51
|
Wei CY, Zhu MX, Yang YW, Zhang PF, Yang X, Peng R, Gao C, Lu JC, Wang L, Deng XY, Lu NH, Qi FZ, Gu JY. Downregulation of RNF128 activates Wnt/β-catenin signaling to induce cellular EMT and stemness via CD44 and CTTN ubiquitination in melanoma. J Hematol Oncol 2019; 12:21. [PMID: 30832692 PMCID: PMC6399928 DOI: 10.1186/s13045-019-0711-z] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/21/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Ring finger proteins (RNFs) were involved in carcinogenesis. Here, we aimed to explore the detailed mechanism of RNF128 in the progression of melanoma. METHODS We reanalyzed several gene expression profiles from the Gene Expression Omnibus (GEO) database and obtained the overlapped differential expressed RNF genes. Among them, RNF128 was selected to further explore its expression, the biological significance, and the underlying molecular mechanism, as well as the clinical relevance in melanoma patients. RESULTS RNF128 was found to be significantly downregulated in the selected datasets, which was further verified in our melanoma tissues. Moreover, RNF128 downregulation was shown to correlate with the malignant phenotype of melanoma, and further functional assays demonstrated that low levels of RNF128 promoted melanoma progression via inducing cell epithelial-mesenchymal transition (EMT) and the acquisition of stemness. Mechanistically, RNF128 interference activated the Wnt pathway via simultaneously ubiquitinating CD44/cortactin (CTTN), resulting in CD44 and c-Myc transcription, thus revealed that RNF128 participated in a positive feedback of the Wnt pathway-CD44 loop. Clinically, we found that patients expressing low RNF128 and high CD44/CTTN levels had a poor prognosis. CONCLUSION Downregulated RNF128 activates Wnt signaling to induce cellular EMT and stemness by ubiquitinating and degrading CD44/CTTN, and RNF128 is a reliable diagnostic and prognostic biomarker, and a deeper understanding of RNF128 may contribute to the treatment of melanoma.
Collapse
Affiliation(s)
- Chuan-Yuan Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.,Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, People's Republic of China
| | - Meng-Xuan Zhu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yan-Wen Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Peng-Fei Zhang
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200032, People's Republic of China
| | - Xuan Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, People's Republic of China
| | - Rui Peng
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, People's Republic of China
| | - Chao Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jia-Cheng Lu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, People's Republic of China
| | - Lu Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xin-Yi Deng
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Nan-Hang Lu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Fa-Zhi Qi
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jian-Ying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
52
|
ProNGF increases breast tumor aggressiveness through functional association of TrkA with EphA2. Cancer Lett 2019; 449:196-206. [PMID: 30771434 DOI: 10.1016/j.canlet.2019.02.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/07/2019] [Accepted: 02/10/2019] [Indexed: 12/19/2022]
Abstract
ProNGF expression has been linked to several types of cancers including breast cancer, and we have previously shown that proNGF stimulates breast cancer invasion in an autocrine manner through membrane receptors sortilin and TrkA. However, little is known regarding TrkA-associated protein partners upon proNGF stimulation. By proteomic analysis and proximity ligation assays, we found that proNGF binding to sortilin induced sequential formation of the functional sortilin/TrkA/EphA2 complex, leading to TrkA-phosphorylation dependent Akt activation and EphA2-dependent Src activation. EphA2 inhibition using siRNA approach abolished proNGF-stimulated clonogenic growth of breast cancer cell lines. Combinatorial targeting of TrkA and EphA2 dramatically reduced colony formation in vitro, primary tumor growth and metastatic dissemination towards the brain in vivo. Finally, proximity ligation assay in breast tumor samples revealed that increased TrkA/EphA2 proximity ligation assay signals were correlated with a decrease of overall survival in patients. All together, these data point out the importance of TrkA/EphA2 functional association in proNGF-induced tumor promoting effects, and provide a rationale to target proNGF/TrkA/EphA2 axis by alternative methods other than the simple use of tyrosine kinase inhibitors in breast cancer.
