1
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [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: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
2
|
Joseph J, Mathew J, Alexander J. Scaffold Proteins in Autoimmune Disorders. Curr Rheumatol Rev 2024; 20:14-26. [PMID: 37670692 DOI: 10.2174/1573397119666230904151024] [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: 04/03/2023] [Revised: 06/26/2023] [Accepted: 08/10/2023] [Indexed: 09/07/2023]
Abstract
Cells transmit information to the external environment and within themselves through signaling molecules that modulate cellular activities. Aberrant cell signaling disturbs cellular homeostasis causing a number of different diseases, including autoimmunity. Scaffold proteins, as the name suggests, serve as the anchor for binding and stabilizing signaling proteins at a particular locale, allowing both intra and intercellular signal amplification and effective signal transmission. Scaffold proteins play a critical role in the functioning of tight junctions present at the intersection of two cells. In addition, they also participate in cleavage formation during cytokinesis, and in the organization of neural synapses, and modulate receptor management outcomes. In autoimmune settings such as lupus, scaffold proteins can lower the cell activation threshold resulting in uncontrolled signaling and hyperactivity. Scaffold proteins, through their binding domains, mediate protein- protein interaction and play numerous roles in cellular communication and homeostasis. This review presents an overview of scaffold proteins, their influence on the different signaling pathways, and their role in the pathogenesis of autoimmune and auto inflammatory diseases. Since these proteins participate in many roles and interact with several other signaling pathways, it is necessary to gain a thorough understanding of these proteins and their nuances to facilitate effective target identification and therapeutic design for the treatment of autoimmune disorders.
Collapse
Affiliation(s)
- Josna Joseph
- Department of Clinical Immunology & Rheumatology, CMC Vellore, Tamil Nadu, India
| | - John Mathew
- Department of Clinical Immunology & Rheumatology, CMC Vellore, Tamil Nadu, India
| | - Jessy Alexander
- Department of Medicine, Jacobs School of Medicine & Biomedical Sciences, University of Buffalo, New York, USA
| |
Collapse
|
3
|
Okletey J, Angelis D, Jones TM, Montagna C, Spiliotis ET. An oncogenic isoform of septin 9 promotes the formation of juxtanuclear invadopodia by reducing nuclear deformability. Cell Rep 2023; 42:112893. [PMID: 37516960 PMCID: PMC10530659 DOI: 10.1016/j.celrep.2023.112893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/17/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
Invadopodia are extracellular matrix (ECM) degrading structures, which promote cancer cell invasion. The nucleus is increasingly viewed as a mechanosensory organelle that determines migratory strategies. However, how the nucleus crosstalks with invadopodia is little known. Here, we report that the oncogenic septin 9 isoform 1 (SEPT9_i1) is a component of breast cancer invadopodia. SEPT9_i1 depletion diminishes invadopodium formation and the clustering of the invadopodium precursor components TKS5 and cortactin. This phenotype is characterized by deformed nuclei and nuclear envelopes with folds and grooves. We show that SEPT9_i1 localizes to the nuclear envelope and juxtanuclear invadopodia. Moreover, exogenous lamin A rescues nuclear morphology and juxtanuclear TKS5 clusters. Importantly, SEPT9_i1 is required for the amplification of juxtanuclear invadopodia, which is induced by the epidermal growth factor. We posit that nuclei of low deformability favor the formation of juxtanuclear invadopodia in a SEPT9_i1-dependent manner, which functions as a tunable mechanism for overcoming ECM impenetrability.
Collapse
Affiliation(s)
- Joshua Okletey
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Dimitrios Angelis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Tia M Jones
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Cristina Montagna
- Department of Radiology and Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
4
|
Petsana M, Roumia AF, Bagos PG, Boleti H, Braliou GG. In Silico Identification and Analysis of Proteins Containing the Phox Homology Phosphoinositide-Binding Domain in Kinetoplastea Protists: Evolutionary Conservation and Uniqueness of Phox-Homology-Domain-Containing Protein Architectures. Int J Mol Sci 2023; 24:11521. [PMID: 37511280 PMCID: PMC10380299 DOI: 10.3390/ijms241411521] [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: 05/29/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Kinetoplastea are free living and parasitic protists with unique features among Eukaryota. Pathogenic Kinetoplastea parasites (i.e., Trypanosoma and Leishmania spp.) undergo several developmental transitions essential for survival in their hosts. These transitions require membrane and cytoskeleton reorganizations that involve phosphoinositides (PIs). Phospholipids like PIs are key regulators of vital functions in all eukaryotes including signal transduction, protein transport and sorting, membrane trafficking, and cytoskeleton and membrane remodeling. A large repertoire of PI-metabolizing enzymes and PI-binding proteins/effectors carrying distinct PI-binding modules like the PX (phox homology) module could play significant roles in the life and virulence of pathogenic Kinetoplastea. The aim of this study was to retrieve the entire spectrum of Kinetoplastea protein sequences containing the PX module (PX-proteins), predict their structures, and identify in them evolutionary conserved and unique traits. Using a large array of bioinformatics tools, protein IDs from two searches (based on PFam's pHMM for PX domain (PF00787)) were combined, aligned, and utilized for the construction of a new Kinetoplastea_PX pHMM. This three-step search retrieved 170 PX-protein sequences. Structural domain configuration analysis identified PX, Pkinase, Lipocalin_5, and Vps5/BAR3-WASP domains and clustered them into five distinct subfamilies. Phylogenetic tree and domain architecture analysis showed that some domain architectures exist in proteomes of all Kinetoplastea spp., while others are genus-specific. Finally, amino acid conservation logos of the Kinetoplastea spp. and Homo sapiens PX domains revealed high evolutionary conservation in residues forming the critical structural motifs for PtdIns3P recognition. This study highlights the PX-Pkinase domain architecture as unique within Trypanosoma spp. and forms the basis for a targeted functional analysis of Kinetoplastea PX-proteins as putative targets for a rational design of anti-parasitic drugs.
Collapse
Affiliation(s)
- Marina Petsana
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 2-4 Papasiopoulou Str., 35131 Lamia, Greece
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Ahmed F Roumia
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 2-4 Papasiopoulou Str., 35131 Lamia, Greece
- Department of Agricultural Biochemistry, Faculty of Agriculture, Menoufia University, Shibin El-Kom 32514, Egypt
| | - Pantelis G Bagos
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 2-4 Papasiopoulou Str., 35131 Lamia, Greece
| | - Haralabia Boleti
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Georgia G Braliou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 2-4 Papasiopoulou Str., 35131 Lamia, Greece
| |
Collapse
|
5
|
Okletey J, Angelis D, Jones TM, Montagna C, Spiliotis ET. An oncogenic isoform of septin 9 promotes the formation of juxtanuclear invadopodia by reducing nuclear deformability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.18.545473. [PMID: 37398172 PMCID: PMC10312791 DOI: 10.1101/2023.06.18.545473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Invadopodia are extracellular matrix (ECM) degrading structures, which promote cancer cell invasion. The nucleus is increasingly viewed as a mechanosensory organelle that determines migratory strategies. However, how the nucleus crosstalks with invadopodia is little known. Here, we report that the oncogenic septin 9 isoform 1 (SEPT9_i1) is a component of breast cancer invadopodia. SEPT9_i1 depletion diminishes invadopodia formation and the clustering of invadopodia precursor components TKS5 and cortactin. This phenotype is characterized by deformed nuclei, and nuclear envelopes with folds and grooves. We show that SEPT9_i1 localizes to the nuclear envelope and juxtanuclear invadopodia. Moreover, exogenous lamin A rescues nuclear morphology and juxtanuclear TKS5 clusters. Importantly, SEPT9_i1 is required for the amplification of juxtanuclear invadopodia, which is induced by the epidermal growth factor. We posit that nuclei of low deformability favor the formation of juxtanuclear invadopodia in a SEPT9_i1-dependent manner, which functions as a tunable mechanism for overcoming ECM impenetrability. Highlights The oncogenic SEPT9_i1 is enriched in breast cancer invadopodia in 2D and 3D ECMSEPT9_i1 promotes invadopodia precursor clustering and invadopodia elongationSEPT9_i1 localizes to the nuclear envelope and reduces nuclear deformabilitySEPT9_i1 is required for EGF-induced amplification of juxtanuclear invadopodia. eTOC Blurb Invadopodia promote the invasion of metastatic cancers. The nucleus is a mechanosensory organelle that determines migratory strategies, but how it crosstalks with invadopodia is unknown. Okletey et al show that the oncogenic isoform SEPT9_i1 promotes nuclear envelope stability and the formation of invadopodia at juxtanuclear areas of the plasma membrane.
Collapse
|
6
|
Kato K, Miyazawa H, Kawashiri S, Lambert DW. Tumour: Fibroblast Interactions Promote Invadopodia-Mediated Migration and Invasion in Oral Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:5277440. [PMID: 36471888 PMCID: PMC9719419 DOI: 10.1155/2022/5277440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/04/2022] [Accepted: 10/28/2022] [Indexed: 08/08/2023]
Abstract
OBJECTIVES In the progression of cancer, interactions between cancer cells and cancer-associated fibroblasts (CAFs) play important roles. Cancer cell invasion is facilitated by filamentous actin (F-actin)-rich membrane protrusions called invadopodia, and the relationship between CAFs and invadopodia has been unclear. We used oral squamous cell carcinoma (OSCC) to investigate CAFs' effects on the formation of invadopodia, and we assessed the expressions of invadopodia markers and CAF markers ex vivo and their relationship with clinical parameters and survival. MATERIALS AND METHODS We examined the effect of culture with normal oral fibroblast (NOF)-derived and CAF-derived conditioned medium on the migration and invasion of two OSCC-derived cell lines using Transwells in the absence/presence of Matrigel. Immunoblotting and immunocytochemistry were conducted to assess the expressions of the invadopodia markers tyrosine kinase substrate 5 (Tks5) and membrane type 1 matrix metalloproteinase (MT1-MMP). We also used immunohistochemistry to examine patients with OSCC for an evaluation of the relationship between the CAF marker alpha smooth muscle actin (αSMA) and the expression of Tks5. The patients' survival was also assessed. RESULTS Compared to the use of culture medium alone, NOF-CM and CAF-CM both significantly increased the OSCC cells' migration and invasion (p < 0.05), and they significantly increased the expressions of both Tks5 and MT1-MMP. After the depletion of Tks5, the OSCC cells' migration and invasion abilities decreased. The expression of Tks5 and that of αSMA were correlated with poor survival, and a high expression of both markers was associated with an especially poor prognosis. CONCLUSIONS These results indicate that the formation of invadopodia is (i) important for OSCC cells' migration and invasion and (ii) regulated by the interaction of OSCC cells and stromal fibroblasts.
