein. J Cell Physiol 2024; 239:e31218. [PMID: 38345408 DOI: 10.1002/jcp.31218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 06/14/2024]
Abstract
One of the pathological hallmarks of Alzheimer's disease (AD) is the presence of extracellular deposits of amyloid beta (Aβ) peptide. In addition to Aβ as the core component of the amyloid plaque, the amyloid precursor protein (APP) processing fragment Aβ was also found accumulated around the plaque. The APPη pathway, mainly mediated by membrane-type 5 matrix metalloproteinase (MT5-MMP), represents an important factor in AD pathogenesis. The proamyloidogenic features of MT5-MMP could result from interactions with APP when trafficking between organelles, so determination of the location within the cell of APPη cleavage and interacting proteins of MT5-MMP affecting this process will be of priority in understanding the role of MT5-MMP in AD. In the present study, MT5-MMP was found to be located in the nucleus, cytosol, and cytosolic subcellular granules of CHO cells that stably expressed wild-type human APP751. MT5-MMP fusion proteins were constructed that could localize enzyme production in the Golgi apparatus, endosome, ER, mitochondria, or plasma membrane. The fusion proteins significantly increased sAPPη when directed to the endosome, Golgi apparatus, plasma membrane, or mitochondria. Since the C-terminal region of MT5-MMP is responsible for its intracellular location and trafficking, this domain was used as the bait in a yeast two-hybrid screen to identify MT5-MMP protein partners in a human brain cDNA library. Identified binding partners included N4BP2L1, TMX3, EIG121, bridging Integrator 1 (BIN1), RUFY4, HTRA1, and TMEM199. The binding of N4BP2L1, EIG121, BIN1, or TMX3 to MT5-MMP resulted in the most significant increase in sAPPη production. Thus, the action of MT5-MMP on APP occurs in multiple locations within the cell and is facilitated by site-specific binding partners.
Collapse
Affiliation(s)
- Hongjie Wang
- Department of Chemistry & Biochemistry, Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, Florida, USA
| | - Madepalli K Lakshmana
- Department of Immunology and Nano-Medicine, Florida International University, Miami, Florida, USA
| | - Gregg B Fields
- Department of Chemistry & Biochemistry, Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, Jupiter, Florida, USA
| |
Collapse
|
5
|
Carvalho LADC, Noma IHY, Uehara AH, Siena ÁDD, Osaki LH, Mori MP, Pinto NCDS, Freitas VM, Junior WAS, Smalley KSM, Maria-Engler SS. Modeling Melanoma Heterogeneity In Vitro: Redox, Resistance and Pigmentation Profiles. Antioxidants (Basel) 2024; 13:555. [PMID: 38790661 PMCID: PMC11118096 DOI: 10.3390/antiox13050555] [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: 03/20/2024] [Revised: 04/21/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Microenvironment and transcriptional plasticity generate subpopulations within the tumor, and the use of BRAF inhibitors (BRAFis) contributes to the rise and selection of resistant clones. We stochastically isolated subpopulations (C1, C2, and C3) from naïve melanoma and found that the clones demonstrated distinct morphology, phenotypic, and functional profiles: C1 was less proliferative, more migratory and invasive, less sensitive to BRAFis, less dependent on OXPHOS, more sensitive to oxidative stress, and less pigmented; C2 was more proliferative, less migratory and invasive, more sensitive to BRAFis, less sensitive to oxidative stress, and more pigmented; and C3 was less proliferative, more migratory and invasive, less sensitive to BRAFis, more dependent on OXPHOS, more sensitive to oxidative stress, and more pigmented. Hydrogen peroxide plays a central role in oxidative stress and cell signaling, and PRDXs are one of its main consumers. The intrinsically resistant C1 and C3 clones had lower MITF, PGC-1α, and PRDX1 expression, while C1 had higher AXL and decreased pigmentation markers, linking PRDX1 to clonal heterogeneity and resistance. PRDX2 is depleted in acquired BRAFi-resistant cells and acts as a redox sensor. Our results illustrate that decreased pigmentation markers are related to therapy resistance and decreased antioxidant defense.
Collapse
Affiliation(s)
- Larissa Anastacio da Costa Carvalho
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL 33612, USA; (L.A.d.C.C.); (K.S.M.S.)
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (I.H.Y.N.); (A.H.U.)
| | - Isabella Harumi Yonehara Noma
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (I.H.Y.N.); (A.H.U.)
| | - Adriana Hiromi Uehara
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (I.H.Y.N.); (A.H.U.)
| | - Ádamo Davi Diógenes Siena
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (Á.D.D.S.); (W.A.S.J.)
| | - Luciana Harumi Osaki
- Department of Cell Biology and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.H.O.); (V.M.F.)
| | - Mateus Prates Mori
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, SP, Brazil; (M.P.M.); (N.C.d.S.P.)
| | - Nadja Cristhina de Souza Pinto
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, SP, Brazil; (M.P.M.); (N.C.d.S.P.)
| | - Vanessa Morais Freitas
- Department of Cell Biology and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.H.O.); (V.M.F.)
| | - Wilson Araújo Silva Junior
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil; (Á.D.D.S.); (W.A.S.J.)
| | - Keiran S. M. Smalley
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL 33612, USA; (L.A.d.C.C.); (K.S.M.S.)
| | - Silvya Stuchi Maria-Engler
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (I.H.Y.N.); (A.H.U.)
| |
Collapse
|
6
|
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent proteinases that belong to the group of endopeptidases or matrixins. They are able to cleave a plethora of substrates, including components of the extracellular matrix and cell-surface-associated proteins, as well as intracellular targets. Accordingly, MMPs play key roles in a variety of physiological and pathological processes, such as tissue homeostasis and cancer cell invasion. MMP activity is exquisitely regulated at several levels, including pro-domain removal, association with inhibitors, intracellular trafficking and transport via extracellular vesicles. Moreover, the regulation of MMP activity is currently being rediscovered for the development of respective therapies for the treatment of cancer, as well as infectious, inflammatory and neurological diseases. In this Cell Science at a Glance article and the accompanying poster, we present an overview of the current knowledge regarding the regulation of MMP activity, the intra- and extra-cellular trafficking pathways of these enzymes and their diverse groups of target proteins, as well as their impact on health and disease.
Collapse
Affiliation(s)
- Sven Hey
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Stefan Linder
- Institut für medizinische Mikrobiologie, Virologie und Hygiene, Universitätsklinikum Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
7
|
Ku M, Yang J. Intracellular lipophilic network transformation induced by protease-specific endocytosis of fluorescent Au nanoclusters. NANO CONVERGENCE 2023; 10:26. [PMID: 37296273 DOI: 10.1186/s40580-023-00376-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023]
Abstract
The understanding of the endocytosis process of internalized nanomedicines through membrane biomarker is essential for the development of molecular-specific nanomedicines. In various recent reports, the metalloproteases have been identified as important markers during the metastasis of cancer cells. In particular, MT1-MMP has provoked concern due to its protease activity in the degradation of the extracellular matrix adjacent to tumors. Thus, in the current work, we have applied fluorescent Au nanoclusters which present strong resistance to chemical quenching to the investigation of MT1-MMP-mediated endocytosis. We synthesized protein-based Au nanocluster (PAuNC) and MT1-MMP-specific peptide was conjugated with PAuNC (pPAuNC) for monitoring protease-mediated endocytosis. The fluorescence capacity of pPAuNC was investigated and MT1-MMP-mediated intracellular uptake of pPAuNC was subsequently confirmed by a co-localization analysis using confocal microscopy and molecular competition test. Furthermore, we confirmed a change in the intracellular lipophilic network after an endocytosis event of pPAuNC. The identical lipophilic network change did not occur with the endocytosis of bare PAuNC. By classification of the branched network between the lipophilic organelles at the nanoscale, the image-based analysis of cell organelle networking allowed the evaluation of nanoparticle internalization and impaired cellular components after intracellular accumulation at a single-cell level. Our analyses suggest a methodology to achieve a better understanding of the mechanism by which nanoparticles enter cells.
Collapse
Affiliation(s)
- Minhee Ku
- Department of Radiology, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- Systems Molecular Radiology at Yonsei (SysMolRaY), Seoul, 03722, Republic of Korea
- Imaging of MechanoBiology (iMechBio) at Yonsei, Seoul, 03722, Republic of Korea
| | - Jaemoon Yang
- Department of Radiology, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea.
- Systems Molecular Radiology at Yonsei (SysMolRaY), Seoul, 03722, Republic of Korea.
- Imaging of MechanoBiology (iMechBio) at Yonsei, Seoul, 03722, Republic of Korea.
- Convergence Research Center for Systems Molecular Radiological Science, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
8
|
Banushi B, Joseph SR, Lum B, Lee JJ, Simpson F. Endocytosis in cancer and cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00574-6. [PMID: 37217781 DOI: 10.1038/s41568-023-00574-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/24/2023]
Abstract
Endocytosis is a complex process whereby cell surface proteins, lipids and fluid from the extracellular environment are packaged, sorted and internalized into cells. Endocytosis is also a mechanism of drug internalization into cells. There are multiple routes of endocytosis that determine the fate of molecules, from degradation in the lysosomes to recycling back to the plasma membrane. The overall rates of endocytosis and temporal regulation of molecules transiting through endocytic pathways are also intricately linked with signalling outcomes. This process relies on an array of factors, such as intrinsic amino acid motifs and post-translational modifications. Endocytosis is frequently disrupted in cancer. These disruptions lead to inappropriate retention of receptor tyrosine kinases on the tumour cell membrane, changes in the recycling of oncogenic molecules, defective signalling feedback loops and loss of cell polarity. In the past decade, endocytosis has emerged as a pivotal regulator of nutrient scavenging, response to and regulation of immune surveillance and tumour immune evasion, tumour metastasis and therapeutic drug delivery. This Review summarizes and integrates these advances into the understanding of endocytosis in cancer. The potential to regulate these pathways in the clinic to improve cancer therapy is also discussed.
Collapse
Affiliation(s)
- Blerida Banushi
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Shannon R Joseph
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Benedict Lum
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Jason J Lee
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
9
|
Cytoplasmic Tail of MT1-MMP: A Hub of MT1-MMP Regulation and Function. Int J Mol Sci 2023; 24:ijms24065068. [PMID: 36982142 PMCID: PMC10049710 DOI: 10.3390/ijms24065068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/03/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
MT1-MMP (MMP-14) is a multifunctional protease that regulates ECM degradation, activation of other proteases, and a variety of cellular processes, including migration and viability in physiological and pathological contexts. Both the localization and signal transduction capabilities of MT1-MMP are dependent on its cytoplasmic domain that constitutes the final 20 C-terminal amino acids, while the rest of the protease is extracellular. In this review, we summarize the ways in which the cytoplasmic tail is involved in regulating and enacting the functions of MT1-MMP. We also provide an overview of known interactors of the MT1-MMP cytoplasmic tail and the functional significance of these interactions, as well as further insight into the mechanisms of cellular adhesion and invasion that are regulated by the cytoplasmic tail.