Collapse
|
53
|
Zhao M, Zhou Q, He C, Zhang Y, Wang Z, Cai R, Ma C, Li Y, Wang X, Zhan L. Stored red blood cells enhance in vivo migration of dendritic cells by promoting reactive oxygen species-induced cytoskeletal rearrangement. Transfusion 2019; 59:1312-1323. [PMID: 30614543 DOI: 10.1111/trf.15123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 10/27/2018] [Accepted: 10/28/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND A complex array of physicochemical changes occurs in red blood cells (RBCs) during storage, leading to enhanced posttransfusion clearance. Dendritic cells (DCs) play crucial roles in the engulfment of aged RBCs; however, it is unclear how stored RBCs (sRBCs) modulate their responses to inflammatory stimuli and DC migration ability. STUDY DESIGN AND METHODS In this study, we examined whether sRBCs affect the migration ability of DCs and elucidated the detailed mechanisms mediating this process. Murine RBCs were incubated with marrow DCs after removing the storage supernatant. The effects of sRBCs on cytokine secretion from DCs, surface marker expression, and homing ability were examined. RESULTS More sRBCs were internalized by DCs than fresh RBCs (fRBCs), and RBC accumulation significantly promoted the expression of allostimulatory molecules and the secretion of Th1-type cytokines in the presence of lipopolysaccharide (LPS). In particular, the lymphoid-tissue homing ability of transfused DCs treated with sRBCs (sRBC-DCs) was also significantly greater than that of fRBCs. Up regulation of CCR7 and improved organization of the cytoskeleton were observed in sRBC-DCs, and blocking Rho/Rho-associated protein kinase (ROCK), PI3K/Akt, and NF-κB pathways greatly hindered cytoskeletal rearrangement. Moreover, high levels of reactive oxygen species (ROS) were detected in sRBC-DCs, and treatment with N-acetylcysteine simultaneously decreased the lymph node-homing ability of DCs and phosphorylation of RhoA, ROCK1, and cortactin. CONCLUSIONS sRBCs initiated differential immune responses compared to fRBCs, and the presence of LPS augmented this phenomenon. Up regulation of CCR7 and ROS production promotes cytoskeletal reorganization and contributes to the increased homing of sRBCs-DCs.
Collapse
Affiliation(s)
- Man Zhao
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China.,Department of Blood Transfusion, Chinese PLA General Hospital, Beijing, China
| | - Qianqian Zhou
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China
| | - Chulin He
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China
| | - Yulong Zhang
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China
| | - Zhengjun Wang
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China
| | - Ruiying Cai
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China
| | - Cong Ma
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China
| | - Yuan Li
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China
| | - Xiaohui Wang
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China
| | - Linsheng Zhan
- Beijing Key Laboratory of Blood Safety and Security, Institute of Health Service and Transfusion Medicine, Beijing, P.R. China.,Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
54
|
Markwell SM, Ammer AG, Interval ET, Allen JL, Papenberg BW, Hames RA, Castaño JE, Schafer DA, Weed SA. Cortactin Phosphorylation by Casein Kinase 2 Regulates Actin-Related Protein 2/3 Complex Activity, Invadopodia Function, and Tumor Cell Invasion. Mol Cancer Res 2019; 17:987-1001. [PMID: 30610108 DOI: 10.1158/1541-7786.mcr-18-0391] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 09/18/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022]
Abstract
Malregulation of the actin cytoskeleton enhances tumor cell motility and invasion. The actin-binding protein cortactin facilitates branched actin network formation through activation of the actin-related protein (Arp) 2/3 complex. Increased cortactin expression due to gene amplification is observed in head and neck squamous cell carcinoma (HNSCC) and other cancers, corresponding with elevated tumor progression and poor patient outcome. Arp2/3 complex activation is responsible for driving increased migration and extracellular matrix (ECM) degradation by governing invadopodia formation and activity. Although cortactin-mediated activation of Arp2/3 complex and invadopodia regulation has been well established, signaling pathways responsible for governing cortactin binding to Arp2/3 are unknown and potentially present a new avenue for anti-invasive therapeutic targeting. Here we identify casein kinase (CK) 2α phosphorylation of cortactin as a negative regulator of Arp2/3 binding. CK2α directly phosphorylates cortactin at a conserved threonine (T24) adjacent to the canonical Arp2/3 binding motif. Phosphorylation of cortactin T24 by CK2α impairs the ability of cortactin to bind Arp2/3 and activate actin nucleation. Decreased invadopodia activity is observed in HNSCC cells with expression of CK2α phosphorylation-null cortactin mutants, shRNA-mediated CK2α knockdown, and with the CK2α inhibitor Silmitasertib. Silmitasertib inhibits HNSCC collective invasion in tumor spheroids and orthotopic tongue tumors in mice. Collectively these data suggest that CK2α-mediated cortactin phosphorylation at T24 is critical in regulating cortactin binding to Arp2/3 complex and pro-invasive activity, identifying a potential targetable mechanism for impairing HNSCC invasion. IMPLICATIONS: This study identifies a new signaling pathway that contributes to enhancing cancer cell invasion.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/17/4/987/F1.large.jpg.