Collapse
Affiliation(s)
- Koroku Kato
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, 13-1 Takara Machi, Kanazawa 9208641, Japan
| | - Hiroki Miyazawa
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, 13-1 Takara Machi, Kanazawa 9208641, Japan
| | - Shuichi Kawashiri
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, 13-1 Takara Machi, Kanazawa 9208641, Japan
| | - Daniel W. Lambert
- School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK
| |
Collapse
|
7
|
Wang W, Zheng X, Azoitei A, John A, Zengerling F, Wezel F, Bolenz C, Günes C. The Role of TKS5 in Chromosome Stability and Bladder Cancer Progression. Int J Mol Sci 2022; 23:ijms232214283. [PMID: 36430759 PMCID: PMC9698602 DOI: 10.3390/ijms232214283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/04/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
TKS5 promotes invasion and migration through the formation of invadopodia in some tumour cells, and it also has an important physiological function in cell migration through podosome formation in various nontumour cells. To date, the role of TKS5 in urothelial cells, and its potential role in BC initiation and progression, has not yet been addressed. Moreover, the contribution of TKS5 to ploidy control and chromosome stability has not been reported in previous studies. Therefore, in the present study, we wished to address the following questions: (i) Is TKS5 involved in the ploidy control of urothelial cells? (ii) What is the mechanism that leads to aneuploidy in response to TKS5 knockdown? (iii) Is TKS5 an oncogene or tumour-suppressor gene in the context of BC? (iv) Does TKS5 affect the proliferation, migration and invasion of BC cells? We assessed the gene and protein expressions via qPCR and Western blot analyses in a set of nontumour cell strains (Y235T, HBLAK and UROtsa) and a set of BC cell lines (RT4, T24, UMUC3 and J82). Following the shRNA knockdown in the TKS5-proficient cells and the ectopic TKS5 expression in the cell lines with low/absent TKS5 expression, we performed functional experiments, such as metaphase, invadopodia and gelatine degradation assays. Moreover, we determined the invasion and migration abilities of these genetically modified cells by using the Boyden chamber and wound-healing assays. The TKS5 expression was lower in the bladder cancer cell lines with higher invasive capacities (T24, UMUC3 and J82) compared to the nontumour cell lines from human ureter (Y235T, HBLAK and UROtsa) and the noninvasive BC cell line RT4. The reduced TKS5 expression in the Y235T cells resulted in augmented aneuploidy and impaired cell division. According to the Boyden chamber and wound-healing assays, TKS5 promotes the invasion and migration of bladder cancer cells. According to the present study, TKS5 regulates the migration and invasion processes of bladder cancer (BC) cell lines and plays an important role in genome stability.
Collapse
|
8
|
Weber K, Hey S, Cervero P, Linder S. The circle of life: Phases of podosome formation, turnover and reemergence. Eur J Cell Biol 2022; 101:151218. [PMID: 35334303 DOI: 10.1016/j.ejcb.2022.151218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 01/06/2023] Open
Abstract
Podosomes are highly dynamic actin-rich structures in a variety of cell types, especially monocytic cells. They fulfill multiple functions such as adhesion, mechanosensing, or extracellular matrix degradation, thus allowing cells to detect and respond to a changing environment. These abilities are based on an intricate architecture that enables podosomes to sense mechanical properties of their substratum and to transduce them intracellularly in order to generate an appropriate cellular response. These processes are enabled through the tightly orchestrated interplay of more than 300 different components that are dynamically recruited during podosome formation and turnover. In this review, we discuss the different phases of the podosome life cycle and the current knowledge on regulatory factors that impact on the genesis, activity, dissolution and reemergence of podosomes. We also highlight mechanoregulatory processes that become important during these different stages, on the level of individual podosomes, and also at podosome sub- and superstructures.
Collapse
Affiliation(s)
- Kathrin Weber
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany
| | - Sven Hey
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany
| | - Pasquale Cervero
- 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
|
9
|
Merő B, Koprivanacz K, Cserkaszky A, Radnai L, Vas V, Kudlik G, Gógl G, Sok P, Póti ÁL, Szeder B, Nyitray L, Reményi A, Geiszt M, Buday L. Characterization of the Intramolecular Interactions and Regulatory Mechanisms of the Scaffold Protein Tks4. Int J Mol Sci 2021; 22:ijms22158103. [PMID: 34360869 PMCID: PMC8348221 DOI: 10.3390/ijms22158103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
The scaffold protein Tks4 is a member of the p47phox-related organizer superfamily. It plays a key role in cell motility by being essential for the formation of podosomes and invadopodia. In addition, Tks4 is involved in the epidermal growth factor (EGF) signaling pathway, in which EGF induces the translocation of Tks4 from the cytoplasm to the plasma membrane. The evolutionarily-related protein p47phox and Tks4 share many similarities in their N-terminal region: a phosphoinositide-binding PX domain is followed by two SH3 domains (so called “tandem SH3”) and a proline-rich region (PRR). In p47phox, the PRR is followed by a relatively short, disordered C-terminal tail region containing multiple phosphorylation sites. These play a key role in the regulation of the protein. In Tks4, the PRR is followed by a third and a fourth SH3 domain connected by a long (~420 residues) unstructured region. In p47phox, the tandem SH3 domain binds the PRR while the first SH3 domain interacts with the PX domain, thereby preventing its binding to the membrane. Based on the conserved structural features of p47phox and Tks4 and the fact that an intramolecular interaction between the third SH3 and the PX domains of Tks4 has already been reported, we hypothesized that Tks4 is similarly regulated by autoinhibition. In this study, we showed, via fluorescence-based titrations, MST, ITC, and SAXS measurements, that the tandem SH3 domain of Tks4 binds the PRR and that the PX domain interacts with the third SH3 domain. We also investigated a phosphomimicking Thr-to-Glu point mutation in the PRR as a possible regulator of intramolecular interactions. Phosphatidylinositol-3-phosphate (PtdIns(3)P) was identified as the main binding partner of the PX domain via lipid-binding assays. In truncated Tks4 fragments, the presence of the tandem SH3, together with the PRR, reduced PtdIns(3)P binding, while the presence of the third SH3 domain led to complete inhibition.
Collapse
Affiliation(s)
- Balázs Merő
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Kitti Koprivanacz
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Anna Cserkaszky
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - László Radnai
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Virag Vas
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Gyöngyi Kudlik
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - Gergő Gógl
- Department of Biochemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (G.G.); (L.N.)
| | - Péter Sok
- Research Centre for Natural Sciences, Institute of Organic Chemistry, 1117 Budapest, Hungary; (P.S.); (Á.L.P.); (A.R.)
| | - Ádám L. Póti
- Research Centre for Natural Sciences, Institute of Organic Chemistry, 1117 Budapest, Hungary; (P.S.); (Á.L.P.); (A.R.)
| | - Bálint Szeder
- Research Centre for Natural Sciences, Institute of Enzymology, 1117 Budapest, Hungary; (B.M.); (K.K.); (A.C.); (L.R.); (V.V.); (G.K.); (B.S.)
| | - László Nyitray
- Department of Biochemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (G.G.); (L.N.)
| | - Attila Reményi
- Research Centre for Natural Sciences, Institute of Organic Chemistry, 1117 Budapest, Hungary; (P.S.); (Á.L.P.); (A.R.)
| | - Miklós Geiszt
- Department of Physiology, Semmelweis University, 1094 Budapest, Hungary;
| | - László Buday
- Department of Biochemistry, Eötvös Loránd University, 1117 Budapest, Hungary; (G.G.); (L.N.)
- Department of Molecular Biology, Semmelweis University Medical School, 1094 Budapest, Hungary
- Correspondence:
| |
Collapse
|
10
|
Vermot A, Petit-Härtlein I, Smith SME, Fieschi F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants (Basel) 2021; 10:890. [PMID: 34205998 PMCID: PMC8228183 DOI: 10.3390/antiox10060890] [Citation(s) in RCA: 297] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 01/17/2023] Open
Abstract
The reactive oxygen species (ROS)-producing enzyme NADPH oxidase (NOX) was first identified in the membrane of phagocytic cells. For many years, its only known role was in immune defense, where its ROS production leads to the destruction of pathogens by the immune cells. NOX from phagocytes catalyzes, via one-electron trans-membrane transfer to molecular oxygen, the production of the superoxide anion. Over the years, six human homologs of the catalytic subunit of the phagocyte NADPH oxidase were found: NOX1, NOX3, NOX4, NOX5, DUOX1, and DUOX2. Together with the NOX2/gp91phox component present in the phagocyte NADPH oxidase assembly itself, the homologs are now referred to as the NOX family of NADPH oxidases. NOX are complex multidomain proteins with varying requirements for assembly with combinations of other proteins for activity. The recent structural insights acquired on both prokaryotic and eukaryotic NOX open new perspectives for the understanding of the molecular mechanisms inherent to NOX regulation and ROS production (superoxide or hydrogen peroxide). This new structural information will certainly inform new investigations of human disease. As specialized ROS producers, NOX enzymes participate in numerous crucial physiological processes, including host defense, the post-translational processing of proteins, cellular signaling, regulation of gene expression, and cell differentiation. These diversities of physiological context will be discussed in this review. We also discuss NOX misregulation, which can contribute to a wide range of severe pathologies, such as atherosclerosis, hypertension, diabetic nephropathy, lung fibrosis, cancer, or neurodegenerative diseases, giving this family of membrane proteins a strong therapeutic interest.
Collapse
Affiliation(s)
- Annelise Vermot
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Isabelle Petit-Härtlein
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| | - Susan M. E. Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA 30144, USA;
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France; (A.V.); (I.P.-H.)
| |
Collapse
|
11
|
Abstract
Endosome-to-cell surface recycling is mediated by retromer and Snx27. In this issue, Mao et al. (2021. J. Cell Biol.https://doi.org/10.1083/jcb.202010048) detail how endosomal protein sorting responds to external stimuli and reveal that phosphorylation of Snx27 regulates its cargo-binding function resulting in reduced endosome-to-cell surface recycling.
Collapse
Affiliation(s)
- Matthew N.J. Seaman
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
12
|
Iizuka S, Quintavalle M, Navarro JC, Gribbin KP, Ardecky RJ, Abelman MM, Ma CT, Sergienko E, Zeng FY, Pass I, Thomas GV, McWeeney SK, Hassig CA, Pinkerton AB, Courtneidge SA. Serine-Threonine Kinase TAO3-Mediated Trafficking of Endosomes Containing the Invadopodia Scaffold TKS5α Promotes Cancer Invasion and Tumor Growth. Cancer Res 2021; 81:1472-1485. [PMID: 33414172 DOI: 10.1158/0008-5472.can-20-2383] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/13/2020] [Accepted: 01/04/2021] [Indexed: 11/16/2022]
Abstract
Invadopodia are actin-based proteolytic membrane protrusions required for invasive behavior and tumor growth. In this study, we used our high-content screening assay to identify kinases whose activity affects invadopodia formation. Among the top hits selected for further analysis was TAO3, an STE20-like kinase of the GCK subfamily. TAO3 was overexpressed in many human cancers and regulated invadopodia formation in melanoma, breast, and bladder cancers. Furthermore, TAO3 catalytic activity facilitated melanoma growth in three-dimensional matrices and in vivo. A novel, potent catalytic inhibitor of TAO3 was developed that inhibited invadopodia formation and function as well as tumor cell extravasation and growth. Treatment with this inhibitor demonstrated that TAO3 activity is required for endosomal trafficking of TKS5α, an obligate invadopodia scaffold protein. A phosphoproteomics screen for TAO3 substrates revealed the dynein subunit protein LIC2 as a relevant substrate. Knockdown of LIC2 or expression of a phosphomimetic form promoted invadopodia formation. Thus, TAO3 is a new therapeutic target with a distinct mechanism of action. SIGNIFICANCE: An unbiased screening approach identifies TAO3 as a regulator of invadopodia formation and function, supporting clinical development of this class of target.