Collapse
|
10
|
Genetic Biodiversity and Posttranslational Modifications of Protease Serine Endopeptidase in Different Strains of Sordaria fimicola. BIOMED RESEARCH INTERNATIONAL 2023; 2023:2088988. [PMID: 36814796 PMCID: PMC9940969 DOI: 10.1155/2023/2088988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/15/2023]
Abstract
Genetic variations (mutation, crossing over, and recombination) act as a source for the gradual alternation in phenotype along a geographic transect where the environment changes. Posttranslational modifications (PTMs) predicted modifications successfully in different and the same species of living organisms. Protein diversity of living organisms is predicted by PTMs. Environmental stresses change nucleotides to produce alternations in protein structures, and these alternations have been examined through bioinformatics tools. The goal of the current study is to search the diversity of genes and posttranslational modifications of protease serine endopeptidase in various strains of Sordaria fimicola. The S. fimicola's genomic DNA was utilized to magnify the protease serine endopeptidase (SP2) gene; the size of the product was 700 and 1400 base pairs. Neurospora crassa was taken as the reference strain for studying the multiple sequence alignment of the nucleotide sequence. Six polymorphic sites of six strains of S. fimicola with respect to N. crassa were under observation. Different bioinformatics tools, i.e., NetPhos 3.1, NetNES 1.1 Server, YinOYang1.2, and Mod Pred, to search phosphorylation sites, acetylation, nuclear export signals, O-glycosylation, and methylation, respectively, were used to predict PTMs. The findings of the current study were 35 phosphorylation sites on the residues of serine for protease SP2 in SFS and NFS strains of S. fimicola and N. crassa. The current study supported us to get the reality of genes involved in protease production in experimental fungi. Our study examined the genetic biodiversity in six strains of S. fimicola which were caused by stressful environments, and these variations are a strong reason for evolution. In this manuscript, we predicted posttranslational modifications of protease serine endopeptidase in S. fimicola obtained from different sites, for the first time, to see the effect of environmental stress on nucleotides, amino acids, and proteases and to study PTMs by using various bioinformatics tools. This research confirmed the genetic biodiversity and PTMs in six strains of S. fimicola, and the designed primers also provided strong evidence for the presence of protease serine endopeptidase in each strain of S. fimicola.
Collapse
|
11
|
Linder S, Cervero P, Eddy R, Condeelis J. Mechanisms and roles of podosomes and invadopodia. Nat Rev Mol Cell Biol 2023; 24:86-106. [PMID: 36104625 DOI: 10.1038/s41580-022-00530-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 01/28/2023]
Abstract
Cell invasion into the surrounding extracellular matrix or across tissue boundaries and endothelial barriers occurs in both physiological and pathological scenarios such as immune surveillance or cancer metastasis. Podosomes and invadopodia, collectively called 'invadosomes', are actin-based structures that drive the proteolytic invasion of cells, by forming highly regulated platforms for the localized release of lytic enzymes that degrade the matrix. Recent advances in high-resolution microscopy techniques, in vivo imaging and high-throughput analyses have led to considerable progress in understanding mechanisms of invadosomes, revealing the intricate inner architecture of these structures, as well as their growing repertoire of functions that extends well beyond matrix degradation. In this Review, we discuss the known functions, architecture and regulatory mechanisms of podosomes and invadopodia. In particular, we describe the molecular mechanisms of localized actin turnover and microtubule-based cargo delivery, with a special focus on matrix-lytic enzymes that enable proteolytic invasion. Finally, we point out topics that should become important in the invadosome field in the future.
Collapse
Affiliation(s)
- Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, Hamburg, Germany.
| | - Pasquale Cervero
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Robert Eddy
- Department of Pathology, Albert Einstein College of Medicine, New York, NY, USA
| | - John Condeelis
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Cartón-García F, Brotons B, Anguita E, Dopeso H, Tarragona J, Nieto R, García-Vidal E, Macaya I, Zagyva Z, Dalmau M, Sánchez-Martín M, van Ijzendoorn SCD, Landolfi S, Hernandez-Losa J, Schwartz Jr S, Matias-Guiu X, Ramón y Cajal S, Martínez-Barriocanal Á, Arango D. Myosin Vb as a tumor suppressor gene in intestinal cancer. Oncogene 2022; 41:5279-5288. [DOI: 10.1038/s41388-022-02508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022]
|
13
|
Sandoval L, Fuentealba LM, Marzolo MP. Participation of OCRL1, and APPL1, in the expression, proteolysis, phosphorylation and endosomal trafficking of megalin: Implications for Lowe Syndrome. Front Cell Dev Biol 2022; 10:911664. [PMID: 36340038 PMCID: PMC9630597 DOI: 10.3389/fcell.2022.911664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Megalin/LRP2 is the primary multiligand receptor for the re-absorption of low molecular weight proteins in the proximal renal tubule. Its function is significantly dependent on its endosomal trafficking. Megalin recycling from endosomal compartments is altered in an X-linked disease called Lowe Syndrome (LS), caused by mutations in the gene encoding for the phosphatidylinositol 5-phosphatase OCRL1. LS patients show increased low-molecular-weight proteins with reduced levels of megalin ectodomain in the urine and accumulation of the receptor in endosomal compartments of the proximal tubule cells. To gain insight into the deregulation of megalin in the LS condition, we silenced OCRL1 in different cell lines to evaluate megalin expression finding that it is post-transcriptionally regulated. As an indication of megalin proteolysis, we detect the ectodomain of the receptor in the culture media. Remarkably, in OCRL1 silenced cells, megalin ectodomain secretion appeared significantly reduced, according to the observation in the urine of LS patients. Besides, the silencing of APPL1, a Rab5 effector associated with OCRL1 in endocytic vesicles, also reduced the presence of megalin’s ectodomain in the culture media. In both silencing conditions, megalin cell surface levels were significantly decreased. Considering that GSK3ß-mediated megalin phosphorylation reduces receptor recycling, we determined that the endosomal distribution of megalin depends on its phosphorylation status and OCRL1 function. As a physiologic regulator of GSK3ß, we focused on insulin signaling that reduces kinase activity. Accordingly, megalin phosphorylation was significantly reduced by insulin in wild-type cells. Moreover, even though in cells with low activity of OCRL1 the insulin response was reduced, the phosphorylation of megalin was significantly decreased and the receptor at the cell surface increased, suggesting a protective role of insulin in a LS cellular model.
Collapse
Affiliation(s)
- Lisette Sandoval
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Luz M. Fuentealba
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María-Paz Marzolo
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: María-Paz Marzolo,
| |
Collapse
|
14
|
Novel Roles of MT1-MMP and MMP-2: Beyond the Extracellular Milieu. Int J Mol Sci 2022; 23:ijms23179513. [PMID: 36076910 PMCID: PMC9455801 DOI: 10.3390/ijms23179513] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 12/14/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are critical enzymes involved in a variety of cellular processes. MMPs are well known for their ability to degrade the extracellular matrix (ECM) and their extracellular role in cell migration. Recently, more research has been conducted on investigating novel subcellular localizations of MMPs and their intracellular roles at their respective locations. In this review article, we focus on the subcellular localization and novel intracellular roles of two closely related MMPs: membrane-type-1 matrix metalloproteinase (MT1-MMP) and matrix metalloproteinase-2 (MMP-2). Although MT1-MMP is commonly known to localize on the cell surface, the protease also localizes to the cytoplasm, caveolae, Golgi, cytoskeleton, centrosome, and nucleus. At these subcellular locations, MT1-MMP functions in cell migration, macrophage metabolism, invadopodia development, spindle formation and gene expression, respectively. Similar to MT1-MMP, MMP-2 localizes to the caveolae, mitochondria, cytoskeleton, nucleus and nucleolus and functions in calcium regulation, contractile dysfunction, gene expression and ribosomal RNA transcription. Our particular interest lies in the roles MMP-2 and MT1-MMP serve within the nucleus, as they may provide critical insights into cancer epigenetics and tumor migration and invasion. We suggest that targeting nuclear MT1-MMP or MMP-2 to reduce or halt cell proliferation and migration may lead to the development of new therapies for cancer and other diseases.
Collapse
|
15
|
Shannon N, Gravelle R, Cunniff B. Mitochondrial trafficking and redox/phosphorylation signaling supporting cell migration phenotypes. Front Mol Biosci 2022; 9:925755. [PMID: 35936783 PMCID: PMC9355248 DOI: 10.3389/fmolb.2022.925755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Regulation of cell signaling cascades is critical in making sure the response is activated spatially and for a desired duration. Cell signaling cascades are spatially and temporally controlled through local protein phosphorylation events which are determined by the activation of specific kinases and/or inactivation of phosphatases to elicit a complete and thorough response. For example, A-kinase-anchoring proteins (AKAPs) contribute to the local regulated activity protein kinase A (PKA). The activity of kinases and phosphatases can also be regulated through redox-dependent cysteine modifications that mediate the activity of these proteins. A primary example of this is the activation of the epidermal growth factor receptor (EGFR) and the inactivation of the phosphatase and tensin homologue (PTEN) phosphatase by reactive oxygen species (ROS). Therefore, the local redox environment must play a critical role in the timing and magnitude of these events. Mitochondria are a primary source of ROS and energy (ATP) that contributes to redox-dependent signaling and ATP-dependent phosphorylation events, respectively. The strategic positioning of mitochondria within cells contributes to intracellular gradients of ROS and ATP, which have been shown to correlate with changes to protein redox and phosphorylation status driving downstream cellular processes. In this review, we will discuss the relationship between subcellular mitochondrial positioning and intracellular ROS and ATP gradients that support dynamic oxidation and phosphorylation signaling and resulting cellular effects, specifically associated with cell migration signaling.
Collapse
Affiliation(s)
- Nathaniel Shannon
- Department of Pathology and Laboratory Medicine, Redox Biology Program, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Randi Gravelle
- Department of Pathology and Laboratory Medicine, Redox Biology Program, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, Redox Biology Program, University of Vermont Larner College of Medicine, Burlington, VT, United States
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States
| |
Collapse
|
16
|
de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, Devel L, Dufour A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol Rev 2022; 74:712-768. [PMID: 35738680 DOI: 10.1124/pharmrev.121.000349] [Citation(s) in RCA: 195] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The first matrix metalloproteinase (MMP) was discovered in 1962 from the tail of a tadpole by its ability to degrade collagen. As their name suggests, matrix metalloproteinases are proteases capable of remodeling the extracellular matrix. More recently, MMPs have been demonstrated to play numerous additional biologic roles in cell signaling, immune regulation, and transcriptional control, all of which are unrelated to the degradation of the extracellular matrix. In this review, we will present milestones and major discoveries of MMP research, including various clinical trials for the use of MMP inhibitors. We will discuss the reasons behind the failures of most MMP inhibitors for the treatment of cancer and inflammatory diseases. There are still misconceptions about the pathophysiological roles of MMPs and the best strategies to inhibit their detrimental functions. This review aims to discuss MMPs in preclinical models and human pathologies. We will discuss new biochemical tools to track their proteolytic activity in vivo and ex vivo, in addition to future pharmacological alternatives to inhibit their detrimental functions in diseases. SIGNIFICANCE STATEMENT: Matrix metalloproteinases (MMPs) have been implicated in most inflammatory, autoimmune, cancers, and pathogen-mediated diseases. Initially overlooked, MMP contributions can be both beneficial and detrimental in disease progression and resolution. Thousands of MMP substrates have been suggested, and a few hundred have been validated. After more than 60 years of MMP research, there remain intriguing enigmas to solve regarding their biological functions in diseases.