Collapse
Affiliation(s)
- Steven M Markwell
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - Amanda G Ammer
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - Erik T Interval
- Department of Otolaryngology, Head and Neck Surgery, West Virginia University, Morgantown, West Virginia
| | - Jessica L Allen
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - Brenen W Papenberg
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - River A Hames
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia
| | - Johnathan E Castaño
- Department of Otolaryngology, Head and Neck Surgery, West Virginia University, Morgantown, West Virginia
| | - Dorothy A Schafer
- Department of Biology, University of Virginia, Charlottesville, Virginia
| | - Scott A Weed
- Program in Cancer Cell Biology, Department of Biochemistry, West Virginia University, Morgantown, West Virginia.
| |
Collapse
|
55
|
Ramos‐García P, González‐Moles MÁ, Ayén Á, González‐Ruiz L, Ruiz‐Ávila I, Gil‐Montoya JA. Prognostic and clinicopathological significance of
CTTN
/cortactin alterations in head and neck squamous cell carcinoma: Systematic review and meta‐analysis. Head Neck 2018; 41:1963-1978. [DOI: 10.1002/hed.25632] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/29/2018] [Accepted: 12/13/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
| | | | - Ángela Ayén
- School of MedicineUniversity of Granada Granada Spain
| | - Lucía González‐Ruiz
- Servicio de DermatologíaHospital General Universitario de Ciudad Real Ciudad Real Spain
| | - Isabel Ruiz‐Ávila
- Instituto de Investigación Biosanitaria Granada Spain
- Servicio de Anatomía PatológicaComplejo Hospitalario Universitario de Granada Granada Spain
| | - José Antonio Gil‐Montoya
- School of DentistryUniversity of Granada Granada Spain
- Instituto de Investigación Biosanitaria Granada Spain
| |
Collapse
|
56
|
Hasan MK, Rassenti L, Widhopf GF, Yu J, Kipps TJ. Wnt5a causes ROR1 to complex and activate cortactin to enhance migration of chronic lymphocytic leukemia cells. Leukemia 2018; 33:653-661. [PMID: 30568170 PMCID: PMC6462876 DOI: 10.1038/s41375-018-0306-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 09/15/2018] [Accepted: 11/14/2018] [Indexed: 01/29/2023]
Abstract
Chronic lymphocytic leukemia cells (CLL) migrate between the blood and lymphoid tissues in response to chemokines. Such migration requires structured cytoskeletal-actin polymerization, which may involve the protein cortactin. We discovered that treatment of CLL cells with Wnt5a causes Receptor tyosin kinase-like orphan receptor 1 (ROR1) to bind cortactin, which undergoes tyrosine phosphorylation at Y421, recruits ARHGEF1, and activates RhoA, thereby enhancing leukemia-cell migration; such effects could be inhibited by cirmtuzumab, a humanized mAb specific for ROR1. We transfected the CLL-cell-line MEC1 with either full-length ROR1 or various mutant forms of ROR1 to examine the structural features required for binding cortactin. We found that the proline-rich domain (PRD) was necessary for ROR1 to recruit cortactin. We generated MEC1 cells that each expressed a mutant form of ROR1 with a single amino-acid substitution of alanine (A) for proline (P) in potential SH3-binding sites in the ROR1-PRD at positions 784, 808, 826, 841, or 850. In contrast to wild-type ROR1, or other ROR1P=>A mutants, ROR1P(841)A failed to complex with cortactin or ARHGEF1 in response to Wnt5a. Moreover, Wnt5a could not induce MEC1-ROR1P(841)A to phosphorylate cortactin or enhance CLL-cell F-actin polymerization. Taken together, these studies show that cortactin plays an important role in ROR1-dependent Wnt5a-enhanced CLL-cell migration.