Collapse
Affiliation(s)
- Shinji Iizuka
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California.,Department of Cell Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | | | - Jose C Navarro
- Department of Cell Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Kyle P Gribbin
- Department of Cell Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Robert J Ardecky
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Matthew M Abelman
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Chen-Ting Ma
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Eduard Sergienko
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Fu-Yue Zeng
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Ian Pass
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - George V Thomas
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Shannon K McWeeney
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon.,Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health and Science University, Portland, Oregon.,Oregon Clinical and Translational Research Institute, Oregon Health and Science University, Portland, Oregon
| | - Christian A Hassig
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | | | - Sara A Courtneidge
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California. .,Department of Cell Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon.,Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
13
|
Advances in Understanding TKS4 and TKS5: Molecular Scaffolds Regulating Cellular Processes from Podosome and Invadopodium Formation to Differentiation and Tissue Homeostasis. Int J Mol Sci 2020; 21:ijms21218117. [PMID: 33143131 PMCID: PMC7663256 DOI: 10.3390/ijms21218117] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Scaffold proteins are typically thought of as multi-domain "bridging molecules." They serve as crucial regulators of key signaling events by simultaneously binding multiple participants involved in specific signaling pathways. In the case of epidermal growth factor (EGF)-epidermal growth factor receptor (EGFR) binding, the activated EGFR contacts cytosolic SRC tyrosine-kinase, which then becomes activated. This process leads to the phosphorylation of SRC-substrates, including the tyrosine kinase substrates (TKS) scaffold proteins. The TKS proteins serve as a platform for the recruitment of key players in EGFR signal transduction, promoting cell spreading and migration. The TKS4 and the TKS5 scaffold proteins are tyrosine kinase substrates with four or five SH3 domains, respectively. Their structural features allow them to recruit and bind a variety of signaling proteins and to anchor them to the cytoplasmic surface of the cell membrane. Until recently, TKS4 and TKS5 had been recognized for their involvement in cellular motility, reactive oxygen species-dependent processes, and embryonic development, among others. However, a number of novel functions have been discovered for these molecules in recent years. In this review, we attempt to cover the diverse nature of the TKS molecules by discussing their structure, regulation by SRC kinase, relevant signaling pathways, and interaction partners, as well as their involvement in cellular processes, including migration, invasion, differentiation, and adipose tissue and bone homeostasis. We also describe related pathologies and the established mouse models.
Collapse
|
14
|
Iizuka S, Leon RP, Gribbin KP, Zhang Y, Navarro J, Smith R, Devlin K, Wang LG, Gibbs SL, Korkola J, Nan X, Courtneidge SA. Crosstalk between invadopodia and the extracellular matrix. Eur J Cell Biol 2020; 99:151122. [PMID: 33070041 DOI: 10.1016/j.ejcb.2020.151122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/24/2020] [Accepted: 08/12/2020] [Indexed: 12/27/2022] Open
Abstract
The scaffold protein Tks5α is required for invadopodia-mediated cancer invasion both in vitro and in vivo. We have previously also revealed a role for Tks5 in tumor cell growth using three-dimensional (3D) culture model systems and mouse transplantation experiments. Here we use both 3D and high-density fibrillar collagen (HDFC) culture to demonstrate that native collagen-I, but not a form lacking the telopeptides, stimulated Tks5-dependent growth, which was dependent on the DDR collagen receptors. We used microenvironmental microarray (MEMA) technology to determine that laminin, fibronectin and tropoelastin also stimulated invadopodia formation. A Tks5α-specific monoclonal antibody revealed its expression both on microtubules and at invadopodia. High- and super-resolution microscopy of cells in and on collagen was then used to place Tks5α at the base of invadopodia, separated from much of the actin and cortactin, but coincident with both matrix metalloprotease and cathepsin proteolytic activity. Inhibition of the Src family kinases, cathepsins or metalloproteases all reduced invadopodia length but each had distinct effects on Tks5α localization. These studies highlight the crosstalk between invadopodia and extracellular matrix components, and reveal the invadopodium to be a spatially complex structure.
Collapse
Affiliation(s)
- Shinji Iizuka
- Departments of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon, USA.
| | - Ronald P Leon
- Departments of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon, USA
| | - Kyle P Gribbin
- Departments of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon, USA
| | - Ying Zhang
- Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Jose Navarro
- Departments of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon, USA
| | - Rebecca Smith
- Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Kaylyn Devlin
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Lei G Wang
- Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Summer L Gibbs
- Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - James Korkola
- Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Xiaolin Nan
- Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Sara A Courtneidge
- Departments of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon, USA; Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA.
| |
Collapse
|
15
|
Thuault S, Mamelonet C, Salameh J, Ostacolo K, Chanez B, Salaün D, Baudelet E, Audebert S, Camoin L, Badache A. A proximity-labeling proteomic approach to investigate invadopodia molecular landscape in breast cancer cells. Sci Rep 2020; 10:6787. [PMID: 32321993 PMCID: PMC7176661 DOI: 10.1038/s41598-020-63926-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/06/2020] [Indexed: 12/27/2022] Open
Abstract
Metastatic progression is the leading cause of mortality in breast cancer. Invasive tumor cells develop invadopodia to travel through basement membranes and the interstitial matrix. Substantial efforts have been made to characterize invadopodia molecular composition. However, their full molecular identity is still missing due to the difficulty in isolating them. To fill this gap, we developed a non-hypothesis driven proteomic approach based on the BioID proximity biotinylation technology, using the invadopodia-specific protein Tks5α fused to the promiscuous biotin ligase BirA* as bait. In invasive breast cancer cells, Tks5α fusion concentrated to invadopodia and selectively biotinylated invadopodia components, in contrast to a fusion which lacked the membrane-targeting PX domain (Tks5β). Biotinylated proteins were isolated by affinity capture and identified by mass spectrometry. We identified known invadopodia components, revealing the pertinence of our strategy. Furthermore, we observed that Tks5 newly identified close neighbors belonged to a biologically relevant network centered on actin cytoskeleton organization. Analysis of Tks5β interactome demonstrated that some partners bound Tks5 before its recruitment to invadopodia. Thus, the present strategy allowed us to identify novel Tks5 partners that were not identified by traditional approaches and could help get a more comprehensive picture of invadopodia molecular landscape.
Collapse
Affiliation(s)
- Sylvie Thuault
- Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Univ, INSERM, Institut Paoli-Calmettes, CNRS, Marseille, France.
| | - Claire Mamelonet
- Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Univ, INSERM, Institut Paoli-Calmettes, CNRS, Marseille, France
| | - Joëlle Salameh
- Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Univ, INSERM, Institut Paoli-Calmettes, CNRS, Marseille, France.,INSERM UMR-S 1193, Univ. Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Kevin Ostacolo
- Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Univ, INSERM, Institut Paoli-Calmettes, CNRS, Marseille, France.,Department of Biochemistry and Molecular Biology, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Brice Chanez
- Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Univ, INSERM, Institut Paoli-Calmettes, CNRS, Marseille, France.,Institut Paoli-Calmettes, Department of Medical Oncology, Marseille, France
| | - Danièle Salaün
- Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Univ, INSERM, Institut Paoli-Calmettes, CNRS, Marseille, France
| | - Emilie Baudelet
- CRCM, Marseille Proteomics, Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Stéphane Audebert
- CRCM, Marseille Proteomics, Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Luc Camoin
- CRCM, Marseille Proteomics, Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Ali Badache
- Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Univ, INSERM, Institut Paoli-Calmettes, CNRS, Marseille, France
| |
Collapse
|
16
|
Burton KM, Cao H, Chen J, Qiang L, Krueger EW, Johnson KM, Bamlet WR, Zhang L, McNiven MA, Razidlo GL. Dynamin 2 interacts with α-actinin 4 to drive tumor cell invasion. Mol Biol Cell 2020; 31:439-451. [PMID: 31967944 PMCID: PMC7185896 DOI: 10.1091/mbc.e19-07-0395] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/23/2019] [Accepted: 01/15/2020] [Indexed: 12/21/2022] Open
Abstract
The large GTPase Dynamin 2 (Dyn2) is known to increase the invasiveness of pancreatic cancer tumor cells, but the mechanisms by which Dyn2 regulates changes in the actin cytoskeleton to drive cell migration are still unclear. Here we report that a direct interaction between Dyn2 and the actin-bundling protein alpha-actinin (α-actinin) 4 is critical for tumor cell migration and remodeling of the extracellular matrix in pancreatic ductal adenocarcinoma (PDAC) cells. The direct interaction is mediated through the C-terminal tails of both Dyn2 and α-actinin 4, and these proteins interact at invasive structures at the plasma membrane. While Dyn2 binds directly to both α-actinin 1 and α-actinin 4, only the interaction with α-actinin 4 is required to promote tumor cell invasion. Specific disruption of the Dyn2-α-actinin 4 interaction blocks the ability of PDAC cells to migrate in either two dimensions or invade through extracellular matrix as a result of impaired invadopodia stability. Analysis of human PDAC tumor tissue additionally reveals that elevated α-actinin 4 or Dyn2 expression are predictive of poor survival. Overall, these data demonstrate that Dyn2 regulates cytoskeletal dynamics, in part, by interacting with the actin-binding protein α-actinin 4 during tumor cell invasion.
Collapse
Affiliation(s)
- Kevin M. Burton
- Mayo Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905
| | - Hong Cao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Jing Chen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Li Qiang
- Mayo Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Eugene W. Krueger
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | | | - William R. Bamlet
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905
| | - Lizhi Zhang
- Department of Anatomic Pathology, Mayo Clinic, Rochester, MN 55905
| | - Mark A. McNiven
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Gina L. Razidlo
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
17
|
Daly C, Logan B, Breeyear J, Whitaker K, Ahmed M, Seals DF. Tks5 SH3 domains exhibit differential effects on invadopodia development. PLoS One 2020; 15:e0227855. [PMID: 31999741 PMCID: PMC6991978 DOI: 10.1371/journal.pone.0227855] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 12/31/2019] [Indexed: 11/23/2022] Open
Abstract
The Src substrate Tks5 helps scaffold matrix-remodeling invadopodia in invasive cancer cells. Focus was directed here on how the five SH3 domains of Tks5 impact that activity. Mutations designed to inhibit protein-protein interactions were created in the individual SH3 domains of Tks5, and the constructs were introduced into the LNCaP prostate carcinoma cell line, a model system with intrinsically low Tks5 expression and which our lab had previously showed the dependence of Src-dependent Tks5 phosphorylation on invadopodia development. In LNCaP cells, acute increases in wild-type Tks5 led to increased gelatin matrix degradation. A similar result was observed when Tks5 was mutated in its 4th or 5th SH3 domains. This was in contrast to the 1st, 2nd, and 3rd SH3 domain mutations of Tks5 where each had a remarkable accentuating effect on gelatin degradation. Conversely, in the invadopodia-competent Src-3T3 model system, mutations in any one of the first three SH3 domains had a dominant negative effect that largely eliminated the presence of invadopodia, inhibited gelatin degradation activity, and redistributed both Src, cortactin, and Tks5 to what are likely endosomal compartments. A hypothesis involving Tks5 conformational states and the regulation of endosomal trafficking is presented as an explanation for these seemingly disparate results.