Collapse
Affiliation(s)
- Luiz G N de Almeida
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Hayley Thode
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Yekta Eslambolchi
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sameeksha Chopra
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Daniel Young
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sean Gill
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Laurent Devel
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Antoine Dufour
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| |
Collapse
|
17
|
Linking Late Endosomal Cholesterol with Cancer Progression and Anticancer Drug Resistance. Int J Mol Sci 2022; 23:ijms23137206. [PMID: 35806209 PMCID: PMC9267071 DOI: 10.3390/ijms23137206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer cells undergo drastic metabolic adaptions to cover increased bioenergetic needs, contributing to resistance to therapies. This includes a higher demand for cholesterol, which often coincides with elevated cholesterol uptake from low-density lipoproteins (LDL) and overexpression of the LDL receptor in many cancers. This implies the need for cancer cells to accommodate an increased delivery of LDL along the endocytic pathway to late endosomes/lysosomes (LE/Lys), providing a rapid and effective distribution of LDL-derived cholesterol from LE/Lys to other organelles for cholesterol to foster cancer growth and spread. LDL-cholesterol exported from LE/Lys is facilitated by Niemann–Pick Type C1/2 (NPC1/2) proteins, members of the steroidogenic acute regulatory-related lipid transfer domain (StARD) and oxysterol-binding protein (OSBP) families. In addition, lysosomal membrane proteins, small Rab GTPases as well as scaffolding proteins, including annexin A6 (AnxA6), contribute to regulating cholesterol egress from LE/Lys. Here, we summarize current knowledge that links upregulated activity and expression of cholesterol transporters and related proteins in LE/Lys with cancer growth, progression and treatment outcomes. Several mechanisms on how cellular distribution of LDL-derived cholesterol from LE/Lys influences cancer cell behavior are reviewed, some of those providing opportunities for treatment strategies to reduce cancer progression and anticancer drug resistance.
Collapse
|
18
|
Thuault S, Ghossoub R, David G, Zimmermann P. A Journey on Extracellular Vesicles for Matrix Metalloproteinases: A Mechanistic Perspective. Front Cell Dev Biol 2022; 10:886381. [PMID: 35669514 PMCID: PMC9163832 DOI: 10.3389/fcell.2022.886381] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/27/2022] [Indexed: 12/15/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are key players in matrix remodeling and their function has been particularly investigated in cancer biology. Indeed, through extracellular matrix (ECM) degradation and shedding of diverse cell surface macromolecules, they are implicated in different steps of tumor development, from local expansion by growth to tissue invasion and metastasis. Interestingly, MMPs are also components of extracellular vesicles (EVs). EVs are membrane-limited organelles that cells release in their extracellular environment. These "secreted" vesicles are now well accepted players in cell-to-cell communication. EVs have received a lot of interest in recent years as they are also envisioned as sources of biomarkers and as potentially outperforming vehicles for the delivery of therapeutics. Molecular machineries governing EV biogenesis, cargo loading and delivery to recipient cells are complex and still under intense investigation. In this review, we will summarize the state of the art of our knowledge about the molecular mechanisms implicated in MMP trafficking and secretion. We focus on MT1-MMP, a major effector of invasive cell behavior. We will also discuss how this knowledge is of interest for a better understanding of EV-loading of MMPs. Such knowledge might be of use to engineer novel strategies for cancer treatment. A better understanding of these mechanisms could also be used to design more efficient EV-based therapies.
Collapse
Affiliation(s)
- Sylvie Thuault
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue 2018, CNRS, Inserm, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
| | - Rania Ghossoub
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue 2018, CNRS, Inserm, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
| | - Guido David
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue 2018, CNRS, Inserm, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
- Department of Human Genetics, KU Leuven, University of Leuven, Leuven, Belgium
| | - Pascale Zimmermann
- Centre de Recherche en Cancérologie de Marseille (CRCM), Equipe Labellisée Ligue 2018, CNRS, Inserm, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
- Department of Human Genetics, KU Leuven, University of Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Brasher MI, Chafe SC, McDonald PC, Nemirovsky O, Gorshtein G, Gerbec ZJ, Brown WS, Grafinger OR, Marchment M, Matus E, Dedhar S, Coppolino MG. Syntaxin4-Munc18c Interaction Promotes Breast Tumor Invasion and Metastasis by Regulating MT1-MMP Trafficking. Mol Cancer Res 2022; 20:434-445. [PMID: 34876482 PMCID: PMC9306282 DOI: 10.1158/1541-7786.mcr-20-0527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/18/2020] [Accepted: 11/24/2021] [Indexed: 01/07/2023]
Abstract
Invasion of neighboring extracellular matrix (ECM) by malignant tumor cells is a hallmark of metastatic progression. This invasion can be mediated by subcellular structures known as invadopodia, the function of which depends upon soluble N-ethylmaleimide-sensitive factor-activating protein receptor (SNARE)-mediated vesicular transport of cellular cargo. Recently, it has been shown the SNARE Syntaxin4 (Stx4) mediates trafficking of membrane type 1-matrix metalloproteinase (MT1-MMP) to invadopodia, and that Stx4 is regulated by Munc18c in this context. Here, it is observed that expression of a construct derived from the N-terminus of Stx4, which interferes with Stx4-Munc18c interaction, leads to perturbed trafficking of MT1-MMP, and reduced invadopodium-based invasion in vitro, in models of triple-negative breast cancer (TNBC). Expression of Stx4 N-terminus also led to increased survival and markedly reduced metastatic burden in multiple TNBC models in vivo. The findings are the first demonstration that disrupting Stx4-Munc18c interaction can dramatically alter metastatic progression in vivo, and suggest that this interaction warrants further investigation as a potential therapeutic target. IMPLICATIONS Disrupting the interaction of Syntaxin4 and Munc18c may be a useful approach to perturb trafficking of MT1-MMP and reduce metastatic potential of breast cancers.
Collapse
Affiliation(s)
- Megan I. Brasher
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Shawn C. Chafe
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Paul C. McDonald
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Oksana Nemirovsky
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Genya Gorshtein
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Zachary J. Gerbec
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Wells S. Brown
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Olivia R. Grafinger
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Matthew Marchment
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Esther Matus
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Shoukat Dedhar
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, British Columbia, Canada.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Marc G. Coppolino
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada.,Corresponding Author: Marc G. Coppolino, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario N1G 2W1, Canada. E-mail:
| |
Collapse
|
20
|
Hey S, Ratt A, Linder S. There and back again: Intracellular trafficking, release and recycling of matrix metalloproteinases. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119189. [PMID: 34973301 DOI: 10.1016/j.bbamcr.2021.119189] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/26/2022]
Abstract
Matrix metalloproteinases are a family of zinc-dependent endopeptidases that are involved in a large variety of proteolytic processes in physiological and pathological scenarios, including immune cell surveillance, tissue homeostasis, or tumor cell metastasis. This is based on their ability to cleave a plethora of substrates that include components of the extracellular matrix, but also cell surface-associated and intracellular proteins. Accordingly, a tight regulatory web has evolved that closely regulates spatiotemporal activity of specific MMPs. An often underappreciated mechanism of MMP regulation involves their trafficking to and from specific subcellular sites that require MMP activity only for a certain period. In this review, we focus on the current knowledge of MMP intracellular trafficking, their secretion or surface exposure, as well as their recycling back from the cell surface. We discuss molecular mechanisms that enable these steps, in particular microtubule-dependent motility of vesicles that is driven by molecular motors and directed by vesicle regulatory proteins. Finally, we also point out open questions in the field of MMP motility that may become important in the future.
Collapse
Affiliation(s)
- Sven Hey
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany
| | - Artur Ratt
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany
| | - Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
21
|
Matrix Metalloproteinases Shape the Tumor Microenvironment in Cancer Progression. Int J Mol Sci 2021; 23:ijms23010146. [PMID: 35008569 PMCID: PMC8745566 DOI: 10.3390/ijms23010146] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer progression with uncontrolled tumor growth, local invasion, and metastasis depends largely on the proteolytic activity of numerous matrix metalloproteinases (MMPs), which affect tissue integrity, immune cell recruitment, and tissue turnover by degrading extracellular matrix (ECM) components and by releasing matrikines, cell surface-bound cytokines, growth factors, or their receptors. Among the MMPs, MMP-14 is the driving force behind extracellular matrix and tissue destruction during cancer invasion and metastasis. MMP-14 also influences both intercellular as well as cell-matrix communication by regulating the activity of many plasma membrane-anchored and extracellular proteins. Cancer cells and other cells of the tumor stroma, embedded in a common extracellular matrix, interact with their matrix by means of various adhesive structures, of which particularly invadopodia are capable to remodel the matrix through spatially and temporally finely tuned proteolysis. As a deeper understanding of the underlying functional mechanisms is beneficial for the development of new prognostic and predictive markers and for targeted therapies, this review examined the current knowledge of the interplay of the various MMPs in the cancer context on the protein, subcellular, and cellular level with a focus on MMP14.
Collapse
|
22
|
Liu BHM, Tey SK, Mao X, Ma APY, Yeung CLS, Wong SWK, Ng TH, Xu Y, Yao Y, Fung EYM, Tan KV, Khong P, Ho DW, Ng IO, Tang AHN, Cai SH, Yun JP, Yam JWP. TPI1-reduced extracellular vesicles mediated by Rab20 downregulation promotes aerobic glycolysis to drive hepatocarcinogenesis. J Extracell Vesicles 2021; 10:e12135. [PMID: 34401050 PMCID: PMC8357635 DOI: 10.1002/jev2.12135] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/30/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
Rab GTPases are major mediators that ensure the proper spatiotemporal regulation of intracellular trafficking. Functional impairment and altered expression of Rab proteins have been revealed in various human cancers. There is an emerging evidence about the role of Rab proteins in the biogenesis of extracellular vesicles (EVs). In hepatocellular carcinoma (HCC), using RNA sequencing comparing expression profiles of adjacent non-tumorous tissues and HCC, Rab20 is identified to be the most frequently downregulated Rab member in HCC. Functionally, restoration of Rab20 in metastatic HCC cells results in the release of EVs with a diminished activity to promote cell growth, motility and metastasis. Conversely, EVs released from normal liver cells with Rab20 knockdown loses suppressive effect on HCC cell growth and motility. Proteomic profiling revealed the level of triosephosphate isomerase 1 (TPI1), a glycolytic enzyme, in EVs to be positively associated with Rab20 expression of the releasing cells. TPI1 targeted to be expressed in EVs released by Rab20 knockdown cells compromises the oncogenic activity of EVs. Besides, EVs released by TPI1 knockdown cells recapitulates the promoting effect of EVs derived from HCC cells with Rab20 underexpression. Aerobic glycolysis is beneficial to the survival and proliferation of tumour cells. Here, we observed that the enhanced cell growth and motility are driven by the enhanced aerobic glycolysis induced by EVs with reduced TPI1. The addition of glycolytic inhibitor blocks the promoting effect of EVs with reduced TPI1. Taken together, our study provides a mechanistic link among tumour cell-derived EVs and glucose metabolism in HCC with Rab20 deregulation.