Collapse
Affiliation(s)
- Md Kamrul Hasan
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Laura Rassenti
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - George F Widhopf
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Jian Yu
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Thomas J Kipps
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
57
|
Abe T, La TM, Miyagaki Y, Oya E, Wei FY, Sumida K, Fujise K, Takeda T, Tomizawa K, Takei K, Yamada H. Phosphorylation of cortactin by cyclin-dependent kinase 5 modulates actin bundling by the dynamin 1-cortactin ring-like complex and formation of filopodia and lamellipodia in NG108-15 glioma-derived cells. Int J Oncol 2018; 54:550-558. [PMID: 30570111 PMCID: PMC6317663 DOI: 10.3892/ijo.2018.4663] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/09/2018] [Indexed: 12/14/2022] Open
Abstract
Dynamin copolymerizes with cortactin to form a ring-like complex that bundles and stabilizes actin filaments. Actin bundle formation is crucial for generation of filopodia and lamellipodia, which guide migration, invasion, and metastasis of cancer cells. However, it is unknown how the dynamin-cortactin complex regulates actin bundle formation. The present study investigated phosphorylation of cortactin by cyclin-dependent kinase 5 (CDK5) and its effect on actin bundle formation by the dynamin-cortactin complex. CDK5 directly phosphorylated cortactin at T145/T219 in vitro. Phosphomimetic mutants in which one or both of these threonine residues was substituted by aspartate were used. The three phosphomimetic mutants (T145D, T219D and T145DT219D) had a decreased affinity for F-actin. Furthermore, electron microscopy demonstrated that these phosphomimetic mutants could not form a ring-like complex with dynamin 1. Consistently, the dynamin 1-phosphomimetic cortactin complexes exhibited decreased actin-bundling activity. Expression of the phosphomimetic mutants resulted in not only aberrant lamellipodia and short filopodia but also cell migration in NG108-15 glioma-derived cells. These results indicate that phosphorylation of cortactin by CDK5 regulates formation of lamellipodia and filopodia by modulating dynamin 1/cortactin-dependent actin bundling. Taken together, these findings suggest that CDK5 is a potential molecular target for anticancer therapy.
Collapse
Affiliation(s)
- Tadashi Abe
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - The Mon La
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Yuuzi Miyagaki
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Eri Oya
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Fan-Yan Wei
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kento Sumida
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Kenshiro Fujise
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Tetsuya Takeda
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Kazuhito Tomizawa
- Department of Molecular Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kohji Takei
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hiroshi Yamada
- Department of Neuroscience, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
58
|
Horn D, Gross M, Dyckhoff G, Fuchs J, Grabe N, Weichert W, Herpel E, Herold‐Mende C, Lichter P, Hoffmann J, Hess J, Freier K. Cortactin expression: Association with disease progression and survival in oral squamous cell carcinoma. Head Neck 2018; 40:2685-2694. [DOI: 10.1002/hed.25515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/21/2018] [Accepted: 10/06/2018] [Indexed: 01/01/2023] Open
Affiliation(s)
- Dominik Horn
- Department of Oral and Cranio‐Maxillofacial SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| | - Madeleine Gross
- Department of Oral and Cranio‐Maxillofacial SurgeryUniversity Hospital Heidelberg Heidelberg Germany
- Division of Molecular GeneticsGerman Cancer Research Center (DKFZ) Heidelberg Germany
| | - Gerhard Dyckhoff
- Department of Otorhinolaryngology, Head and Neck SurgeryUniversity Hospital Heidelberg Heidelberg Germany
- Molecular Cell Biology Group, Department of Otorhinolaryngology, Head and Neck SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| | - Jennifer Fuchs
- Department of Oral and Cranio‐Maxillofacial SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| | - Niels Grabe
- Hamamatsu Tissue Imaging and Analysis Center (TIGA)BIOQUANT, University of Heidelberg Heidelberg Germany
| | - Wilko Weichert
- Institute of PathologyUniversity Hospital Heidelberg Heidelberg Germany
| | - Esther Herpel
- Institute of PathologyUniversity Hospital Heidelberg Heidelberg Germany
- Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg Germany
| | - Christel Herold‐Mende
- Molecular Cell Biology Group, Department of Otorhinolaryngology, Head and Neck SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| | - Peter Lichter
- Division of Molecular GeneticsGerman Cancer Research Center (DKFZ) Heidelberg Germany
| | - Jürgen Hoffmann
- Department of Oral and Cranio‐Maxillofacial SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck SurgeryUniversity Hospital Heidelberg Heidelberg Germany
- Research Group Molecular Mechanisms of Head and Neck TumorsGerman Cancer Research Center (DKFZ) Heidelberg Germany
| | - Kolja Freier
- Department of Oral and Cranio‐Maxillofacial SurgeryUniversity Hospital Heidelberg Heidelberg Germany
| |
Collapse
|
59
|
Amorim S, da Costa DS, Freitas D, Reis CA, Reis RL, Pashkuleva I, Pires RA. Molecular weight of surface immobilized hyaluronic acid influences CD44-mediated binding of gastric cancer cells. Sci Rep 2018; 8:16058. [PMID: 30375477 PMCID: PMC6207784 DOI: 10.1038/s41598-018-34445-0] [Citation(s) in RCA: 35] [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: 06/16/2017] [Accepted: 08/30/2018] [Indexed: 01/02/2023] Open
Abstract
The physiological importance of the interactions between hyaluronic acid (HA) and its main membrane receptor, CD44, in pathological processes, e.g. cancer, is well recognized. However, these interactions are mainly studied in solution, whereas HA in the extracellular matrix (ECM) is partially immobilized via its interactions with other ECM components. We therefore, developed substrates in which HA is presented in an ECM-relevant manner. We immobilized HA with different molecular weights (Mw) in a Layer-by-Layer (LbL) fashion and studied the interactions of the substrates with CD44 and two human gastric cancer cell lines that overexpress this receptor, namely AGS and MKN45. We demonstrate that MKN45 cells are more sensitive to the LbL substrates as compared with AGS. This difference is due to different CD44 expression: while CD44 is detected mainly in the cytoplasm of AGS, MKN45 express CD44 predominantly at the cell membrane where it is involved in the recognition and binding of HA. The invasiveness of the studied cell lines was also evaluated as a function of HA Mw. Invasive profile characterized by low cell adhesion, high cell motility, high expression of cortactin, formation of invadopodia and cell clusters was observed for MKN45 cells when they are in contact with substrates presenting HA of high Mw.