Collapse
Affiliation(s)
- Christina Daly
- Department of Biology, Appalachian State University, Boone, North Carolina, United States of America
| | - Brewer Logan
- Department of Biology, Appalachian State University, Boone, North Carolina, United States of America
| | - Joseph Breeyear
- Department of Biology, Appalachian State University, Boone, North Carolina, United States of America
| | - Kelley Whitaker
- Department of Biology, Appalachian State University, Boone, North Carolina, United States of America
| | - Maryam Ahmed
- Department of Biology, Appalachian State University, Boone, North Carolina, United States of America
| | - Darren F Seals
- Department of Biology, Appalachian State University, Boone, North Carolina, United States of America
| |
Collapse
|
18
|
Ijuin T. Phosphoinositide phosphatases in cancer cell dynamics-Beyond PI3K and PTEN. Semin Cancer Biol 2019; 59:50-65. [PMID: 30922959 DOI: 10.1016/j.semcancer.2019.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 03/18/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Abstract
Phosphoinositides are a group of lipids that regulate intracellular signaling and subcellular biological events. The signaling by phosphatidylinositol-3,4,5-trisphosphate and Akt mediates the action of growth factors that are essential for cell proliferation, gene transcription, cell migration, and polarity. The hyperactivation of this signaling has been identified in different cancer cells; and, it has been implicated in oncogenic transformation and cancer cell malignancy. Recent studies have argued the role of phosphoinositides in cancer cell dynamics, including actin cytoskeletal rearrangement at the plasma membrane and the organization of intracellular compartments. The focus of this review is to summarize the impact of the activities of phosphoinositide phosphatases on intracellular signaling related to cancer cell dynamics and to discuss how the abnormalities in the activities of the enzymes alter the levels of phosphoinositides in cancer cells.
Collapse
Affiliation(s)
- Takeshi Ijuin
- Division of Biochemistry, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chu-o, Kobe 650-0017, Japan.
| |
Collapse
|
19
|
Ma X, Zhang L, Song J, Nguyen E, Lee RS, Rodgers SJ, Li F, Huang C, Schittenhelm RB, Chan H, Chheang C, Wu J, Brown KK, Mitchell CA, Simpson KJ, Daly RJ. Characterization of the Src-regulated kinome identifies SGK1 as a key mediator of Src-induced transformation. Nat Commun 2019; 10:296. [PMID: 30655532 PMCID: PMC6336867 DOI: 10.1038/s41467-018-08154-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 12/20/2018] [Indexed: 12/13/2022] Open
Abstract
Despite significant progress, our understanding of how specific oncogenes transform cells is still limited and likely underestimates the complexity of downstream signalling events. To address this gap, we use mass spectrometry-based chemical proteomics to characterize the global impact of an oncogene on the expressed kinome, and then functionally annotate the regulated kinases. As an example, we identify 63 protein kinases exhibiting altered expression and/or phosphorylation in Src-transformed mammary epithelial cells. An integrated siRNA screen identifies nine kinases, including SGK1, as being essential for Src-induced transformation. Accordingly, we find that Src positively regulates SGK1 expression in triple negative breast cancer cells, which exhibit a prominent signalling network governed by Src family kinases. Furthermore, combined inhibition of Src and SGK1 reduces colony formation and xenograft growth more effectively than either treatment alone. Therefore, this approach not only provides mechanistic insights into oncogenic transformation but also aids the design of improved therapeutic strategies. The systemic understanding of oncogenic kinase signalling is still limited. Here, the authors combine chemical proteomics with functional screens to assess the impact of oncogenic Src on the expressed kinome and identify SGK1 as a critical mediator of Src-induced cell transformation.
Collapse
Affiliation(s)
- Xiuquan Ma
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Luxi Zhang
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Jiangning Song
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Monash Centre for Data Science, Faculty of Information Technology, Monash University, Melbourne, VIC, 3800, Australia
| | - Elizabeth Nguyen
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Rachel S Lee
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Samuel J Rodgers
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Fuyi Li
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Cheng Huang
- Monash Biomedical Proteomics Facility and Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Ralf B Schittenhelm
- Monash Biomedical Proteomics Facility and Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Howard Chan
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Chanly Chheang
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Jianmin Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Centre for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Kristin K Brown
- Cancer Therapeutics Program and Cancer Metabolism Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC, 3010, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Christina A Mitchell
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Kaylene J Simpson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia. .,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
20
|
Chen YC, Baik M, Byers JT, Chen KT, French SW, Díaz B. Experimental supporting data on TKS5 and Cortactin expression and localization in human pancreatic cancer cells and tumors. Data Brief 2018; 22:132-136. [PMID: 30581916 PMCID: PMC6297239 DOI: 10.1016/j.dib.2018.11.138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 11/27/2022] Open
Abstract
In this article, using human pancreatic cancer cell lines and tumor specimens, we analyze the expression and localization of the invadopodia-related proteins TKS5 and Cortactin. Specifically, we present data on: a) TKS5 expression and localization by immunofluorescence in human pancreatic tumors, b) Cortactin expression by western blotting in various human pancreatic adenocarcinoma cell lines, c) TKS5 and Cortactin localization at invadopodia in BxPC-3 pancreatic adenocarcinoma cells, and d) TKS5 and Cortactin localization by co-immunofluorescence in human pancreatic cancer specimens. Data presented here is related to and supportive of the research article by Chen et al., "TKS5-positive invadopodia-like structures in human tumor surgical specimens" (Chen et al., 2019), where interpretation of the research data presented here is available.
Collapse
Affiliation(s)
- Yu-Chuan Chen
- Division of Medical Hematology Oncology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California USA
| | - Matthew Baik
- Division of Medical Hematology Oncology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California USA
| | - Joshua T Byers
- Department of Pathology. Harbor-UCLA Medical Center, Torrance, California USA
| | - Kathryn T Chen
- Department of Surgery. Harbor-UCLA Medical Center, Torrance, California USA.,David Geffen School of Medicine at UCLA, Los Angeles, California USA
| | - Samuel W French
- Department of Pathology. Harbor-UCLA Medical Center, Torrance, California USA.,David Geffen School of Medicine at UCLA, Los Angeles, California USA
| | - Begoña Díaz
- Division of Medical Hematology Oncology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California USA.,David Geffen School of Medicine at UCLA, Los Angeles, California USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California USA
| |
Collapse
|
21
|
Cui Y, Sun G. Structural versatility that serves the function of the HRD motif in the catalytic loop of protein tyrosine kinase, Src. Protein Sci 2018; 28:533-542. [PMID: 30461096 DOI: 10.1002/pro.3554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/19/2022]
Abstract
Site-directed mutagenesis is a traditional approach for structure-function analysis of protein tyrosine kinases, and it requires the generation, expression, purification, and analysis of each mutant enzyme. In this study, we report a versatile high throughput bacterial screening system that can identify functional kinase mutants by immunological detection of tyrosine phosphorylation. Two key features of this screening system are noteworthy. First, instead of blotting bacterial colonies directly from Agar plates to nitrocellulose membrane, the colonies were cultured in 96-well plates, and then spotted in duplicate onto the membrane with appropriate controls. This made the screening much more reliable compared with direct colony blotting transfer. A second feature is the parallel use of a protein tyrosine phosphatase (PTP)-expressing host and a non-PTP-expressing host. Because high activity Src mutants are toxic to the host, the PTP system allowed the identification of Src mutants with high activity, while the non-PTP system identified Src mutants with low activity. This approach was applied to Src mutant libraries randomized in the highly conserved HRD motif in the catalytic loop, and revealed that structurally diverse residues can replace the His and Arg residues, while the Asp residue is irreplaceable for catalytic activity.
Collapse
Affiliation(s)
- Yixin Cui
- Department of Cell and Molecular Biology, Center for Biotechnology and Life Science, University of Rhode Island, 120 Flagg Road, Kingston, Rhode Island, 02881
| | - Gongqin Sun
- Department of Cell and Molecular Biology, Center for Biotechnology and Life Science, University of Rhode Island, 120 Flagg Road, Kingston, Rhode Island, 02881
| |
Collapse
|
22
|
Chen YC, Baik M, Byers JT, Chen KT, French SW, Díaz B. TKS5-positive invadopodia-like structures in human tumor surgical specimens. Exp Mol Pathol 2018; 106:17-26. [PMID: 30439350 DOI: 10.1016/j.yexmp.2018.11.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/02/2018] [Accepted: 11/09/2018] [Indexed: 12/15/2022]
Abstract
Invadopodia, cancer cell protrusions with proteolytic activity, are functionally associated with active remodeling of the extracellular matrix. Here, we show that the invadopodia-related protein TKS5 is expressed in human pancreatic adenocarcinoma lines, and demonstrate that pancreatic cancer cells depend on TKS5 for invadopodia formation and function. Immunofluorescence staining of human pancreatic cancer cells reveals that TKS5 is a marker of mature and immature invadopodia. We also analyze the co-staining patterns of TKS5 and the commonly used invadopodia marker Cortactin, and find only partial co-localization of these two proteins at invadopodia, with a large fraction of TKS5-positive invadopodia lacking detectable levels of Cortactin. Whereas compelling evidence exist on the role of invadopodia as mediators of invasive migration in cultured cells and in animal models of cancer, these structures have never been detected inside human tumors. Here, using antibodies against TKS5 and Cortactin, we describe for the first time structures strongly resembling invadopodia in various paraffin-embedded human tumor surgical specimens from pancreas and other organs. Our results strongly suggest that invadopodia are present inside human tumors, and warrants further investigation on their regulation and occurrence in surgical specimens, and on the value of TKS5 antibodies as pathological research and diagnostic tools.
Collapse
Affiliation(s)
- Yu-Chuan Chen
- Division on Medical Oncology Hematology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Matthew Baik
- Division on Medical Oncology Hematology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joshua T Byers
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kathryn T Chen
- Department of Surgery, Harbor-UCLA Medical Center, Torrance, CA, USA; David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Samuel W French
- Department of Pathology, Harbor-UCLA Medical Center, Torrance, CA, USA; David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Begoña Díaz
- Division on Medical Oncology Hematology, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA; David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Moodley S, Derouet M, Bai XH, Xu F, Kapus A, Yang BB, Liu M. Stimulus-dependent dissociation between XB130 and Tks5 scaffold proteins promotes airway epithelial cell migration. Oncotarget 2018; 7:76437-76452. [PMID: 27835612 PMCID: PMC5363521 DOI: 10.18632/oncotarget.13261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/02/2016] [Indexed: 02/07/2023] Open
Abstract
Repair of airway epithelium after injury requires migration of neighboring epithelial cells to injured areas. However, the molecular mechanisms regulating airway epithelial cell migration is not well defined. We have previously shown that XB130, a scaffold protein, is required for airway epithelial repair and regeneration in vivo, and interaction between XB130 and another scaffold protein, Tks5, regulates cell proliferation and survival in human bronchial epithelial cells. The objective of the present study was to determine the role of XB130 and Tks5 interaction in airway epithelial cell migration. Interestingly, we found that XB130 only promotes lateral cell migration, whereas, Tks5 promotes cell migration/invasion via proteolysis of extracellular matrix. Upon stimulation with EGF, PKC activator phorbol 12, 13-dibutyrate or a nicotinic acetylcholine receptor ligand, XB130 and Tks5 translocated to the cell membrane in a stimulus-dependent manner. The translocation and distribution of XB130 is similar to lamellipodial marker, WAVE2; whereas Tks5 is similar to podosome marker, N-WASP. Over-expression of XB130 or Tks5 alone enhances cell migration, whereas co-expression of both XB130 and Tks5 inhibits cell migration processes and signaling. Furthermore, XB130 interacts with Rac1 whereas Tks5 interacts with Cdc42 to promote Rho GTPase activity. Our results suggest that dissociation between XB130 and Tks5 may facilitate lateral cell migration via XB130/Rac1, and vertical cell migration via Tks5/Cdc42. These molecular mechanisms will help our understanding of airway epithelial repair and regeneration.