Collapse
Affiliation(s)
- Bonnie Hei Man Liu
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Sze Keong Tey
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Xiaowen Mao
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Angel Po Yee Ma
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Cherlie Lot Sum Yeung
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Samuel Wan Ki Wong
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Tung Him Ng
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Yi Xu
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Department of Hepatopancreatobiliary SurgerySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Yue Yao
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- Department of EndocrinologySecond Affiliated Hospital of Harbin Medical UniversityHarbinChina
| | - Eva Yi Man Fung
- Department of ChemistryState Key Laboratory of Synthetic ChemistryThe University of Hong KongHong KongChina
| | - Kel Vin Tan
- Department of Diagnostic RadiologyQueen Mary Hospitalthe University of Hong KongHong KongChina
| | - Pek‐Lan Khong
- Department of Diagnostic RadiologyQueen Mary Hospitalthe University of Hong KongHong KongChina
| | - Daniel Wai‐Hung Ho
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- State Key Laboratory of Liver Research (The University of Hong Kong)Hong KongChina
| | - Irene Oi‐Lin Ng
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- State Key Laboratory of Liver Research (The University of Hong Kong)Hong KongChina
| | - Alexander Hin Ning Tang
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Shao Hang Cai
- Department of Infectious DiseasesNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jing Ping Yun
- Department of PathologySun Yat‐sen University Cancer CentreGuangzhouChina
| | - Judy Wai Ping Yam
- Department of PathologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- State Key Laboratory of Liver Research (The University of Hong Kong)Hong KongChina
| |
Collapse
|
23
|
Xia XD, Alabi A, Wang M, Gu HM, Yang RZ, Wang G, Zhang DW. Membrane-type I matrix metalloproteinase (MT1-MMP), lipid metabolism and therapeutic implications. J Mol Cell Biol 2021; 13:513-526. [PMID: 34297054 PMCID: PMC8530520 DOI: 10.1093/jmcb/mjab048] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 11/13/2022] Open
Abstract
Lipids exert many essential physiological functions, such as serving as a structural component of biological membranes, storing energy, and regulating cell signal transduction. Dysregulation of lipid metabolism can lead to dyslipidemia related to various human diseases, such as obesity, diabetes, and cardiovascular disease. Therefore, lipid metabolism is strictly regulated through multiple mechanisms at different levels, including the extracellular matrix. Membrane-type I matrix metalloproteinase (MT1-MMP), a zinc-dependent endopeptidase, proteolytically cleaves extracellular matrix components, and non-matrix proteins, thereby regulating many physiological and pathophysiological processes. Emerging evidence supports the vital role of MT1-MMP in lipid metabolism. For example, MT1-MMP mediates ectodomain shedding of low-density lipoprotein receptor and increases plasma low-density lipoprotein cholesterol levels and the development of atherosclerosis. It also increases the vulnerability of atherosclerotic plaque by promoting collagen cleavage. Furthermore, it can cleave the extracellular matrix of adipocytes, affecting adipogenesis and the development of obesity. Therefore, the activity of MT1-MMP is strictly regulated by multiple mechanisms, such as autocatalytic cleavage, endocytosis and exocytosis, and post-translational modifications. Here, we summarize the latest advances in MT1-MMP, mainly focusing on its role in lipid metabolism, the molecular mechanisms regulating the function and expression of MT1-MMP, and their pharmacotherapeutic implications.
Collapse
Affiliation(s)
- Xiao-Dan Xia
- Department of Orthopedics, The Sixth Affiliated Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan 511500, China.,Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6R 2G3, Canada
| | - Adekunle Alabi
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6R 2G3, Canada
| | - Maggie Wang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6R 2G3, Canada
| | - Hong-Mei Gu
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6R 2G3, Canada
| | - Rui Zhe Yang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6R 2G3, Canada
| | - Guiqing Wang
- Department of Orthopedics, The Sixth Affiliated Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan 511500, China
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6R 2G3, Canada
| |
Collapse
|
24
|
Romano R, Calcagnile M, Margiotta A, Franci L, Chiariello M, Alifano P, Bucci C. RAB7A Regulates Vimentin Phosphorylation through AKT and PAK. Cancers (Basel) 2021; 13:cancers13092220. [PMID: 34066419 PMCID: PMC8125308 DOI: 10.3390/cancers13092220] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary RAB7A (RAs-related in Brain 7A) is a master regulator of intracellular traffic controlling transport to late endosomes and lysosomes, two organelles of the endocytic pathway important for degradation. Thanks to this function, RAB7A is also involved in cellular processes linked to cancer, such as apoptosis, cytoskeletal reorganization, and cell migration. Therefore, the interest in the role of RAB7A in cancer progression is increasing. Previously, we demonstrated that RAB7A regulates phosphorylation and assembly of vimentin, a cytoskeletal intermediate filament protein, which is also an important mesenchymal marker of cancer cells. The aim of the present study is the identification of the kinases responsible for vimentin phosphorylation whose activity is affected by the modulation of RAB7A expression. We found that RAB7A is able to regulate AKT (also called protein kinase B or PKB) and PAK1 (P21-Activated Kinase 1) and several of their downstream effectors, which control proliferation, apoptosis, survival, migration, and invasion. These data suggest that RAB7A could have a key role in cancer development. Abstract RAB7A is a small GTPase that controls the late endocytic pathway but also cell migration through RAC1 (Ras-related C3 botulinum toxin substrate 1) and vimentin. In fact, RAB7A regulates vimentin phosphorylation at different sites and vimentin assembly, and, in this study, we identified vimentin domains interacting with RAB7A. As several kinases could be responsible for vimentin phosphorylation, we investigated whether modulation of RAB7A expression affects the activity of these kinases. We discovered that RAB7A regulates AKT and PAK1, and we demonstrated that increased vimentin phosphorylation at Ser38 (Serine 38), observed upon RAB7A overexpression, is due to AKT activity. As AKT and PAK1 are key regulators of several cellular events, we investigated if RAB7A could have a role in these processes by modulating AKT and PAK1 activity. We found that RAB7A protein levels affected beta-catenin and caspase 9 expression. We also observed the downregulation of cofilin-1 and decreased matrix metalloproteinase 2 (MMP2) activity upon RAB7A silencing. Altogether these results demonstrate that RAB7A regulates AKT and PAK1 kinases, affecting their downstream effectors and the processes they regulate, suggesting that RAB7A could have a role in a number of cancer hallmarks.
Collapse
Affiliation(s)
- Roberta Romano
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, 73100 Lecce, Italy; (R.R.); (M.C.); (A.M.); (P.A.)
| | - Matteo Calcagnile
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, 73100 Lecce, Italy; (R.R.); (M.C.); (A.M.); (P.A.)
| | - Azzurra Margiotta
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, 73100 Lecce, Italy; (R.R.); (M.C.); (A.M.); (P.A.)
| | - Lorenzo Franci
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), 53100 Siena, Italy; (L.F.); (M.C.)
- Core Research Laboratory (CRL), Istituto per lo Studio, La Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Mario Chiariello
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR), 53100 Siena, Italy; (L.F.); (M.C.)
- Core Research Laboratory (CRL), Istituto per lo Studio, La Prevenzione e la Rete Oncologica (ISPRO), 53100 Siena, Italy
| | - Pietro Alifano
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, 73100 Lecce, Italy; (R.R.); (M.C.); (A.M.); (P.A.)
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, 73100 Lecce, Italy; (R.R.); (M.C.); (A.M.); (P.A.)
- Correspondence: ; Tel.: +39-0832-298900
| |
Collapse
|
25
|
Hu J, Ding X, Tian S, Chu Y, Liu Z, Li Y, Li X, Wang G, Wang L, Wang Z. TRIM39 deficiency inhibits tumor progression and autophagic flux in colorectal cancer via suppressing the activity of Rab7. Cell Death Dis 2021; 12:391. [PMID: 33846303 PMCID: PMC8041807 DOI: 10.1038/s41419-021-03670-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022]
Abstract
The biological function of TRIM39, a member of TRIM family, remains largely unexplored in cancer, especially in colorectal cancer (CRC). In this study, we show that TRIM39 is upregulated in tumor tissues compared to adjacent normal tissues and associated with poor prognosis in CRC. Functional studies demonstrate that TRIM39 deficiency restrains CRC progression in vitro and in vivo. Our results further find that TRIM39 is a positive regulator of autophagosome–lysosome fusion. Mechanistically, TRIM39 interacts with Rab7 and promotes its activity via inhibiting its ubiquitination at lysine 191 residue. Depletion of TRIM39 inhibits CRC progression and autophagic flux in a Rab7 activity-dependent manner. Moreover, TRIM39 deficiency suppresses CRC progression through inhibiting autophagic degradation of p53. Thus, our findings uncover the roles as well as the relevant mechanisms of TRIM39 in CRC and establish a functional relationship between autophagy and CRC progression, which may provide promising approaches for the treatment of CRC.
Collapse
Affiliation(s)
- Jia Hu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Xueliang Ding
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Shaobo Tian
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Yanan Chu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Zhibo Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Yuqin Li
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Xiaoqiong Li
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China. .,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China. .,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
26
|
Pedersen NM, Wenzel EM, Wang L, Antoine S, Chavrier P, Stenmark H, Raiborg C. Protrudin-mediated ER-endosome contact sites promote MT1-MMP exocytosis and cell invasion. J Cell Biol 2021; 219:151827. [PMID: 32479595 PMCID: PMC7401796 DOI: 10.1083/jcb.202003063] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer cells break tissue barriers by use of small actin-rich membrane protrusions called invadopodia. Complete invadopodia maturation depends on protrusion outgrowth and the targeted delivery of the matrix metalloproteinase MT1-MMP via endosomal transport by mechanisms that are not known. Here, we show that the ER protein Protrudin orchestrates invadopodia maturation and function. Protrudin formed contact sites with MT1-MMP-positive endosomes that contained the RAB7-binding Kinesin-1 adaptor FYCO1, and depletion of RAB7, FYCO1, or Protrudin inhibited MT1-MMP-dependent extracellular matrix degradation and cancer cell invasion by preventing anterograde translocation and exocytosis of MT1-MMP. Moreover, when endosome translocation or exocytosis was inhibited by depletion of Protrudin or Synaptotagmin VII, respectively, invadopodia were unable to expand and elongate. Conversely, when Protrudin was overexpressed, noncancerous cells developed prominent invadopodia-like protrusions and showed increased matrix degradation and invasion. Thus, Protrudin-mediated ER-endosome contact sites promote cell invasion by facilitating translocation of MT1-MMP-laden endosomes to the plasma membrane, enabling both invadopodia outgrowth and MT1-MMP exocytosis.