Collapse
Affiliation(s)
- Sara Amorim
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Diana Soares da Costa
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Daniela Freitas
- Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
- Department of Pathology and Oncology, Faculty of Medicine, Porto University, Porto, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Ricardo A Pires
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal.
| |
Collapse
|
60
|
Koya Y, Liu W, Yamakita Y, Senga T, Shibata K, Yamashita M, Nawa A, Kikkawa F, Kajiyama H. Hematopoietic lineage cell-specific protein 1 (HS1), a hidden player in migration, invasion, and tumor formation, is over-expressed in ovarian carcinoma cells. Oncotarget 2018; 9:32609-32623. [PMID: 30220969 PMCID: PMC6135686 DOI: 10.18632/oncotarget.25975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic lineage cell-specific protein 1 (HS1), which is the hematopoietic homolog of cortactin, is an actin-binding protein and Lyn substrate. It is upregulated in several cancers and its expression level is associated with increased cell migration, metastasis, and poor prognosis. Here we investigated the expression and roles of HS1 in ovarian carcinoma cells. We analyzed the expression of HS1 in 171 ovarian cancer specimens and determined the association between HS1 expression and clinicopathological characteristics, including patient outcomes. In patients with stage II-IV disease, positive HS1 expression was associated with significantly worse overall survival than negative expression (P < 0.05). HS1 was localized in invadopodia in some ovarian cancer cells and was required for invadopodia formation. Migration and invasion of ovarian cancer cells were suppressed by down-regulation of HS1, but increased in cells that over-expressed exogenous HS1. Furthermore, ovarian cancer cells that expressed HS1 shRNA exhibited reduced tumor formation in a mouse xenograft model. Finally, we found that tyrosine phosphorylation of HS1 was essential for cell migration and invasion. These findings show that HS1 is a useful biomarker for the prognosis of patients with ovarian carcinoma and is a critical regulator of cytoskeleton remodeling involved in cell migration and invasion.
Collapse
Affiliation(s)
- Yoshihiro Koya
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan.,Bell Research Center for Reproductive Health and Cancer, Nagoya, Aichi, Japan
| | - Wenting Liu
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan.,Bell Research Center for Reproductive Health and Cancer, Nagoya, Aichi, Japan
| | - Yoshihiko Yamakita
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan.,Bell Research Center for Reproductive Health and Cancer, Nagoya, Aichi, Japan
| | | | - Kiyosumi Shibata
- Department of Obstetrics and Gynecology, Fujita Health University, Banbuntane Hotokukai Hospital, Nakagawa-ku, Nagoya, Japan
| | - Mamoru Yamashita
- Bell Research Center for Reproductive Health and Cancer, Nagoya, Aichi, Japan
| | - Akihiro Nawa
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan.,Bell Research Center for Reproductive Health and Cancer, Nagoya, Aichi, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| |
Collapse
|
61
|
Belvitch P, Rizzo AN, Dudek SM. Cortactin in Atherosclerosis: Just Say NO. Arterioscler Thromb Vasc Biol 2018; 36:2278-2280. [PMID: 27879274 DOI: 10.1161/atvbaha.116.308497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Patrick Belvitch
- From the Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois College of Medicine, Chicago
| | - Alicia N Rizzo
- From the Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois College of Medicine, Chicago
| | - Steven M Dudek
- From the Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois College of Medicine, Chicago.