Collapse
Affiliation(s)
- Serisha Moodley
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Mathieu Derouet
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Xiao Hui Bai
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Feng Xu
- Advanced Optical Microscopy Facility, UHN, Toronto, Canada
| | - Andras Kapus
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Burton B Yang
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Mingyao Liu
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, Canada
| |
Collapse
|
24
|
Iizuka S, Abdullah C, Buschman MD, Diaz B, Courtneidge SA. The role of Tks adaptor proteins in invadopodia formation, growth and metastasis of melanoma. Oncotarget 2018; 7:78473-78486. [PMID: 27802184 PMCID: PMC5346654 DOI: 10.18632/oncotarget.12954] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/22/2016] [Indexed: 01/07/2023] Open
Abstract
Metastatic cancer cells are characterized by their ability to degrade and invade through extracellular matrix. We previously showed that the Tks adaptor proteins, Tks4 and Tks5, are required for invadopodia formation and/or function in Src-transformed fibroblasts and a number of human cancer cell types. In this study, we investigated the role of Tks adaptor proteins in melanoma cell invasion and metastasis. Knockdown of either Tks4 or Tks5 in both mouse and human melanoma cell lines resulted in a decreased ability to form invadopodia and degrade extracellular matrix. In addition, Tks-knockdown melanoma cells had decreased proliferation in a 3-dimensional type l collagen matrix, but not in 2-dimensional culture conditions. We also investigated the role of Tks proteins in melanoma progression in vivo using xenografts and experimental metastasis assays. Consistent with our in vitro results, reduction of Tks proteins markedly reduced subcutaneous melanoma growth as well as metastatic growth in the lung. We explored the clinical relevance of Tks protein expression in human melanoma specimens using a tissue microarray. Compared to non-malignant nevi, both Tks proteins were highly expressed in melanoma tissues. Moreover, metastatic melanoma cases showed higher expression of Tks5 than primary melanoma cases. Taken together, these findings suggest the importance of Tks adaptor proteins in melanoma growth and metastasis in vivo, likely via functional invadopodia formation.
Collapse
Affiliation(s)
- Shinji Iizuka
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Christopher Abdullah
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Matthew D Buschman
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, USA
| | - Begoña Diaz
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sara A Courtneidge
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Department of Biomedical Engineering and Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
25
|
Gorai S, Paul D, Borah R, Haloi N, Santra MK, Manna D. Role of Cationic Groove and Hydrophobic Residues in Phosphatidylinositol-Dependent Membrane-Binding Properties of Tks5-Phox Homology Domain. ChemistrySelect 2018. [DOI: 10.1002/slct.201702558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Sukhamoy Gorai
- Department of Chemistry; Indian Institute of Technology Guwahati; Guwahati - 781039, Assam India
| | - Debasish Paul
- National Centre for Cell Science; Pune 411007, Maharashtra India
| | - Rituparna Borah
- Department of Chemistry; Indian Institute of Technology Guwahati; Guwahati - 781039, Assam India
| | - Nandan Haloi
- Department of Chemistry; Indian Institute of Technology Guwahati; Guwahati - 781039, Assam India
| | | | - Debasis Manna
- Department of Chemistry; Indian Institute of Technology Guwahati; Guwahati - 781039, Assam India
| |
Collapse
|
26
|
Abstract
Tyrosine kinase substrate (Tks) adaptor proteins are considered important regulators of various physiological and/or pathological processes, particularly cell migration and invasion, and cancer progression. These proteins contain PX and SH3 domains, and act as scaffolds, bringing membrane and cellular components in close proximity in structures known as invadopodia or podosomes. Tks proteins, analogous to the related proteins p47phox, p40phox and NoxO1, also facilitate local generation of reactive oxygen species (ROS), which aid in signaling at invadopodia and/or podosomes to promote their activity. As their name suggests, Tks adaptor proteins are substrates for tyrosine kinases, especially Src. In this Cell Science at a Glance article and accompanying poster, we discuss the known structural and functional aspects of Tks adaptor proteins. As the science of Tks proteins is evolving, this article will point out where we stand and what still needs to be explored. We also underscore pathological conditions involving these proteins, providing a basis for future research to develop therapies for treatment of these diseases.
Collapse
Affiliation(s)
- Priyanka Saini
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Sara A Courtneidge
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
27
|
Paterson EK, Courtneidge SA. Invadosomes are coming: new insights into function and disease relevance. FEBS J 2017; 285:8-27. [PMID: 28548369 DOI: 10.1111/febs.14123] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/09/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022]
Abstract
Invadopodia and podosomes are discrete, actin-based molecular protrusions that form in cancer cells and normal cells, respectively, in response to diverse signaling pathways and extracellular matrix cues. Although they participate in a host of different cellular processes, they share a common functional theme of controlling pericellular proteolytic activity, which sets them apart from other structures that function in migration and adhesion, including focal adhesions, lamellipodia, and filopodia. In this review, we highlight research that explores the function of these complex structures, including roles for podosomes in embryonic and postnatal development, in angiogenesis and remodeling of the vasculature, in maturation of the postsynaptic membrane, in antigen sampling and recognition, and in cell-cell fusion mechanisms, as well as the involvement of invadopodia at multiple steps of the metastatic cascade, and how all of this may apply in the treatment of human disease states. Finally, we explore recent research that implicates a novel role for exosomes and microvesicles in invadopodia-dependent and invadopodia-independent mechanisms of invasion, respectively.
Collapse
Affiliation(s)
- Elyse K Paterson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Sara A Courtneidge
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
28
|
NADPH Oxidases: Insights into Selected Functions and Mechanisms of Action in Cancer and Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017. [PMID: 28626501 PMCID: PMC5463201 DOI: 10.1155/2017/9420539] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
NADPH oxidases (NOX) are reactive oxygen species- (ROS-) generating enzymes regulating numerous redox-dependent signaling pathways. NOX are important regulators of cell differentiation, growth, and proliferation and of mechanisms, important for a wide range of processes from embryonic development, through tissue regeneration to the development and spread of cancer. In this review, we discuss the roles of NOX and NOX-derived ROS in the functioning of stem cells and cancer stem cells and in selected aspects of cancer cell physiology. Understanding the functions and complex activities of NOX is important for the application of stem cells in tissue engineering, regenerative medicine, and development of new therapies toward invasive forms of cancers.
Collapse
|
29
|
Ke Y, Bao T, Zhou Q, Wang Y, Ge J, Fu B, Wu X, Tang H, Shi Z, Lei X, Zhang C, Tan Y, Chen H, Guo Z, Wang L. Discs large homolog 5 decreases formation and function of invadopodia in human hepatocellular carcinoma via Girdin and Tks5. Int J Cancer 2017; 141:364-376. [PMID: 28390157 DOI: 10.1002/ijc.30730] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 02/23/2017] [Accepted: 03/22/2017] [Indexed: 12/21/2022]
Abstract
Invadopodium formation is a crucial early event of invasion and metastasis of hepatocellular carcinoma (HCC). However, the molecular mechanisms underlying regulation of invadopodia remain elusive. This study aimed to investigate the potential role of discs large homolog 5 (Dlg5) in invadopodium formation and function in HCC. We found that Dlg5 expression was significantly lower in human HCC tissues and cell lines than adjacent nontumor tissues and liver cells. Lower Dlg5 expression was associated with advanced stages of HCC, and poor overall and disease-free survival of HCC patients. Dlg5-silencing promoted epithelial-mesenchymal transition, invadopodium formation, gelatin degradation function, and invadopodium-associated invasion of HepG2 cells. In contrast, Dlg5 overexpression inhibited epithelial-mesenchymal transition, functional invadopodium formation, and invasion of SK-Hep1 cells. Both Girdin and Tks5, but not the Tks5 nonphosphorylatable mutant, were responsible for the enhanced invadopodium formation and invasion of Dlg5-silenced HepG2 cells. Furthermore, Dlg5 interacted with Girdin and interfered with the interaction of Girdin and Tks5. Dlg5 silencing promoted Girdin and Tks5 phosphorylation, which was abrogated by Girdin silencing and rescued by inducing shRNA-resistant Girdin expression. Moreover, Dlg5 overexpression significantly inhibited HCC intrahepatic and lung metastasis in vivo. Taken together, our data indicate that Dlg5 acts as a novel regulator of invadopodium-associated invasion via Girdin and by interfering with the interaction between Girdin and Tks5, which might be important for Tks5 phosphorylation in HCC cells. Conceivably, Dlg5 may act as a new biomarker for prognosis of HCC patients.
Collapse
Affiliation(s)
- Yang Ke
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tianhao Bao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China.,The Mental Health Center of Kunming Medical University, Kunming, China
| | - Qixin Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yan Wang
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiayun Ge
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bimang Fu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuesong Wu
- Department of Gastroenterological Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haoran Tang
- Department of Gastroenterological Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhitian Shi
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuefen Lei
- Deparment of Medical Oncology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Cheng Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuqi Tan
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haotian Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhitang Guo
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
30
|
Significance of kinase activity in the dynamic invadosome. Eur J Cell Biol 2016; 95:483-492. [PMID: 27465307 DOI: 10.1016/j.ejcb.2016.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/08/2016] [Accepted: 07/13/2016] [Indexed: 12/19/2022] Open
Abstract
Invadosomes are actin rich protrusive structures that facilitate invasive migration in multiple cell types. Comprised of invadopodia and podosomes, these highly dynamic structures adhere to and degrade the extracellular matrix, and are also thought to play a role in mechanosensing. Many extracellular signals have been implicated in invadosome stimulation, activating complex signalling cascades to drive the formation, activity and turnover of invadosomes. While the structural components of invadosomes have been well studied, the regulation of invadosome dynamics is still poorly understood. Protein kinases are essential to this regulation, affecting all stages of invadosome dynamics and allowing tight spatiotemporal control of their activity. Invadosome organisation and function have been linked to pathophysiological states such as cancer invasion and metastasis; therapeutic targeting of invadosome regulatory components is thus warranted. In this review, we discuss the involvement of kinase signalling in every stage of the invadosome life cycle and evaluate its significance.
Collapse
|
31
|
Bendris N, Stearns CJS, Reis CR, Rodriguez-Canales J, Liu H, Witkiewicz AW, Schmid SL. Sorting nexin 9 negatively regulates invadopodia formation and function in cancer cells. J Cell Sci 2016; 129:2804-16. [PMID: 27278018 DOI: 10.1242/jcs.188045] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/26/2016] [Indexed: 01/11/2023] Open
Abstract
The ability of cancer cells to degrade the extracellular matrix and invade interstitial tissues contributes to their metastatic potential. We recently showed that overexpression of sorting nexin 9 (SNX9) leads to increased cell invasion and metastasis in animal models, which correlates with increased SNX9 protein expression in metastases from human mammary cancers. Here, we report that SNX9 expression is reduced relative to neighboring normal tissues in primary breast tumors, and progressively reduced in more aggressive stages of non-small-cell lung cancers. We show that SNX9 is localized at invadopodia where it directly binds the invadopodia marker TKS5 and negatively regulates invadopodia formation and function. SNX9 depletion increases invadopodia number and the local recruitment of MT1-MMP by decreasing its internalization. Together, these effects result in increased localized matrix degradation. We further identify SNX9 as a Src kinase substrate and show that this phosphorylation is important for SNX9 activity in regulating cell invasion, but is dispensable for its function in regulating invadopodia. The diversified changes associated with SNX9 expression in cancer highlight its importance as a central regulator of cancer cell behavior.