Collapse
Affiliation(s)
- Nina Marie Pedersen
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Eva Maria Wenzel
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ling Wang
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Sandra Antoine
- Research Center, Institut Curie, Membrane and Cytoskeleton Dynamics and Cell and Tissue Imaging Facility, Centre National de la Recherche Scientifique UMR 144, Paris, France
| | - Philippe Chavrier
- Research Center, Institut Curie, Membrane and Cytoskeleton Dynamics and Cell and Tissue Imaging Facility, Centre National de la Recherche Scientifique UMR 144, Paris, France
| | - Harald Stenmark
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Camilla Raiborg
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
27
|
Grafinger OR, Gorshtein G, Stirling T, Geddes-McAlister J, Coppolino MG. Inhibition of β1 integrin induces its association with MT1-MMP and decreases MT1-MMP internalization and cellular invasiveness. Cell Signal 2021; 83:109984. [PMID: 33744418 DOI: 10.1016/j.cellsig.2021.109984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 11/17/2022]
Abstract
Integrin signaling plays a fundamental role in the establishment of focal adhesions and the subsequent formation of invadopodia in malignant cancer cells. Invadopodia facilitate localized adhesion and degradation of the extracellular matrix (ECM), which promote tumour cell invasion and metastasis. Degradation of ECM components is often driven by membrane type-1 matrix metalloproteinase (MT1-MMP), and we have recently shown that regulation of enzyme internalization is dependent on signaling downstream of β1 integrin. Phosphorylation of the cytoplasmic tail of MT1-MMP is required for its internalization and delivery to Rab5-marked early endosomes, where it is then able to be recycled to new sites of invadopodia formation and promote invasion. Here we found that inhibition of β1 integrin, using the antibody AIIB2, inhibited the internalization and recycling of MT1-MMP that is necessary to support long-term cellular invasion. MT1-MMP and β1 integrin were sequestered at the cell surface when β1-integrin was inhibited, and their association under these conditions was detected using immunoprecipitation and mass spectrometry analyses. Sequestration of β1 integrin and MT1-MMP at the cell surface resulted in the formation of large invadopodia and local ECM degradation; however, the impaired internalization and recycling of MT1-MMP and β1 integrin ultimately led to a loss of invasive behaviour.
Collapse
Affiliation(s)
- Olivia R Grafinger
- Department of Molecular and Cellular Biology, University of Guelph, ON N1G 2W1, Canada
| | - Genya Gorshtein
- Department of Molecular and Cellular Biology, University of Guelph, ON N1G 2W1, Canada
| | - Tyler Stirling
- Department of Molecular and Cellular Biology, University of Guelph, ON N1G 2W1, Canada
| | | | - Marc G Coppolino
- Department of Molecular and Cellular Biology, University of Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
28
|
Decotret LR, Wadsworth BJ, Li LV, Lim CJ, Bennewith KL, Pallen CJ. Receptor-type protein tyrosine phosphatase alpha (PTPα) mediates MMP14 localization and facilitates triple-negative breast cancer cell invasion. Mol Biol Cell 2021; 32:567-578. [PMID: 33566639 PMCID: PMC8101463 DOI: 10.1091/mbc.e20-01-0060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The ability of cancer cells to invade surrounding tissues requires degradation of the extracellular matrix (ECM). Invasive structures, such as invadopodia, form on the plasma membranes of cancer cells and secrete ECM-degrading proteases that play crucial roles in cancer cell invasion. We have previously shown that the protein tyrosine phosphatase alpha (PTPα) regulates focal adhesion formation and migration of normal cells. Here we report a novel role for PTPα in promoting triple-negative breast cancer cell invasion in vitro and in vivo. We show that PTPα knockdown reduces ECM degradation and cellular invasion of MDA-MB-231 cells through Matrigel. PTPα is not a component of TKS5-positive structures resembling invadopodia; rather, PTPα localizes with endosomal structures positive for MMP14, caveolin-1, and early endosome antigen 1. Furthermore, PTPα regulates MMP14 localization to plasma membrane protrusions, suggesting a role for PTPα in intracellular trafficking of MMP14. Importantly, we show that orthotopic MDA-MB-231 tumors depleted in PTPα exhibit reduced invasion into the surrounding mammary fat pad. These findings suggest a novel role for PTPα in regulating the invasion of triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Lisa R Decotret
- Integrative Oncology, BC Cancer, Vancouver, British Columbia, BC V5Z 4E6, Canada.,Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, British Columbia, BC V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, BC V6H 3V4, Canada
| | - Brennan J Wadsworth
- Integrative Oncology, BC Cancer, Vancouver, British Columbia, BC V5Z 4E6, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, BC V6H 3V4, Canada
| | - Ling Vicky Li
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, British Columbia, BC V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, BC V6H 3V4, Canada
| | - Chinten J Lim
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, British Columbia, BC V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, BC V6H 3V4, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, BC V6H 3V4, Canada
| | - Kevin L Bennewith
- Integrative Oncology, BC Cancer, Vancouver, British Columbia, BC V5Z 4E6, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, BC V6H 3V4, Canada
| | - Catherine J Pallen
- Michael Cuccione Childhood Cancer Research Program, BC Children's Hospital Research Institute, Vancouver, British Columbia, BC V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, BC V6H 3V4, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, BC V6H 3V4, Canada
| |
Collapse
|
29
|
Niland S, Eble JA. Hold on or Cut? Integrin- and MMP-Mediated Cell-Matrix Interactions in the Tumor Microenvironment. Int J Mol Sci 2020; 22:ijms22010238. [PMID: 33379400 PMCID: PMC7794804 DOI: 10.3390/ijms22010238] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) has become the focus of interest in cancer research and treatment. It includes the extracellular matrix (ECM) and ECM-modifying enzymes that are secreted by cancer and neighboring cells. The ECM serves both to anchor the tumor cells embedded in it and as a means of communication between the various cellular and non-cellular components of the TME. The cells of the TME modify their surrounding cancer-characteristic ECM. This in turn provides feedback to them via cellular receptors, thereby regulating, together with cytokines and exosomes, differentiation processes as well as tumor progression and spread. Matrix remodeling is accomplished by altering the repertoire of ECM components and by biophysical changes in stiffness and tension caused by ECM-crosslinking and ECM-degrading enzymes, in particular matrix metalloproteinases (MMPs). These can degrade ECM barriers or, by partial proteolysis, release soluble ECM fragments called matrikines, which influence cells inside and outside the TME. This review examines the changes in the ECM of the TME and the interaction between cells and the ECM, with a particular focus on MMPs.
Collapse
|
30
|
Sharma P, Parveen S, Shah LV, Mukherjee M, Kalaidzidis Y, Kozielski AJ, Rosato R, Chang JC, Datta S. SNX27-retromer assembly recycles MT1-MMP to invadopodia and promotes breast cancer metastasis. J Cell Biol 2020; 219:132732. [PMID: 31820782 PMCID: PMC7039210 DOI: 10.1083/jcb.201812098] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 07/26/2019] [Accepted: 10/21/2019] [Indexed: 12/25/2022] Open
Abstract
Recycling of MT-MMPs to actin-rich membrane-protrusive structures promotes breast cancer invasion. This study shows that SNX27–retromer, an endosomal sorting and recycling machinery, interacts with MT1-MMP and regulates its transport to the cell surface, thus promoting matrix invasive activity of the breast cancer cells. A variety of metastatic cancer cells use actin-rich membrane protrusions, known as invadopodia, for efficient ECM degradation, which involves trafficking of proteases from intracellular compartments to these structures. Here, we demonstrate that in the metastatic breast cancer cell line MDA-MB-231, retromer regulates the matrix invasion activity by recycling matrix metalloprotease, MT1-MMP. We further found that MT2-MMP, another abundantly expressed metalloprotease, is also invadopodia associated. MT1- and MT2-MMP showed a high degree of colocalization but were located on the distinct endosomal domains. Retromer and its associated sorting nexin, SNX27, phenocopied each other in matrix degradation via selectively recycling MT1-MMP but not MT2-MMP. ITC-based studies revealed that both SNX27 and retromer could directly interact with MT1-MMP. Analysis from a publicly available database showed SNX27 to be overexpressed or frequently altered in the patients having invasive breast cancer. In xenograft-based studies, SNX27-depleted cell lines showed prolonged survival of SCID mice, suggesting a possible implication for overexpression of the sorting nexin in tumor samples.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Bhopal, India
| | - Sameena Parveen
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Bhopal, India
| | - Lekha V Shah
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Bhopal, India
| | - Madhumita Mukherjee
- Department of Chemistry, Indian Institute of Science Education and Research, Bhopal, Bhopal, India
| | - Yannis Kalaidzidis
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Faculty of Bioengineering and Bioinformatics, Moscow State University, Moscow, Russia
| | | | | | | | - Sunando Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Bhopal, India
| |
Collapse
|
31
|
Tagliatela AC, Hempstead SC, Hibshman PS, Hockenberry MA, Brighton HE, Pecot CV, Bear JE. Coronin 1C inhibits melanoma metastasis through regulation of MT1-MMP-containing extracellular vesicle secretion. Sci Rep 2020; 10:11958. [PMID: 32686704 PMCID: PMC7371684 DOI: 10.1038/s41598-020-67465-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
Coronin 1C is overexpressed in multiple tumors, leading to the widely held view that this gene drives tumor progression, but this hypothesis has not been rigorously tested in melanoma. Here, we combined a conditional knockout of Coronin 1C with a genetically engineered mouse model of PTEN/BRAF-driven melanoma. Loss of Coronin 1C in this model increases both primary tumor growth rates and distant metastases. Coronin 1C-null cells isolated from this model are more invasive in vitro and produce more metastatic lesions in orthotopic transplants than Coronin 1C-reexpressing cells due to the shedding of extracellular vesicles (EVs) containing MT1-MMP. Interestingly, these vesicles contain melanosome markers suggesting a melanoma-specific mechanism of EV release, regulated by Coronin 1C, that contributes to the high rates of metastasis in melanoma.
Collapse
Affiliation(s)
- Alicia C Tagliatela
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Stephanie C Hempstead
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Priya S Hibshman
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Max A Hockenberry
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hailey E Brighton
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chad V Pecot
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
32
|
Liu H, Xu J, Yao Q, Zhang Z, Guo Q, Lin J. Rab7 Is Associated with Poor Prognosis of Gastric Cancer and Promotes Proliferation, Invasion, and Migration of Gastric Cancer Cells. Med Sci Monit 2020; 26:e922217. [PMID: 32591494 PMCID: PMC7339976 DOI: 10.12659/msm.922217] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Rab7 belongs to the Ras oncogene family. Many studies have shown that its dysfunction is associated with many types of malignant tumors, but its effect on the pathogenesis of gastric cancer (GC) is still unknown. Therefore, we investigated the effect and mechanism of Rab7 in GC. MATERIAL AND METHODS The expression of Rab7 in GC and adjacent tissues was detected by immunohistochemistry, Western blot analysis, and qRT-PCR. The relationship of Rab7 with clinicopathological parameters and prognosis was analyzed. The expressions of Rab7, PI3K, and AKT in GC cells were assessed by Western blot. Overexpressed and silenced GC cell lines were constructed and AGS cells were treated with LY294002. The proliferation capacity of GC cells was detected by CCK8 assay, cell cycle changes were detected by flow cytometry, and the invasion and migration abilities of GC cells were assessed by transwell assay. RESULTS The expression of Rab7 was upregulated in the samples and cells, and was positively correlated with lymph node metastasis but negatively correlated with histological differentiation and clinical prognosis. In cell function experiments, overexpression of Rab7 induced the transition from S phase to G2 phase and promoted the proliferation, invasion, and migration of GC cells. Our assessment of the molecular mechanism showed that Rab7 promoted the phosphorylation of PI3K and AKT in GC cells. Incubation with the PI3K inhibitor Ly294002 impaired the enhanced effect of Rab7 overexpression on proliferation, migration, and invasion abilities of GC cells. These results show that the Rab7 affects GC cell progression by modulating the PI3K/AKT pathway. CONCLUSIONS Rab7 could be a prognostic biomarker and therapeutic target of the PI3K/AKT pathway in GC.