| |
Collapse
|
62
|
Ramos-García P, González-Moles MÁ, González-Ruiz L, Ayén Á, Ruiz-Ávila I, Navarro-Triviño FJ, Gil-Montoya JA. An update of knowledge on cortactin as a metastatic driver and potential therapeutic target in oral squamous cell carcinoma. Oral Dis 2018; 25:949-971. [PMID: 29878474 DOI: 10.1111/odi.12913] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/15/2018] [Accepted: 06/05/2018] [Indexed: 12/12/2022]
Abstract
Cortactin is a protein encoded by the CTTN gene, localized on chromosome band 11q13. As a result of the amplification of this band, an important event in oral carcinogenesis, CTTN is also usually amplified, promoting the frequent overexpression of cortactin. Cortactin enhances cell migration in oral cancer, playing a key role in the regulation of filamentous actin and of protrusive structures (invadopodia and lamellipodia) on the cell membrane that are necessary for the acquisition of a migratory phenotype. We also analyze a series of emerging functions that cortactin may exert in oral cancer (cell proliferation, angiogenesis, regulation of exosomes, and interactions with the tumor microenvironment). We review its molecular structure, its most important interactions (with Src, Arp2/3 complex, and SH3-binding partners), the regulation of its functions, and its specific oncogenic role in oral cancer. We explore the mechanisms of its overexpression in cancer, mainly related to genetic amplification. We analyze the prognostic implications of the oncogenic activation of cortactin in potentially malignant disorders and in head and neck cancer, where it appears to be relevant in the development of lymph node metastasis. Finally, we discuss its usefulness as a therapeutic target and suggest future research lines.
Collapse
Affiliation(s)
| | - Miguel Ángel González-Moles
- School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria, Granada, Spain
| | - Lucía González-Ruiz
- Servicio de Dermatología, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Ángela Ayén
- School of Medicine, University of Granada, Granada, Spain
| | - Isabel Ruiz-Ávila
- Instituto de Investigación Biosanitaria, Granada, Spain.,Servicio de Anatomía Patológica, Complejo Hospitalario Universitario de Granada, Granada, Spain
| | | | - José Antonio Gil-Montoya
- School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria, Granada, Spain
| |
Collapse
|
63
|
Castosa R, Martinez-Iglesias O, Roca-Lema D, Casas-Pais A, Díaz-Díaz A, Iglesias P, Santamarina I, Graña B, Calvo L, Valladares-Ayerbes M, Concha Á, Figueroa A. Hakai overexpression effectively induces tumour progression and metastasis in vivo. Sci Rep 2018; 8:3466. [PMID: 29472634 PMCID: PMC5823865 DOI: 10.1038/s41598-018-21808-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/12/2018] [Indexed: 12/31/2022] Open
Abstract
At early stages of carcinoma progression, epithelial cells undergo a program named epithelial-to-mesenchymal transition characterized by the loss of the major component of the adherens junctions, E-cadherin, which in consequence causes the disruption of cell-cell contacts. Hakai is an E3 ubiquitin-ligase that binds to E-cadherin in a phosphorylated-dependent manner and induces its degradation; thus modulating cell adhesions. Here, we show that Hakai expression is gradually increased in adenoma and in different TNM stages (I-IV) from colon adenocarcinomas compared to human colon healthy tissues. Moreover, we confirm that Hakai overexpression in epithelial cells drives transformation in cells, a mesenchymal and invasive phenotype, accompanied by the downregulation of E-cadherin and the upregulation of N-cadherin, and an increased proliferation and an oncogenic potential. More importantly, for the first time, we have studied the role of Hakai during cancer progression in vivo. We show that Hakai-transformed MDCK cells dramatically induce tumour growth and local invasion in nude mice and tumour cells exhibit a mesenchymal phenotype. Furthermore, we have detected the presence of micrometastasis in the lung mice, further confirming Hakai role during tumour metastasis in vivo. These results lead to the consideration of Hakai as a potential new therapeutic target to block tumour development and metastasis.