Collapse
Affiliation(s)
- Nawal Bendris
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX75390, USA
| | - Carrie J S Stearns
- Department of Molecular Medicine, Veterinary Medical Center, Cornell University, Ithaca, NY14853, USA
| | - Carlos R Reis
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX75390, USA
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX77030, USA
| | - Hui Liu
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX77030, USA Department of Pathology, Xuzhou Medical College, Province of Jiangsu, China
| | - Agnieszka W Witkiewicz
- Simmons Cancer Center, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX390, USA
| | - Sandra L Schmid
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX75390, USA
| |
Collapse
|
32
|
Wang H, Leung M, Wandinger-Ness A, Hudson LG, Song M. Constrained inference of protein interaction networks for invadopodium formation in cancer. IET Syst Biol 2016; 10:76-85. [PMID: 26997662 PMCID: PMC4804358 DOI: 10.1049/iet-syb.2015.0009] [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: 01/29/2015] [Revised: 06/27/2015] [Accepted: 07/10/2015] [Indexed: 11/19/2022] Open
Abstract
Integrating prior molecular network knowledge into interpretation of new experimental data is routine practice in biology research. However, a dilemma for deciphering interactome using Bayes' rule is the demotion of novel interactions with low prior probabilities. Here the authors present constrained generalised logical network (CGLN) inference to predict novel interactions in dynamic networks, respecting previously known interactions and observed temporal coherence. It encodes prior interactions as probabilistic logic rules called local constraints, and forms global constraints using observed dynamic patterns. CGLN finds constraint-satisfying trajectories by solving a k-stops problem in the state space of dynamic networks and then reconstructs candidate networks. They benchmarked CGLN on randomly generated networks, and CGLN outperformed its alternatives when 50% or more interactions in a network are given as local constraints. CGLN is then applied to infer dynamic protein interaction networks regulating invadopodium formation in motile cancer cells. CGLN predicted 134 novel protein interactions for their involvement in invadopodium formation. The most frequently predicted interactions centre around focal adhesion kinase and tyrosine kinase substrate TKS4, and 14 interactions are supported by the literature in molecular contexts related to invadopodium formation. As an alternative to the Bayesian paradigm, the CGLN method offers constrained network inference without requiring prior probabilities and thus can promote novel interactions, consistent with the discovery process of scientific facts that are not yet in common beliefs.
Collapse
Affiliation(s)
- Haizhou Wang
- Department of Computer Science, New Mexico State University, Las Cruces, NM 88003, USA
| | - Ming Leung
- Departments of Biology and Computer Science, Duke University, Durham, NC 27708, USA
| | | | - Laurie G Hudson
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131, USA
| | - Mingzhou Song
- Department of Computer Science, New Mexico State University, Las Cruces, NM 88003, USA.
| |
Collapse
|
33
|
Jensen HH, Pedersen HN, Stenkjær E, Pedersen GA, Login FH, Nejsum LN. Tir Is Essential for the Recruitment of Tks5 to Enteropathogenic Escherichia coli Pedestals. PLoS One 2015; 10:e0141871. [PMID: 26536015 PMCID: PMC4633291 DOI: 10.1371/journal.pone.0141871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/14/2015] [Indexed: 11/24/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) is a bacterial pathogen that infects the epithelial lining of the small intestine and causes diarrhea. Upon attachment to the intestinal epithelium, EPEC uses a Type III Secretion System to inject its own high affinity receptor Translocated intimin receptor (Tir) into the host cell. Tir facilitates tight adhesion and recruitment of actin-regulating proteins leading to formation of an actin pedestal beneath the infecting bacterium. The pedestal has several similarities with podosomes, which are basolateral actin-rich extensions found in some migrating animal cells. Formation of podosomes is dependent upon the early podosome-specific scavenger protein Tks5, which is involved in actin recruitment. Although Tks5 is expressed in epithelial cells, and podosomes and EPEC pedestals share many components in their structure and mechanism of formation, the potential role of Tks5 in EPEC infections has not been studied. The aim of this study was to determine the subcellular localization of Tks5 in epithelial cells and to investigate if Tks5 is recruited to the EPEC pedestal. In an epithelial MDCK cell line stably expressing Tks5-EGFP, Tks5 localized to actin bundles. Upon infection, EPEC recruited Tks5-EGFP. Tir, but not Tir phosphorylation was essential for the recruitment. Time-lapse microscopy revealed that Tks5-EGFP was recruited instantly upon EPEC attachment to host cells, simultaneously with actin and N-WASp. EPEC infection of cells expressing a ΔPX-Tks5 deletion version of Tks5 showed that EPEC was able to both infect and form pedestals when the PX domain was deleted from Tks5. Future investigations will clarify the role of Tks5 in EPEC infection and pedestal formation.
Collapse
Affiliation(s)
- Helene H. Jensen
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Hans N. Pedersen
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Eva Stenkjær
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Gitte A. Pedersen
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Frédéric H. Login
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
| | - Lene N. Nejsum
- Institute of Molecular Biology and Genetics and Interdiciplinary Nanoscience Center, Aarhus University, C. F. Moellers Allé 3, Aarhus, Denmark
- * E-mail:
| |
Collapse
|
34
|
Sirvent A, Urbach S, Roche S. Contribution of phosphoproteomics in understanding SRC signaling in normal and tumor cells. Proteomics 2015; 15:232-44. [PMID: 25403792 DOI: 10.1002/pmic.201400162] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/30/2014] [Accepted: 11/12/2014] [Indexed: 01/02/2023]
Abstract
The membrane-anchored, non-receptor tyrosine kinase (non-RTK) SRC is a critical regulator of signal transduction induced by a large variety of cell-surface receptors, including RTKs that bind to growth factors to control cell growth and migration. When deregulated, SRC shows strong oncogenic activity, probably because of its capacity to promote RTK-mediated downstream signaling even in the absence of extracellular stimuli. Accordingly, SRC is frequently deregulated in human cancer and is thought to play important roles during tumorigenesis. However, our knowledge on the molecular mechanism by which SRC controls signaling is incomplete due to the limited number of key substrates identified so far. Here, we review how phosphoproteomic methods have changed our understanding of the mechanisms underlying SRC signaling in normal and tumor cells and discuss how these novel findings can be used to improve therapeutic strategies aimed at targeting SRC signaling in human cancer.
Collapse
Affiliation(s)
- Audrey Sirvent
- CNRS UMR5237, University Montpellier 1 and 2, CRBM, Montpellier, France
| | | | | |
Collapse
|
35
|
Regulation of sarcoma cell migration, invasion and invadopodia formation by AFAP1L1 through a phosphotyrosine-dependent pathway. Oncogene 2015. [DOI: 10.1038/onc.2015.272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
36
|
Di Martino J, Paysan L, Gest C, Lagrée V, Juin A, Saltel F, Moreau V. Cdc42 and Tks5: a minimal and universal molecular signature for functional invadosomes. Cell Adh Migr 2015; 8:280-92. [PMID: 24840388 DOI: 10.4161/cam.28833] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Invadosomes are actin-based structures involved in extracellular-matrix degradation. Invadosomes, either known as podosomes or invadopodia, are found in an increasing number of cell types. Moreover, their overall organization and molecular composition may vary from one cell type to the other. Some are constitutive such as podosomes in hematopoietic cells whereas others are inducible. However, they share the same feature, their ability to interact and to degrade the extracellular matrix. Based on the literature and our own experiments, the aim of this study was to establish a minimal molecular definition of active invadosomes. We first highlighted that Cdc42 is the key RhoGTPase involved in invadosome formation in all described models. Using different cellular models, such as NIH-3T3, HeLa, and endothelial cells, we demonstrated that overexpression of an active form of Cdc42 is sufficient to form invadosome actin cores. Therefore, active Cdc42 must be considered not only as an inducer of filopodia, but also as an inducer of invadosomes. Depending on the expression level of Tks5, these Cdc42-dependent actin cores were endowed or not with a proteolytic activity. In fact, Tks5 overexpression rescued this activity in Tks5 low expressing cells. We thus described the adaptor protein Tks5 as a major actor of the invadosome degradation function. Surprisingly, we found that Src kinases are not always required for invadosome formation and function. These data suggest that even if Src family members are the principal kinases involved in the majority of invadosomes, it cannot be considered as a common element for all invadosome structures. We thus define a minimal and universal molecular signature of invadosome that includes Cdc42 activity and Tks5 presence in order to drive the actin machinery and the proteolytic activity of these invasive structures.
Collapse
Affiliation(s)
- Julie Di Martino
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Lisa Paysan
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Caroline Gest
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Valérie Lagrée
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Amélie Juin
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Frédéric Saltel
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Violaine Moreau
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| |
Collapse
|
37
|
Tsujita K, Itoh T. Phosphoinositides in the regulation of actin cortex and cell migration. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:824-31. [DOI: 10.1016/j.bbalip.2014.10.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Revised: 10/08/2014] [Accepted: 10/22/2014] [Indexed: 10/25/2022]
|
38
|
Blouw B, Patel M, Iizuka S, Abdullah C, You WK, Huang X, Li JL, Diaz B, Stallcup WB, Courtneidge SA. The invadopodia scaffold protein Tks5 is required for the growth of human breast cancer cells in vitro and in vivo. PLoS One 2015; 10:e0121003. [PMID: 25826475 PMCID: PMC4380437 DOI: 10.1371/journal.pone.0121003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 02/09/2015] [Indexed: 01/07/2023] Open
Abstract
The ability of cancer cells to invade underlies metastatic progression. One mechanism by which cancer cells can become invasive is through the formation of structures called invadopodia, which are dynamic, actin-rich membrane protrusions that are sites of focal extracellular matrix degradation. While there is a growing consensus that invadopodia are instrumental in tumor metastasis, less is known about whether they are involved in tumor growth, particularly in vivo. The adaptor protein Tks5 is an obligate component of invadopodia, and is linked molecularly to both actin-remodeling proteins and pericellular proteases. Tks5 appears to localize exclusively to invadopodia in cancer cells, and in vitro studies have demonstrated its critical requirement for the invasive nature of these cells, making it an ideal surrogate to investigate the role of invadopodia in vivo. In this study, we examined how Tks5 contributes to human breast cancer progression. We used immunohistochemistry and RNA sequencing data to evaluate Tks5 expression in clinical samples, and we characterized the role of Tks5 in breast cancer progression using RNA interference and orthotopic implantation in SCID-Beige mice. We found that Tks5 is expressed to high levels in approximately 50% of primary invasive breast cancers. Furthermore, high expression was correlated with poor outcome, particularly in those patients with late relapse of stage I/II disease. Knockdown of Tks5 expression in breast cancer cells resulted in decreased growth, both in 3D in vitro cultures and in vivo. Moreover, our data also suggest that Tks5 is important for the integrity and permeability of the tumor vasculature. Together, this work establishes an important role for Tks5 in tumor growth in vivo, and suggests that invadopodia may play broad roles in tumor progression.