Collapse
Affiliation(s)
- Huiyong Liu
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China (mainland)
| | - Jie Xu
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China (mainland)
| | - Qingzhi Yao
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China (mainland)
| | - Zhongyi Zhang
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China (mainland)
| | - Qiaonan Guo
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China (mainland)
| | - Jianqing Lin
- Department of Surgical Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China (mainland)
| |
Collapse
|
33
|
Li Z, Dong K, Guo P, Tan Z, Zhang F, Tian Y, Lv H. Identification of Autophagy-Related Genes and Small-Molecule Drugs in Esophageal Carcinoma. Med Sci Monit 2020; 26:e921855. [PMID: 32415055 PMCID: PMC7247420 DOI: 10.12659/msm.921855] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Esophageal carcinoma (ESCA) is associated with a poor prognosis and high mortality rate. Autophagy plays important roles in promoting or suppressing tumor cell survival at different stages of cancer development. However, the roles of autophagy-related genes (ARGs) during ESCA progression and in patient prognosis remain unclear. Accordingly, in this study, we aimed to identify the relationships of ARGs with ESCA progression and patient prognosis. Material/Methods Clinicopathological information for patients with ESCA was downloaded from The Cancer Genome Atlas (TCGA) database. Transcriptome expression profiles were downloaded from TCGA and GTEx databases, and ARGs were downloaded from the Human Autophagy Database. We investigated the functions of ARGs by bioinformatics analysis. Moreover, statistical analysis of these genes was performed to identify independent prognostic markers. Results Differentially expressed genes between normal and tumor tissues were detected and identified. GO and KEGG analyses of differentially expressed ARGs were performed. Moreover, we derived a risk signature based on the identified independent prognostic markers. The identified genes also could predict the clinicopathological features of ESCA. Conclusions ARGs were key participants in the tumorigenesis and development of ESCA. Our findings may be useful for developing improved therapeutic approaches for ESCA.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Keqin Dong
- School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Peiyuan Guo
- School of Basic Medical Sciences, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Zirui Tan
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Fan Zhang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Yang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Huilai Lv
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| |
Collapse
|
34
|
Grafinger OR, Gorshtein G, Stirling T, Brasher MI, Coppolino MG. β1 integrin-mediated signaling regulates MT1-MMP phosphorylation to promote tumor cell invasion. J Cell Sci 2020; 133:jcs239152. [PMID: 32205364 DOI: 10.1242/jcs.239152] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/12/2020] [Indexed: 12/15/2022] Open
Abstract
Malignant cancer cells can invade extracellular matrix (ECM) through the formation of F-actin-rich subcellular structures termed invadopodia. ECM degradation at invadopodia is mediated by matrix metalloproteinases (MMPs), and recent findings indicate that membrane-anchored membrane type 1-matrix metalloproteinase (MT1-MMP, also known as MMP14) has a primary role in this process. Maintenance of an invasive phenotype is dependent on internalization of MT1-MMP from the plasma membrane and its recycling to sites of ECM remodeling. Internalization of MT1-MMP is dependent on its phosphorylation, and here we examine the role of β1 integrin-mediated signaling in this process. Activation of β1 integrin using the antibody P4G11 induced phosphorylation and internalization of MT1-MMP and resulted in increased cellular invasiveness and invadopodium formation in vitro We also observed phosphorylation of Src and epidermal growth factor receptor (EGFR) and an increase in their association in response to β1 integrin activation, and determined that Src and EGFR promote phosphorylation of MT1-MMP on Thr567 These results suggest that MT1-MMP phosphorylation is regulated by a β1 integrin-Src-EGFR signaling pathway that promotes recycling of MT1-MMP to sites of invadopodia formation during cancer cell invasion.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Olivia R Grafinger
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Genya Gorshtein
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Tyler Stirling
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Megan I Brasher
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Marc G Coppolino
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| |
Collapse
|
35
|
Suárez H, López-Martín S, Toribio V, Zamai M, Hernández-Riquer MV, Genís L, Arroyo AG, Yáñez-Mó M. Regulation of MT1-MMP Activity through Its Association with ERMs. Cells 2020; 9:cells9020348. [PMID: 32028690 PMCID: PMC7072721 DOI: 10.3390/cells9020348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/31/2020] [Accepted: 02/01/2020] [Indexed: 02/08/2023] Open
Abstract
Membrane-bound proteases play a key role in biology by degrading matrix proteins or shedding adhesion receptors. MT1-MMP metalloproteinase is critical during cancer invasion, angiogenesis, and development. MT1-MMP activity is strictly regulated by internalization, recycling, autoprocessing but also through its incorporation into tetraspanin-enriched microdomains (TEMs), into invadopodia, or by its secretion on extracellular vesicles (EVs). We identified a juxtamembrane positively charged cluster responsible for the interaction of MT1-MMP with ERM (ezrin/radixin/moesin) cytoskeletal connectors in breast carcinoma cells. Linkage to ERMs regulates MT1-MMP subcellular distribution and internalization, but not its incorporation into extracellular vesicles. MT1-MMP association to ERMs and insertion into TEMs are independent phenomena, so that mutation of the ERM-binding motif in the cytoplasmic region of MT1-MMP does not preclude its association with the tetraspanin CD151, but impairs the accumulation and coalescence of CD151/MT1-MMP complexes at actin-rich structures. Conversely, gene deletion of CD151 does not impact on MT1-MMP colocalization with ERM molecules. At the plasma membrane MT1-MMP autoprocessing is severely dependent on ERM association and seems to be the dominant regulator of the enzyme collagenolytic activity. This newly characterized MT1-MMP/ERM association can thus be of relevance for tumor cell invasion.
Collapse
Affiliation(s)
- Henar Suárez
- Molecular Biology Department, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (H.S.); (V.T.)
- Severo Ochoa Molecular Biology Center (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049 Madrid, Spain;
| | - Soraya López-Martín
- Severo Ochoa Molecular Biology Center (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049 Madrid, Spain;
| | - Víctor Toribio
- Molecular Biology Department, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (H.S.); (V.T.)
- Severo Ochoa Molecular Biology Center (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049 Madrid, Spain;
| | - Moreno Zamai
- Unit of Microscopy and Dynamic Imaging, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain;
| | - M. Victoria Hernández-Riquer
- Vascular Pathophysiology Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (M.V.H.-R.); (L.G.); (A.G.A.)
| | - Laura Genís
- Vascular Pathophysiology Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (M.V.H.-R.); (L.G.); (A.G.A.)
| | - Alicia G. Arroyo
- Vascular Pathophysiology Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (M.V.H.-R.); (L.G.); (A.G.A.)
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), 28040 Madrid, Spain
| | - María Yáñez-Mó
- Molecular Biology Department, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain; (H.S.); (V.T.)
- Severo Ochoa Molecular Biology Center (CBM-SO), Instituto de Investigación Sanitaria Princesa (IIS-IP), 28049 Madrid, Spain;
- Correspondence:
| |
Collapse
|
36
|
Solga R, Behrens J, Ziemann A, Riou A, Berwanger C, Becker L, Garrett L, de Angelis MH, Fischer L, Coras R, Barkovits K, Marcus K, Mahabir E, Eichinger L, Schröder R, Noegel AA, Clemen CS. CRN2 binds to TIMP4 and MMP14 and promotes perivascular invasion of glioblastoma cells. Eur J Cell Biol 2019; 98:151046. [PMID: 31677819 DOI: 10.1016/j.ejcb.2019.151046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/13/2019] [Accepted: 09/30/2019] [Indexed: 12/21/2022] Open
Abstract
CRN2 is an actin filament binding protein involved in the regulation of various cellular processes including cell migration and invasion. CRN2 has been implicated in the malignant progression of different types of human cancer. We used CRN2 knock-out mice for analyses as well as for crossbreeding with a Tp53/Pten knock-out glioblastoma mouse model. CRN2 knock-out mice were subjected to a phenotyping screen at the German Mouse Clinic. Murine glioblastoma tissue specimens as well as cultured murine brain slices and glioblastoma cell lines were investigated by immunohistochemistry, immunofluorescence, and cell biological experiments. Protein interactions were studied by immunoprecipitation, pull-down, and enzyme activity assays. CRN2 knock-out mice displayed neurological and behavioural alterations, e.g. reduced hearing sensitivity, reduced acoustic startle response, hypoactivity, and less frequent urination. While glioblastoma mice with or without the additional CRN2 knock-out allele exhibited no significant difference in their survival rates, the increased levels of CRN2 in transplanted glioblastoma cells caused a higher tumour cell encasement of murine brain slice capillaries. We identified two important factors of the tumour microenvironment, the tissue inhibitor of matrix metalloproteinase 4 (TIMP4) and the matrix metalloproteinase 14 (MMP14, synonym: MT1-MMP), as novel binding partners of CRN2. All three proteins mutually interacted and co-localised at the front of lamellipodia, and CRN2 was newly detected in exosomes. On the functional level, we demonstrate that CRN2 increased the secretion of TIMP4 as well as the catalytic activity of MMP14. Our results imply that CRN2 represents a pro-invasive effector within the tumour cell microenvironment of glioblastoma multiforme.
Collapse
Affiliation(s)
- Roxana Solga
- Centre for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany
| | - Juliane Behrens
- Centre for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany
| | - Anja Ziemann
- Centre for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany
| | - Adrien Riou
- In-vivo NMR, Max Planck Institute for Metabolism Research, 50931, Cologne, Germany
| | - Carolin Berwanger
- Centre for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany; Institute of Aerospace Medicine, German Aerospace Center (DLR), 51147, Cologne, Germany
| | - Lore Becker
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Centre Munich, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
| | - Lillian Garrett
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Centre Munich, German Research Centre for Environmental Health, 85764, Neuherberg, Germany; Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Centre Munich, German Research Centre for Environmental Health, 85764, Neuherberg, Germany; Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, 85354, Freising, Germany; German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Lisa Fischer
- Comparative Medicine, Center for Molecular Medicine Cologne, University of Cologne, 50931, Cologne, Germany
| | - Roland Coras
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Katalin Barkovits
- Medizinisches Proteom‑Center, Medical Faculty, Ruhr-University Bochum, 44801, Bochum, Germany
| | - Katrin Marcus
- Medizinisches Proteom‑Center, Medical Faculty, Ruhr-University Bochum, 44801, Bochum, Germany
| | - Esther Mahabir
- Comparative Medicine, Center for Molecular Medicine Cologne, University of Cologne, 50931, Cologne, Germany
| | - Ludwig Eichinger
- Centre for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Angelika A Noegel
- Centre for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany.
| | - Christoph S Clemen
- Centre for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, 50931, Cologne, Germany; Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054, Erlangen, Germany; Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Medical Faculty, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
37
|
Gonzalez-Molina J, Gramolelli S, Liao Z, Carlson JW, Ojala PM, Lehti K. MMP14 in Sarcoma: A Regulator of Tumor Microenvironment Communication in Connective Tissues. Cells 2019; 8:cells8090991. [PMID: 31466240 PMCID: PMC6770050 DOI: 10.3390/cells8090991] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/24/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Sarcomas are deadly malignant tumors of mesenchymal origin occurring at all ages. The expression and function of the membrane-type matrix metalloproteinase MMP14 is closely related to the mesenchymal cell phenotype, and it is highly expressed in most sarcomas. MMP14 regulates the activity of multiple extracellular and plasma membrane proteins, influencing cell–cell and cell–extracellular matrix (ECM) communication. This regulation mediates processes such as ECM degradation and remodeling, cell invasion, and cancer metastasis. Thus, a comprehensive understanding of the biology of MMP14 in sarcomas will shed light on the mechanisms controlling the key processes in these diseases. Here, we provide an overview of the function and regulation of MMP14 and we discuss their relationship with clinical and pre-clinical MMP14 data in both adult and childhood sarcomas.