Collapse
Affiliation(s)
- Raquel Castosa
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Universidade da Coruña (UDC), Sergas, Spain
| | - Olaia Martinez-Iglesias
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Universidade da Coruña (UDC), Sergas, Spain
| | - Daniel Roca-Lema
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Universidade da Coruña (UDC), Sergas, Spain
| | - Alba Casas-Pais
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Universidade da Coruña (UDC), Sergas, Spain
| | - Andrea Díaz-Díaz
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Universidade da Coruña (UDC), Sergas, Spain
| | - Pilar Iglesias
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Universidade da Coruña (UDC), Sergas, Spain.,Pathology Department, INIBIC, CHUAC, Sergas, UDC, Spain
| | - Isabel Santamarina
- Clinical and Translational Oncology Group, INIBIC, CHUAC, Sergas, UDC, Spain
| | - Begoña Graña
- Clinical and Translational Oncology Group, INIBIC, CHUAC, Sergas, UDC, Spain
| | - Lourdes Calvo
- Clinical and Translational Oncology Group, INIBIC, CHUAC, Sergas, UDC, Spain
| | | | - Ángel Concha
- Pathology Department, INIBIC, CHUAC, Sergas, UDC, Spain
| | - Angélica Figueroa
- Epithelial Plasticity and Metastasis Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Universidade da Coruña (UDC), Sergas, Spain.
| |
Collapse
|
64
|
Cortactin: Cell Functions of A Multifaceted Actin-Binding Protein. Trends Cell Biol 2018; 28:79-98. [DOI: 10.1016/j.tcb.2017.10.009] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022]
|
65
|
Abstract
Actin remodeling plays an essential role in diverse cellular processes such as cell motility, vesicle trafficking or cytokinesis. The scaffold protein and actin nucleation promoting factor Cortactin is present in virtually all actin-based structures, participating in the formation of branched actin networks. It has been involved in the control of endocytosis, and vesicle trafficking, axon guidance and organization, as well as adhesion, migration and invasion. To migrate and invade through three-dimensional environments, cells have developed specialized actin-based structures called invadosomes, a generic term to designate invadopodia and podosomes. Cortactin has emerged as a critical regulator of invadosome formation, function and disassembly. Underscoring this role, Cortactin is frequently overexpressed in several types of invasive cancers. Herein we will review the roles played by Cortactin in these specific invasive structures.
Collapse
Affiliation(s)
- Pauline Jeannot
- CRCT INSERM UMR1037, Université Toulouse III Paul Sabatier , CNRS ERL5294, Toulouse, France.,Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester , Manchester M20 4BX, UK
| | - Arnaud Besson
- CRCT INSERM UMR1037, Université Toulouse III Paul Sabatier , CNRS ERL5294, Toulouse, France.,LBCMCP , Centre de Biologie Intégrative, Université de Toulouse , CNRS, UPS, Toulouse Cedex, France
| |
Collapse
|
66
|
Jeannot P, Besson A. [Invadopodia regulation: a new function for p27 in cellular invasion]. Med Sci (Paris) 2017; 33:1023-1025. [PMID: 29261483 DOI: 10.1051/medsci/20173312002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Pauline Jeannot
- Centre de recherches en cancérologie de Toulouse (CRCT), Inserm UMR1037, université Toulouse III Paul Sabatier, CNRS ERL5294, 31000 Toulouse, France
| | - Arnaud Besson
- Centre de recherches en cancérologie de Toulouse (CRCT), Inserm UMR1037, université Toulouse III Paul Sabatier, CNRS ERL5294, 31000 Toulouse, France
| |
Collapse
|
67
|
Li X, Tao Y, Murphy JW, Scherer AN, Lam TT, Marshall AG, Koleske AJ, Boggon TJ. The repeat region of cortactin is intrinsically disordered in solution. Sci Rep 2017; 7:16696. [PMID: 29196701 PMCID: PMC5711941 DOI: 10.1038/s41598-017-16959-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/19/2017] [Indexed: 01/14/2023] Open
Abstract
The multi-domain protein, cortactin, contains a 37-residue repeating motif that binds to actin filaments. This cortactin repeat region comprises 6½ similar copies of the motif and binds actin filaments. To better understand this region of cortactin, and its fold, we conducted extensive biophysical analysis. Size exclusion chromatography with multi-angle light scattering (SEC-MALS) reveals that neither constructs of the cortactin repeats alone or together with the adjacent helical region homo-oligomerize. Using circular dichroism (CD) we find that in solution the cortactin repeats resemble a coil-like intrinsically disordered protein. Small-angle X-ray scattering (SAXS) also indicates that the cortactin repeats are intrinsically unfolded, and the experimentally observed radius of gyration (Rg) is coincidental to that calculated by the program Flexible-Meccano for an unfolded peptide of this length. Finally, hydrogen-deuterium exchange mass spectrometry (HDX-MS) indicates that the domain contains limited hydrophobic core regions. These experiments therefore provide evidence that in solution the cortactin repeat region of cortactin is intrinsically disordered.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA.,Department of Pathology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Yeqing Tao
- Department of Chemistry, Florida State University, 600 W., College Avenue, Tallahassee, FL, 32306, USA.,Biopharmaceutical Analytical Sciences, Biopharm R&D, GlaxoSmithKline, 709 Swedeland Road, King of Prussia, PA, 19406, USA
| | - James W Murphy
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Alexander N Scherer
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA.,Yale MS & Proteomics Resource, Yale University, New Haven, CT, 06520, USA
| | - Alan G Marshall
- Department of Chemistry, Florida State University, 600 W., College Avenue, Tallahassee, FL, 32306, USA.,Ion Cyclotron Resonance Program, National High Magnetic Field Laboratory, 1800 E. Paul Dirac Dr., Tallahassee, FL, 32310, USA
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA
| | - Titus J Boggon
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA. .,Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06520, USA.