Collapse
Affiliation(s)
- Barbara Blouw
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Manishha Patel
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Shinji Iizuka
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Christopher Abdullah
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Weon Kyoo You
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Xiayu Huang
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Jian-Liang Li
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Begoña Diaz
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - William B. Stallcup
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Sara A. Courtneidge
- Tumor Microenvironment and Metastasis Program, NCI Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
39
|
Chevalier C, Cannet A, Descamps S, Sirvent A, Simon V, Roche S, Benistant C. ABL tyrosine kinase inhibition variable effects on the invasive properties of different triple negative breast cancer cell lines. PLoS One 2015; 10:e0118854. [PMID: 25803821 PMCID: PMC4372365 DOI: 10.1371/journal.pone.0118854] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/16/2015] [Indexed: 12/20/2022] Open
Abstract
The non-receptor tyrosine kinase ABL drives myeloid progenitor expansion in human chronic myeloid leukemia. ABL inhibition by the tyrosine kinase inhibitor nilotinib is a first-line treatment for this disease. Recently, ABL has also been implicated in the transforming properties of solid tumors, including triple negative (TN) breast cancer. TN breast cancers are highly metastatic and several cell lines derived from these tumors display high invasive activity in vitro. This feature is associated with the activation of actin-rich membrane structures called invadopodia that promote extracellular matrix degradation. Here, we investigated nilotinib effect on the invasive and migratory properties of different TN breast cancer cell lines. Nilotinib decreased both matrix degradation and invasion in the TN breast cancer cell lines MDA-MB 231 and MDA-MB 468. However, and unexpectedly, nilotinib increased by two-fold the invasive properties of the TN breast cancer cell line BT-549 and of Src-transformed fibroblasts. Both display much higher levels of ABL kinase activity compared to MDA-MB 231. Similar effects were obtained by siRNA-mediated down-regulation of ABL expression, confirming ABL central role in this process. ABL anti-tumor effect in BT-549 cells and Src-transformed fibroblasts was not dependent on EGF secretion, as recently reported in neck and squamous carcinoma cells. Rather, we identified the TRIO-RAC1 axis as an important downstream element of ABL activity in these cancer cells. In conclusion, the observation that TN breast cancer cell lines respond differently to ABL inhibitors could have implications for future therapies.
Collapse
Affiliation(s)
- Clément Chevalier
- Centre de Recherche de Biochimie Macromoléculaire CNRS UMR5237, University of Montpellier, Montpellier, 34000, France
| | - Aude Cannet
- Centre de Recherche de Biochimie Macromoléculaire CNRS UMR5237, University of Montpellier, Montpellier, 34000, France
| | - Simon Descamps
- Centre de Recherche de Biochimie Macromoléculaire CNRS UMR5237, University of Montpellier, Montpellier, 34000, France
| | - Audrey Sirvent
- Centre de Recherche de Biochimie Macromoléculaire CNRS UMR5237, University of Montpellier, Montpellier, 34000, France
| | - Valérie Simon
- Centre de Recherche de Biochimie Macromoléculaire CNRS UMR5237, University of Montpellier, Montpellier, 34000, France
| | - Serge Roche
- Centre de Recherche de Biochimie Macromoléculaire CNRS UMR5237, University of Montpellier, Montpellier, 34000, France
- * E-mail: (SR); (CB)
| | - Christine Benistant
- Centre de Biochimie Structurale, CNRS UMR 5048, INSERM UMR 1054, University of Montpellier, Montpellier, 34090, France
- * E-mail: (SR); (CB)
| |
Collapse
|
40
|
Genetic disruption of the sh3pxd2a gene reveals an essential role in mouse development and the existence of a novel isoform of tks5. PLoS One 2014; 9:e107674. [PMID: 25259869 PMCID: PMC4178035 DOI: 10.1371/journal.pone.0107674] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/12/2014] [Indexed: 01/07/2023] Open
Abstract
Tks5 is a scaffold protein and Src substrate involved in cell migration and matrix degradation through its essential role in invadosome formation and function. We have previously described that Tks5 is fundamental for zebrafish neural crest cell migration in vivo. In the present study, we sought to investigate the function of Tks5 in mammalian development by analyzing mice mutant for sh3pxd2a, the gene encoding Tks5. Homozygous disruption of the sh3pxd2a gene by gene-trapping in mouse resulted in neonatal death and the presence of a complete cleft of the secondary palate. Interestingly, embryonic fibroblasts from homozygous gene-trap sh3pxd2a mice lacked only the highest molecular weight band of the characteristic Tks5 triplet observed in protein extracts, leaving the lower molecular weight bands unaffected. This finding, together with the existence of two human Expressed Sequence Tags lacking the first 5 exons of SH3PXD2A, made us hypothesize about the presence of a second alternative transcription start site located in intron V. We performed 5′RACE on mouse fibroblasts and isolated a new transcript of the sh3pxd2a gene encoding a novel Tks5 isoform, that we named Tks5β. This novel isoform diverges from the long form of Tks5 in that it lacks the PX-domain, which confers affinity for phosphatidylinositol-3,4-bisphosphate. Instead, Tks5β has a short unique amino terminal sequence encoded by the newly discovered exon 6β; this exon includes a start codon located 29 bp from the 5'-end of exon 6. Tks5β mRNA is expressed in MEFs and all mouse adult tissues analyzed. Tks5β is a substrate for the Src tyrosine kinase and its expression is regulated through the proteasome degradation pathway. Together, these findings indicate the essentiality of the larger Tks5 isoform for correct mammalian development and the transcriptional complexity of the sh3pxd2a gene.
Collapse
|
41
|
Stylli SS, Luwor RB, Kaye AH, I STT, Hovens CM, Lock P. Expression of the adaptor protein Tks5 in human cancer: prognostic potential. Oncol Rep 2014; 32:989-1002. [PMID: 24993883 DOI: 10.3892/or.2014.3310] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/04/2014] [Indexed: 11/05/2022] Open
Abstract
Tks5 (tyrosine kinase substrate with 5 SH3 domains) is an adaptor protein which cooperates with Src tyrosine kinase to promote the formation of protease-enriched, actin-based projections known as invadopodia, which are utilized by invasive cancer cells to degrade the extracellular matrix (ECM). We previously identified a Src-Tks5-Nck pathway which promotes invadopodium formation and ECM proteolysis in melanoma and breast cancer cells. We therefore sought to investigate the significance of Tks5 expression in human cancers. This was undertaken retrospectively through an immunohistochemical evaluation in tissue microarray cores and through data mining of the public database, Oncomine. Here we showed that Tks5 was expressed at higher levels in the microarray cores of breast, colon, lung and prostate cancer tissues compared to the levels in normal tissues. Importantly, mining of Oncomine datasets revealed a strong correlation between Tks5 mRNA overexpression in a number of cancers with increased metastatic events and a poorer prognosis. Collectively, these findings suggest a clinical association of Tks5 expression in human cancers. It identifies the importance for further investigations in examining the full potential of Tks5 as a relevant prognostic marker in a select number of cancers which may have implications for future targeted therapies.
Collapse
Affiliation(s)
- Stanley S Stylli
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3052, Australia
| | - Rodney B Luwor
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3052, Australia
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3052, Australia
| | - Stacey T T I
- Department of Biochemistry, La Trobe Institute of Molecular Sciences, La Trobe University, Bundoora, Victoria 3086, Australia
| | - Christopher M Hovens
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Peter Lock
- Department of Biochemistry, La Trobe Institute of Molecular Sciences, La Trobe University, Bundoora, Victoria 3086, Australia
| |
Collapse
|
42
|
Abstract
The occurrence of invadopodia has been, since its characterization, a hallmark of cancerous cell invasion and metastasis. These structures are now the subject of a controversy concerning their cellular function, molecular regulation, and assembly. The terms invadopodia and podosomes have been used interchangeably since their discovery back in 1980. Since then, these phenotypes are now more established and accepted by the scientific community as vital structures for 3D cancer cell motility. Many characteristics relating to invadopodia and podosomes have been elucidated, which might prove these structures as good targets for metastasis treatment. In this review, we briefly review the actin reorganization process needed in most types of cancer cell motility. We also review the important characteristics of invadopodia, including molecular components, assembly, markers, and the signaling pathways, providing a comprehensive model for invadopodia regulation.
Collapse
Affiliation(s)
- Bechara A Saykali
- Department of Natural Sciences, The Lebanese American University , Beirut , Lebanon
| | | |
Collapse
|
43
|
RIPK1- and RIPK3-induced cell death mode is determined by target availability. Cell Death Differ 2014; 21:1600-12. [PMID: 24902899 DOI: 10.1038/cdd.2014.70] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 04/02/2014] [Accepted: 04/15/2014] [Indexed: 12/25/2022] Open
Abstract
Both receptor-interacting protein kinase 1 (RIPK1) and RIPK3 can signal cell death following death receptor ligation. To study the requirements for RIPK-triggered cell death in the absence of death receptor signaling, we engineered inducible versions of RIPK1 and RIPK3 that can be activated by dimerization with the antibiotic coumermycin. In the absence of TNF or other death ligands, expression and dimerization of RIPK1 was sufficient to cause cell death by caspase- or RIPK3-dependent mechanisms. Dimerized RIPK3 induced cell death by an MLKL-dependent mechanism but, surprisingly, also induced death mediated by FADD, caspase 8 and RIPK1. Catalytically active RIPK3 kinase domains were essential for MLKL-dependent but not for caspase 8-dependent death. When RIPK1 or RIPK3 proteins were dimerized, the mode of cell death was determined by the availability of downstream molecules such as FADD, caspase 8 and MLKL. These observations imply that rather than a 'switch' operating between the two modes of cell death, the final mechanism depends on levels of the respective signaling and effector proteins.
Collapse
|
44
|
Phosphosite mapping of HIP-55 protein in mammalian cells. Int J Mol Sci 2014; 15:4903-14. [PMID: 24651461 PMCID: PMC3975430 DOI: 10.3390/ijms15034903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 02/20/2014] [Accepted: 03/07/2014] [Indexed: 11/16/2022] Open
Abstract
In the present study, hematopoietic progenitor kinase 1 (HPK1)-interacting protein of 55 kDa (HIP-55) protein was over-expressed in HEK293 cells, which was genetically attached with 6x His tag. The protein was purified by nickel-charged resin and was then subjected to tryptic digestion. The phosphorylated peptides within the HIP-55 protein were enriched by TiO2 affinity chromatography, followed by mass spectrometry analysis. Fourteen phosphorylation sites along the primary structure of HIP-55 protein were identified, most of which had not been previously reported. Our results indicate that bio-mass spectrometry coupled with manual interpretation can be used to successfully identify the phosphorylation modification in HIP-55 protein in HEK293 cells.