Collapse
Affiliation(s)
- Jordi Gonzalez-Molina
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, 17177 Stockholm, Sweden.
- Department of Oncology-Pathology, Karolinska Institutet, 17176 Stockholm, Sweden.
| | - Silvia Gramolelli
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Zehuan Liao
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, 17177 Stockholm, Sweden
- School of Biological Sciences, Nanyang Technological University Singapore, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Joseph W Carlson
- Department of Oncology-Pathology, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Päivi M Ojala
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Section of Virology, Division of Infectious Diseases, Department of Medicine, Imperial College London, London W2 1NY, UK
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, 17177 Stockholm, Sweden.
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.
| |
Collapse
|
38
|
Hsu KS, Otsu W, Li Y, Wang HC, Chen S, Tsang SH, Chuang JZ, Sung CH. CLIC4 regulates late endosomal trafficking and matrix degradation activity of MMP14 at focal adhesions in RPE cells. Sci Rep 2019; 9:12247. [PMID: 31439888 PMCID: PMC6706427 DOI: 10.1038/s41598-019-48438-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/05/2019] [Indexed: 12/17/2022] Open
Abstract
Dysregulation in the extracellular matrix (ECM) microenvironment surrounding the retinal pigment epithelium (RPE) has been implicated in the etiology of proliferative vitreoretinopathy and age-related macular degeneration. The regulation of ECM remodeling by RPE cells is not well understood. We show that membrane-type matrix metalloproteinase 14 (MMP14) is central to ECM degradation at the focal adhesions in human ARPE19 cells. The matrix degradative activity, but not the assembly, of the focal adhesion is regulated by chloride intracellular channel 4 (CLIC4). CLIC4 is co-localized with MMP14 in the late endosome. CLIC4 regulates the proper sorting of MMP14 into the lumen of the late endosome and its proteolytic activation in lipid rafts. CLIC4 has the newly-identified “late domain” motif that binds to MMP14 and to Tsg101, a component of the endosomal sorting complex required for transport (ESCRT) complex. Unlike the late domain mutant CLIC4, wild-type CLIC4 can rescue the late endosomal sorting defect of MMP14. Finally, CLIC4 knockdown inhibits the apical secretion of MMP2 in polarized human RPE monolayers. These results, taken together, demonstrate that CLIC4 is a novel matrix microenvironment modulator and a novel regulator for late endosomal cargo sorting. Moreover, the late endosomal sorting of MMP14 actively regulates its surface activation in RPE cells.
Collapse
Affiliation(s)
- Kuo-Shun Hsu
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA.,Department of Surgery, Colorectal Service and Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wataru Otsu
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA.,Department of Biomedical Research Laboratory, Gifu Pharmaceutical University, Gifu, Japan
| | - Yao Li
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Heuy-Ching Wang
- Ocular Trauma Task Area, US Army Institute of Surgical Research, Joint Base San Antonio-Fort Sam Houston, TX, San Antonio, USA
| | - Shuibing Chen
- Department of Surgery and Department of Biochemistry, Weill Medical College of Cornell University, New York, NY, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.,Department of Pathology & Cell Biology, and Columbia Stem Cell Initiative, Columbia University Medical Center, New York, NY, USA
| | - Jen-Zen Chuang
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA
| | - Ching-Hwa Sung
- Department of Ophthalmology, Weill Medical College of Cornell University, New York, NY, USA. .,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY, USA.
| |
Collapse
|
39
|
Guerra F, Bucci C. Role of the RAB7 Protein in Tumor Progression and Cisplatin Chemoresistance. Cancers (Basel) 2019; 11:cancers11081096. [PMID: 31374919 PMCID: PMC6721790 DOI: 10.3390/cancers11081096] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022] Open
Abstract
RAB7 is a small guanosine triphosphatase (GTPase) extensively studied as regulator of vesicular trafficking. Indeed, its role is fundamental in several steps of the late endocytic pathway, including endosome maturation, transport from early endosomes to late endosomes and lysosomes, clustering and fusion of late endosomes and lysosomes in the perinuclear region and lysosomal biogenesis. Besides endocytosis, RAB7 is important for a number of other cellular processes among which, autophagy, apoptosis, signaling, and cell migration. Given the importance of RAB7 in these cellular processes, the interest to study the role of RAB7 in cancer progression is widely grown. Here, we describe the current understanding of oncogenic and oncosuppressor functions of RAB7 analyzing cellular context and other environmental factors in which it elicits pro and/or antitumorigenic effects. We also discuss the role of RAB7 in cisplatin resistance associated with its ability to regulate the late endosomal pathway, lysosomal biogenesis and extracellular vesicle secretion. Finally, we examined the potential cancer therapeutic strategies targeting the different molecular events in which RAB7 is involved.
Collapse
Affiliation(s)
- Flora Guerra
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy.
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy.
| |
Collapse
|
40
|
Röhl J, West ZE, Rudolph M, Zaharia A, Van Lonkhuyzen D, Hickey DK, Semmler ABT, Murray RZ. Invasion by activated macrophages requires delivery of nascent membrane-type-1 matrix metalloproteinase through late endosomes/lysosomes to the cell surface. Traffic 2019; 20:661-673. [PMID: 31297933 DOI: 10.1111/tra.12675] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022]
Abstract
Macrophage migration into injured or infected tissue is a key aspect in the pathophysiology of many diseases where inflammation is a driving factor. Membrane-type-1 matrix metalloproteinase (MT1-MMP) cleaves extracellular matrix components to facilitate invasion. Here we show that, unlike the constitutive MT1-MMP surface recycling seen in cancer cells, unactivated macrophages express low levels of MT1-MMP. Upon lipopolysaccharide (LPS) activation, MT1-MMP synthesis dramatically increases 10-fold at the surface by 15 hours. MT1-MMP is trafficked from the Golgi complex to the surface via late endosomes/lysosomes in a pathway regulated by the late endosome/lysosome R-SNAREs VAMP7 and VAMP8. These form two separate complexes with the surface Q-SNARE complex Stx4/SNAP23 to regulate MT1-MMP delivery to the plasma membrane. Loss of either one of these SNAREs leads to a reduction in surface MT1-MMP, gelatinase activity and reduced invasion. Thus, inhibiting MT1-MMP transport through this pathway could reduce macrophage migration and the resulting inflammation.
Collapse
Affiliation(s)
- Joan Röhl
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Zoe E West
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Maren Rudolph
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Andreea Zaharia
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Derek Van Lonkhuyzen
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Danica K Hickey
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Annalese B T Semmler
- Institute of Health and Biomedical Innovation, School of Clinical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Rachael Z Murray
- Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
41
|
Post-Translational Modification-Dependent Activity of Matrix Metalloproteinases. Int J Mol Sci 2019; 20:ijms20123077. [PMID: 31238509 PMCID: PMC6627178 DOI: 10.3390/ijms20123077] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 12/18/2022] Open
Abstract
Due to their capacity to process different proteins of the extracellular matrix (ECM), matrix metalloproteinases (MMPs) were initially described as a family of secreted proteases, functioning as main ECM regulators. However, through proteolytic processing of various biomolecules, MMPs also modulate intra- and extracellular pathways and networks. Thereby, they are functionally implicated in the regulation of multiple physiological and pathological processes. Consequently, MMP activity is tightly regulated through a combination of epigenetic, transcriptional, and post-transcriptional control of gene expression, proteolytic activation, post-translational modifications (PTMs), and extracellular inhibition. In addition, MMPs, their substrates and ECM binding partners are frequently modified by PTMs, which suggests an important role of PTMs in modulating the pleiotropic activities of these proteases. This review summarizes the recent progress towards understanding the role of PTMs (glycosylation, phosphorylation, glycosaminoglycans) on the activity of several members of the MMP family.
Collapse
|
42
|
Noll B, Benz D, Frey Y, Meyer F, Lauinger M, Eisler SA, Schmid S, Hordijk PL, Olayioye MA. DLC3 suppresses MT1-MMP-dependent matrix degradation by controlling RhoB and actin remodeling at endosomal membranes. J Cell Sci 2019; 132:jcs.223172. [PMID: 31076513 DOI: 10.1242/jcs.223172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/29/2019] [Indexed: 12/21/2022] Open
Abstract
Cancer cells degrade the extracellular matrix through actin-rich protrusions termed invadopodia. The formation of functional invadopodia requires polarized membrane trafficking driven by Rho GTPase-mediated cytoskeletal remodeling. We identify the Rho GTPase-activating protein deleted in liver cancer 3 (DLC3; also known as STARD8) as an integral component of the endosomal transport and sorting machinery. We provide evidence for the direct regulation of RhoB by DLC3 at endosomal membranes to which DLC3 is recruited by interacting with the sorting nexin SNX27. In TGF-β-treated MCF10A breast epithelial cells, DLC3 knockdown enhanced metalloproteinase-dependent matrix degradation, which was partially rescued by RhoB co-depletion. This was recapitulated in MDA-MB-231 breast cancer cells in which early endosomes demonstrated aberrantly enriched F-actin and accumulated the metalloproteinase MT1-MMP (also known as MMP14) upon DLC3 knockdown. Remarkably, Rab4 (herein referring to Rab4A) downregulation fully rescued the enhanced matrix degradation of TGF-β-treated MCF10A and MDA-MB-231 cells. In summary, our findings establish a novel role for DLC3 in the suppression of MT1-MMP-dependent matrix degradation by inactivating RhoB signaling at endosomal membranes. We propose that DLC3 function is required to limit endosomal actin polymerization, Rab4-dependent recycling of MT1-MMP and, consequently, matrix degradation mediated by invadopodial activity.
Collapse
Affiliation(s)
- Bettina Noll
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany.,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, 70569 Stuttgart, Germany
| | - David Benz
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Yannick Frey
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Florian Meyer
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Manuel Lauinger
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Stephan A Eisler
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany.,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, 70569 Stuttgart, Germany
| | - Simone Schmid
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | - Peter L Hordijk
- Department of Physiology, Amsterdam University Medical Center, VUmc, De Boelelaan 1118,1081 HV Amsterdam, The Netherlands
| | - Monilola A Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany .,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, 70569 Stuttgart, Germany
| |
Collapse
|
43
|
MT1-MMP-dependent cell migration: proteolytic and non-proteolytic mechanisms. Biochem Soc Trans 2019; 47:811-826. [PMID: 31064864 PMCID: PMC6599156 DOI: 10.1042/bst20180363] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/22/2019] [Accepted: 04/08/2019] [Indexed: 01/01/2023]
Abstract
Membrane-type 1 matrix metalloproteinase (MT1-MMP) is a type I transmembrane proteinase that belongs to the matrix metalloproteinase (MMP) family. It is a potent modifier of cellular microenvironment and promotes cell migration and invasion of a wide variety of cell types both in physiological and pathological conditions. It promotes cell migration by degrading extracellular matrix on the cell surface and creates a migration path, by modifying cell adhesion property by shedding cell adhesion molecules to increase cell motility, and by altering cellular metabolism. Thus, MT1-MMP is a multifunctional cell motility enhancer. In this review, we will discuss the current understanding of the proteolytic and non-proteolytic mechanism of MT1-MMP-dependent cell migration.