Collapse
|
45
|
Burger KL, Learman BS, Boucherle AK, Sirintrapun SJ, Isom S, Díaz B, Courtneidge SA, Seals DF. Src-dependent Tks5 phosphorylation regulates invadopodia-associated invasion in prostate cancer cells. Prostate 2014; 74:134-48. [PMID: 24174371 PMCID: PMC4083496 DOI: 10.1002/pros.22735] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 09/05/2013] [Indexed: 11/07/2022]
Abstract
BACKGROUND The Src tyrosine kinase substrate and adaptor protein Tks5 had previously been implicated in the invasive phenotype of normal and transformed cell types via regulation of cytoskeletal structures called podosomes/invadopodia. The role of Src-Tks5 signaling in invasive prostate cancer, however, had not been previously evaluated. METHODS We measured the relative expression of Tks5 in normal (n = 20) and cancerous (n = 184, from 92 patients) prostate tissue specimens by immunohistochemistry using a commercially available tumor microarray. We also manipulated the expression and activity of wild-type and mutant Src and Tks5 constructs in the LNCaP and PC-3 prostate cancer cell lines in order to ascertain the role of Src-Tks5 signaling in invadopodia development, matrix-remodeling activity, motility, and invasion. RESULTS Our studies demonstrated that Src was activated and Tks5 upregulated in high Gleason score prostate tumor specimens and in invasive prostate cancer cell lines. Remarkably, overexpression of Tks5 in LNCaP cells was sufficient to induce invadopodia formation and associated matrix degradation. This Tks5-dependent increase in invasive behavior further depended on Src tyrosine kinase activity and the phosphorylation of Tks5 at tyrosine residues 557 and 619. In PC-3 cells we demonstrated that Tks5 phosphorylation at these sites was necessary and sufficient for invadopodia-associated matrix degradation and invasion. CONCLUSIONS Our results suggest a general role for Src-Tks5 signaling in prostate tumor progression and the utility of Tks5 as a marker protein for the staging of this disease.
Collapse
Affiliation(s)
- Karen L. Burger
- Department of Cancer Biology, Wake Forest Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Brian S. Learman
- Department of Cancer Biology, Wake Forest Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Amy K. Boucherle
- Department of Cancer Biology, Wake Forest Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - S. Joseph Sirintrapun
- Department of Pathology, Wake Forest Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Scott Isom
- Department of Biostatistical Sciences, Wake Forest Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Begoña Díaz
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Sara A. Courtneidge
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Darren F. Seals
- Department of Biology, Appalachian State University, Boone, North Carolina
| |
Collapse
|
46
|
Li CMC, Chen G, Dayton TL, Kim-Kiselak C, Hoersch S, Whittaker CA, Bronson RT, Beer DG, Winslow MM, Jacks T. Differential Tks5 isoform expression contributes to metastatic invasion of lung adenocarcinoma. Genes Dev 2013; 27:1557-67. [PMID: 23873940 DOI: 10.1101/gad.222745.113] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Metastasis accounts for the vast majority of cancer-related deaths, yet the molecular mechanisms that drive metastatic spread remain poorly understood. Here we report that Tks5, which has been linked to the formation of proteolytic cellular protrusions known as invadopodia, undergoes an isoform switch during metastatic progression in a genetically engineered mouse model of lung adenocarcinoma. Nonmetastatic primary tumor-derived cells predominantly expressed a short isoform, Tks5short, while metastatic primary tumor- and metastasis-derived cells acquired increased expression of the full-length isoform Tks5long. This elevation of Tks5long to Tks5short ratio correlated with a commensurate increase in invadopodia activity in metastatic cells compared with nonmetastatic cells. Further characterization of these isoforms by knockdown and overexpression experiments demonstrated that Tks5long promoted invadopodia in vitro and increased metastasis in transplant models and an autochthonous model of lung adenocarcinoma. Conversely, Tks5short decreased invadopodia stability and proteolysis, acting as a natural dominant-negative inhibitor to Tks5long. Importantly, high Tks5long and low Tks5short expressions in human lung adenocarcinomas correlated with metastatic disease and predicted worse survival of early stage patients. These data indicate that tipping the Tks5 isoform balance to a high Tks5long to Tks5short ratio promotes invadopodia-mediated invasion and metastasis.
Collapse
Affiliation(s)
- Carman Man-Chung Li
- David H. Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Fekete A, Bőgel G, Pesti S, Péterfi Z, Geiszt M, Buday L. EGF regulates tyrosine phosphorylation and membrane-translocation of the scaffold protein Tks5. J Mol Signal 2013; 8:8. [PMID: 23924390 PMCID: PMC3765130 DOI: 10.1186/1750-2187-8-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 08/06/2013] [Indexed: 02/07/2023] Open
Abstract
Background Tks5/FISH is a scaffold protein comprising of five SH3 domains and one PX domain. Tks5 is a substrate of the tyrosine kinase Src and is required for the organization of podosomes/invadopodia implicated in invasion of tumor cells. Recent data have suggested that a close homologue of Tks5, Tks4, is implicated in the EGF signaling. Results Here, we report that Tks5 is a component of the EGF signaling pathway. In EGF-treated cells, Tks5 is tyrosine phosphorylated within minutes and the level of phosphorylation is sustained for at least 2 hours. Using specific kinase inhibitors, we demonstrate that tyrosine phosphorylation of Tks5 is catalyzed by Src tyrosine kinase. We show that treatment of cells with EGF results in plasma membrane translocation of Tks5. In addition, treatment of cells with LY294002, an inhibitor of PI 3-kinase, or mutation of the PX domain reduces tyrosine phosphorylation and membrane translocation of Tks5. Conclusions Our results identify Tks5 as a novel component of the EGF signaling pathway.
Collapse
Affiliation(s)
- Anna Fekete
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences, Budapest 1113, Hungary
| | - Gábor Bőgel
- Department of Medical Chemistry, Semmelweis University Medical School, Budapest 1094, Hungary
| | - Szabolcs Pesti
- Department of Medical Chemistry, Semmelweis University Medical School, Budapest 1094, Hungary
| | - Zalán Péterfi
- Department of Physiology, Semmelweis University Medical School, Budapest, Hungary
| | - Miklós Geiszt
- Department of Physiology, Semmelweis University Medical School, Budapest, Hungary ; "Lendület" Peroxidase Enzyme Research Group of the Semmelweis University and the Hungarian Academy of Sciences, Budapest 1094, Hungary
| | - László Buday
- Institute of Enzymology, Research Center for Natural Sciences, Hungarian Academy of Sciences, Budapest 1113, Hungary ; Department of Medical Chemistry, Semmelweis University Medical School, Budapest 1094, Hungary
| |
Collapse
|
48
|
Abstract
Remodeling of extracellular matrix (ECM) is a fundamental cell property that allows cells to alter their microenvironment and move through tissues. Invadopodia and podosomes are subcellular actin-rich structures that are specialized for matrix degradation and are formed by cancer and normal cells, respectively. Although initial studies focused on defining the core machinery of these two structures, recent studies have identified inputs from both growth factor and adhesion signaling as crucial for invasive activity. This Commentary will outline the current knowledge on the upstream signaling inputs to invadopodia and podosomes and their role in governing distinct stages of these invasive structures. We discuss invadopodia and podosomes as adhesion structures and highlight new data showing that invadopodia-associated adhesion rings promote the maturation of already-formed invadopodia. We present a model in which growth factor stimulation leads to phosphoinositide 3-kinase (PI3K) activity and formation of invadopodia, whereas adhesion signaling promotes exocytosis of proteinases at invadopodia.
Collapse
Affiliation(s)
- Daisuke Hoshino
- Department of Cancer Biology, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, TN 37232-6840, USA
| | | | | |
Collapse
|
49
|
Amelio I, Lena AM, Viticchiè G, Shalom-Feuerstein R, Terrinoni A, Dinsdale D, Russo G, Fortunato C, Bonanno E, Spagnoli LG, Aberdam D, Knight RA, Candi E, Melino G. miR-24 triggers epidermal differentiation by controlling actin adhesion and cell migration. ACTA ACUST UNITED AC 2013; 199:347-63. [PMID: 23071155 PMCID: PMC3471232 DOI: 10.1083/jcb.201203134] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A differentiation-promoting micro-RNA regulates actin cable dynamics, intercellular adhesion, and cell migration in human and mouse epidermis. During keratinocyte differentiation and stratification, cells undergo extensive remodeling of their actin cytoskeleton, which is important to control cell mobility and to coordinate and stabilize adhesive structures necessary for functional epithelia. Limited knowledge exists on how the actin cytoskeleton is remodeled in epithelial stratification and whether cell shape is a key determinant to trigger terminal differentiation. In this paper, using human keratinocytes and mouse epidermis as models, we implicate miR-24 in actin adhesion dynamics and demonstrate that miR-24 directly controls actin cable formation and cell mobility. miR-24 overexpression in proliferating cells was sufficient to trigger keratinocyte differentiation both in vitro and in vivo and directly repressed cytoskeletal modulators (PAK4, Tks5, and ArhGAP19). Silencing of these targets recapitulated the effects of miR-24 overexpression. Our results uncover a new regulatory pathway involving a differentiation-promoting microribonucleic acid that regulates actin adhesion dynamics in human and mouse epidermis.
Collapse
Affiliation(s)
- Ivano Amelio
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Siddiqui TA, Lively S, Vincent C, Schlichter LC. Regulation of podosome formation, microglial migration and invasion by Ca(2+)-signaling molecules expressed in podosomes. J Neuroinflammation 2012; 9:250. [PMID: 23158496 PMCID: PMC3551664 DOI: 10.1186/1742-2094-9-250] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 10/25/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Microglia migrate during brain development and after CNS injury, but it is not known how they degrade the extracellular matrix (ECM) to accomplish this. Podosomes are tiny structures with the unique ability to adhere to and dissolve ECM. Podosomes have a two-part architecture: a core that is rich in F-actin and actin-regulatory molecules (for example, Arp2/3), surrounded by a ring with adhesion and structural proteins (for example, talin, vinculin). We recently discovered that the lamellum at the leading edge of migrating microglia contains a large F-actin-rich superstructure ('podonut') composed of many podosomes. Microglia that expressed podosomes could degrade ECM molecules. Finely tuned Ca(2+) signaling is important for cell migration, cell-substrate adhesion and contraction of the actomyosin network. Here, we hypothesized that podosomes contain Ca(2+)-signaling machinery, and that podosome expression and function depend on Ca(2+) influx and specific ion channels. METHODS High-resolution immunocytochemistry was used on rat microglia to identify podosomes and novel molecular components. A pharmacological toolbox was applied to functional assays. We analyzed roles of Ca(2+)-entry pathways and ion channels in podosome expression, microglial migration into a scratch-wound, transmigration through pores in a filter, and invasion through Matrigel™-coated filters. RESULTS Microglial podosomes were identified using well-known components of the core (F-actin, Arp2) and ring (talin, vinculin). We discovered four novel podosome components related to Ca(2+) signaling. The core contained calcium release activated calcium (CRAC; Orai1) channels, calmodulin, small-conductance Ca(2+)-activated SK3 channels, and ionized Ca(2+) binding adapter molecule 1 (Iba1), which is used to identify microglia in the CNS. The Orai1 accessory molecule, STIM1, was also present in and around podosomes. Podosome formation was inhibited by removing external Ca(2+) or blocking CRAC channels. Blockers of CRAC channels inhibited migration and invasion, and SK3 inhibition reduced invasion. CONCLUSIONS Microglia podosome formation, migration and/or invasion require Ca(2+) influx, CRAC, and SK3 channels. Both channels were present in microglial podosomes along with the Ca(2+)-regulated molecules, calmodulin, Iba1 and STIM1. These results suggest that the podosome is a hub for sub-cellular Ca(2+)-signaling to regulate ECM degradation and cell migration. The findings have broad implications for understanding migration mechanisms of cells that adhere to, and dissolve ECM.
Collapse
Affiliation(s)
- Tamjeed A Siddiqui
- Toronto Western Research Institute, 399 Bathurst Street, Toronto, Ontario, M5T 2S8, Canada
| | | | | | | |
Collapse
|