Collapse
|
44
|
Rab25 and RCP in cancer progression. Arch Pharm Res 2019; 42:101-112. [DOI: 10.1007/s12272-019-01129-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/29/2019] [Indexed: 01/10/2023]
|
45
|
The role of extracellular vesicles in cancer microenvironment and metastasis: myths and challenges. Biochem Soc Trans 2019; 47:273-280. [PMID: 30647137 DOI: 10.1042/bst20180253] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/04/2018] [Accepted: 12/14/2018] [Indexed: 12/16/2022]
Abstract
The concept of vesicles or cell debris released by cancer cells to promote metastasis is not new, but the mechanisms used to currently ascribe their impact in metastasis are of intense debate. A significant increase in reports describing the role of cancer-derived EVs in cancer metastasis has been followed by a growing amount of uncertainty behind these claims. This review will delve into the role of EVs in promoting cancer metastasis by relying on a balanced perspective that looks at challenges faced previously by extracellular vesicle biologists, current technical limitations in the field, and overlooked physiologic mechanisms that may play a confounding role. This review will also discuss how certain experimental approaches are misleading which ultimately lead to overly optimistic mechanisms that have minimally contributed to the pathophysiology of metastasis.
Collapse
|
46
|
Dingjan I, Linders PTA, Verboogen DRJ, Revelo NH, Ter Beest M, van den Bogaart G. Endosomal and Phagosomal SNAREs. Physiol Rev 2018; 98:1465-1492. [PMID: 29790818 DOI: 10.1152/physrev.00037.2017] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein family is of vital importance for organelle communication. The complexing of cognate SNARE members present in both the donor and target organellar membranes drives the membrane fusion required for intracellular transport. In the endocytic route, SNARE proteins mediate trafficking between endosomes and phagosomes with other endosomes, lysosomes, the Golgi apparatus, the plasma membrane, and the endoplasmic reticulum. The goal of this review is to provide an overview of the SNAREs involved in endosomal and phagosomal trafficking. Of the 38 SNAREs present in humans, 30 have been identified at endosomes and/or phagosomes. Many of these SNAREs are targeted by viruses and intracellular pathogens, which thereby reroute intracellular transport for gaining access to nutrients, preventing their degradation, and avoiding their detection by the immune system. A fascinating picture is emerging of a complex transport network with multiple SNAREs being involved in consecutive trafficking routes.
Collapse
Affiliation(s)
- Ilse Dingjan
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Peter T A Linders
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Danielle R J Verboogen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Natalia H Revelo
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Martin Ter Beest
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen , The Netherlands ; and Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen , Groningen , The Netherlands
| |
Collapse
|
47
|
Verraes A, Cholley B, Galli T, Nola S. Comparative study of commercially available and homemade anti-VAMP7 antibodies using CRISPR/Cas9-depleted HeLa cells and VAMP7 knockout mice. F1000Res 2018; 7:1649. [PMID: 30815249 PMCID: PMC6376254 DOI: 10.12688/f1000research.15707.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2018] [Indexed: 10/06/2023] Open
Abstract
VAMP7 (vesicle-associated membrane protein) belongs to the intracellular membrane fusion SNARE (Soluble N-ethylmaleimide-sensitive factor attachment protein receptors) protein family. In this study, we used CRISPR/Cas9 genome editing technology to generate VAMP7 knockout (KO) human HeLa cells and mouse KO brain extracts in order to test the specificity and the background of a set of commercially available and homemade anti-VAMP7 antibodies. We propose a simple profiling method to analyze western blotting and immunocytochemistry staining profiles and determine the extent of the antibodies' specificity. Using this method, we were able to rank the performance of a set of available antibodies and further showed an optimized procedure for VAMP7 immunoprecipitation, which we validated using wild-type and KO mouse brain extracts.
Collapse
Affiliation(s)
- Agathe Verraes
- Membrane Traffic in Health and Disease, INSERM ERL U950, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, CNRS UMR 7592, Paris, France, 75013, France
| | - Beatrice Cholley
- Membrane Traffic in Healthy & Diseased Brain, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Descartes, Sorbonne Paris-Cité, 75014, France
| | - Thierry Galli
- Membrane Traffic in Health and Disease, INSERM ERL U950, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, CNRS UMR 7592, Paris, France, 75013, France
- Membrane Traffic in Healthy & Diseased Brain, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Descartes, Sorbonne Paris-Cité, 75014, France
| | - Sebastien Nola
- Membrane Traffic in Health and Disease, INSERM ERL U950, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, CNRS UMR 7592, Paris, France, 75013, France
- Membrane Traffic in Healthy & Diseased Brain, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Descartes, Sorbonne Paris-Cité, 75014, France
| |
Collapse
|
48
|
Verraes A, Cholley B, Galli T, Nola S. Comparative study of commercially available and homemade anti-VAMP7 antibodies using CRISPR/Cas9-depleted HeLa cells and VAMP7 knockout mice. F1000Res 2018; 7:1649. [PMID: 30815249 PMCID: PMC6376254 DOI: 10.12688/f1000research.15707.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2019] [Indexed: 11/30/2022] Open
Abstract
VAMP7 (vesicle-associated membrane protein) belongs to the intracellular membrane fusion SNARE (Soluble N-ethylmaleimide-sensitive factor attachment protein receptors) protein family. In this study, we used CRISPR/Cas9 genome editing technology to generate VAMP7 knockout (KO) human HeLa cells and mouse KO brain extracts in order to test the specificity and the background of a set of commercially available and homemade anti-VAMP7 antibodies. We propose a simple profiling method to analyze western blotting and use visual scoring for immunocytochemistry staining to determine the extent of the antibodies' specificity. Thus, we were able to rank the performance of a set of available antibodies and further showed an optimized procedure for VAMP7 immunoprecipitation, which we validated using wild-type and KO mouse brain extracts.
Collapse
Affiliation(s)
- Agathe Verraes
- Membrane Traffic in Health and Disease, INSERM ERL U950, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, CNRS UMR 7592, Paris, France, 75013, France
| | - Beatrice Cholley
- Membrane Traffic in Healthy & Diseased Brain, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Descartes, Sorbonne Paris-Cité, 75014, France
| | - Thierry Galli
- Membrane Traffic in Health and Disease, INSERM ERL U950, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, CNRS UMR 7592, Paris, France, 75013, France
- Membrane Traffic in Healthy & Diseased Brain, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Descartes, Sorbonne Paris-Cité, 75014, France
| | - Sebastien Nola
- Membrane Traffic in Health and Disease, INSERM ERL U950, Univ Paris Diderot, Sorbonne Paris Cité, Institut Jacques Monod, CNRS UMR 7592, Paris, France, 75013, France
- Membrane Traffic in Healthy & Diseased Brain, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université Paris Descartes, Sorbonne Paris-Cité, 75014, France
| |
Collapse
|
49
|
Planchon D, Rios Morris E, Genest M, Comunale F, Vacher S, Bièche I, Denisov EV, Tashireva LA, Perelmuter VM, Linder S, Chavrier P, Bodin S, Gauthier-Rouvière C. MT1-MMP targeting to endolysosomes is mediated by upregulation of flotillins. J Cell Sci 2018; 131:jcs.218925. [PMID: 30111578 DOI: 10.1242/jcs.218925] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/21/2018] [Indexed: 12/31/2022] Open
Abstract
Tumor cell invasion and metastasis formation are the major cause of death in cancer patients. These processes rely on extracellular matrix (ECM) degradation mediated by organelles termed invadopodia, to which the transmembrane matrix metalloproteinase MT1-MMP (also known as MMP14) is delivered from its reservoir, the RAB7-containing endolysosomes. How MT1-MMP is targeted to endolysosomes remains to be elucidated. Flotillin-1 and -2 are upregulated in many invasive cancers. Here, we show that flotillin upregulation triggers a general mechanism, common to carcinoma and sarcoma, which promotes RAB5-dependent MT1-MMP endocytosis and its delivery to RAB7-positive endolysosomal reservoirs. Conversely, flotillin knockdown in invasive cancer cells greatly reduces MT1-MMP accumulation in endolysosomes, its subsequent exocytosis at invadopodia, ECM degradation and cell invasion. Our results demonstrate that flotillin upregulation is necessary and sufficient to promote epithelial and mesenchymal cancer cell invasion and ECM degradation by controlling MT1-MMP endocytosis and delivery to the endolysosomal recycling compartment.
Collapse
Affiliation(s)
- Damien Planchon
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Eduardo Rios Morris
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Mallory Genest
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Franck Comunale
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Sophie Vacher
- Department of Genetics, Institut Curie, 75005 Paris, France
| | - Ivan Bièche
- Department of Genetics, Institut Curie, 75005 Paris, France
| | - Evgeny V Denisov
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk 634050, Russia.,Tomsk State University, Tomsk 634050, Russia
| | - Lubov A Tashireva
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk 634050, Russia
| | - Vladimir M Perelmuter
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk 634050, Russia
| | - Stefan Linder
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, University Medical Center Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Philippe Chavrier
- Cell Dynamics and Compartmentalization Unit, Institut Curie, 75005 Paris, France
| | - Stéphane Bodin
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | | |
Collapse
|
50
|
VAMP8, a vesicle-SNARE required for RAB37-mediated exocytosis, possesses a tumor metastasis suppressor function. Cancer Lett 2018; 437:79-88. [PMID: 30165196 DOI: 10.1016/j.canlet.2018.08.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 08/20/2018] [Indexed: 12/23/2022]
Abstract
We previously identified a metastasis suppressor RAB37 small GTPase that regulated exocytosis of tissue inhibitor of metalloproteinases 1 (TIMP1) to suppress lung cancer metastasis. Here, we show that vesicle-associated membrane protein 8 (VAMP8), a v-SNARE (vesicle soluble N-ethylmaleimide-sensitive factor activating protein receptor), interacts with RAB37 and drives the secretion of TIMP1 to inhibit tumor metastases. Confocal and total internal reflection fluorescence microscopic images demonstrated that VAMP8 co-localized with RAB37 and facilitated trafficking of RAB37-TIMP1 vesicles. Reconstitution experiments using tail-vein injection and lung-to-lung metastasis in mice showed that VAMP8 was essential for RAB37-regulated vesicle trafficking of TIMP1 to suppress cancer metastasis. Lung cancer patients with low VAMP8 showed distant metastasis, poor overall survival and progression-free survival. Importantly, multivariate Cox regression analysis indicated that patients with low VAMP8/low RAB37 expression profile showed significantly high risk of death (hazard ratio = 3.42, P < 0.001) even after adjusting for tumor metastasis parameter. Our findings reveal that VAMP8 is a novel v-SNARE crucial for RAB37-mediated exocytic transport of TIMP1 to suppress lung tumor metastasis. VAMP8 possesses a tumor metastasis suppressor function with a prognostic value in lung cancer.
Collapse
|