| |
Collapse
|
68
|
Martin-Piedra MA, Garzón I, Gómez-Sotelo A, Garcia-Abril E, Jaimes-Parra BD, López-Cantarero M, Alaminos M, Campos A. Generation and Evaluation of Novel Stromal Cell-Containing Tissue Engineered Artificial Stromas for the Surgical Repair of Abdominal Defects. Biotechnol J 2017; 12. [PMID: 28869335 DOI: 10.1002/biot.201700078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/26/2017] [Indexed: 12/25/2022]
Abstract
Repair of abdominal wall defects is one of the major clinical challenges in abdominal surgery. Most biomaterials are associated to infection and severe complications, making necessary safer and more biocompatible approaches. In the present work, the adequate mechanical properties of synthetic polymer meshes with tissue-engineered matrices containing stromal mesenchymal cells is combined to generate a novel cell-containing tissue-like artificial stroma (SCTLAS) for use in abdominal wall repair. SCTLAS consisting on fibrin-agarose hydrogels seeded with stromal cells and reinforced with commercial surgical meshes (SM) are evaluated in vitro and in vivo in animal models of abdominal wall defect. Inflammatory cells, collagen, and extracellular matrix (ECM) components are analyzed and compared with grafted SM. Use of SCTLAS results in less inflammation and less fibrosis than SM, with most ECM components being very similar to control abdominal wall tissues. Cell migration and ECM remodeling within SCTLAS is comparable to control tissues. The use of SCTLAS could contribute to reduce the side-effects associated to currently available SM and regenerated tissues are more similar to control abdominal wall tissues. Bioengineered SCTLAS could contribute to a safer treatment of abdominal wall defects with higher biocompatibility than currently available SM.
Collapse
Affiliation(s)
- Miguel A Martin-Piedra
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Spain, and Instituto de Investigación Biosanitaria ibs.GRANADA, Avda. de la Ilustración, 11, Granada 18016, Spain
| | - Ingrid Garzón
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Spain, and Instituto de Investigación Biosanitaria ibs.GRANADA, Avda. de la Ilustración, 11, Granada 18016, Spain
| | - Ana Gómez-Sotelo
- Division of General and Digestive Surgery, Valme University Hospital, Sevilla, Spain
| | | | - Boris D Jaimes-Parra
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Spain, and Instituto de Investigación Biosanitaria ibs.GRANADA, Avda. de la Ilustración, 11, Granada 18016, Spain
| | - Manuel López-Cantarero
- University Hospital Complex of Granada and Department of Surgery, University of Granada, Granada, Spain
| | - Miguel Alaminos
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Spain, and Instituto de Investigación Biosanitaria ibs.GRANADA, Avda. de la Ilustración, 11, Granada 18016, Spain
| | - Antonio Campos
- Tissue Engineering Group, Department of Histology, School of Medicine, University of Granada, Spain, and Instituto de Investigación Biosanitaria ibs.GRANADA, Avda. de la Ilustración, 11, Granada 18016, Spain
| |
Collapse
|
69
|
Yin M, Ma W, An L. Cortactin in cancer cell migration and invasion. Oncotarget 2017; 8:88232-88243. [PMID: 29152154 PMCID: PMC5675706 DOI: 10.18632/oncotarget.21088] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 08/29/2017] [Indexed: 12/20/2022] Open
Abstract
Cortactin, a substrate of sarcoma (Src) kinases, is an actin-binding protein that is involved in cytoskeletal regulation, and is frequently overexpressed in cancer cells. Binding to the actin related protein 2/3 (Arp2/3) complex stimulates cortactin activity, which promotes F-actin nucleation and assembly. Cortactin promotes cancer cell migration and invasion, and plays a pivotal role in invadopodia formation and extra cellular matrix degradation. Overexpression of cortactin, by amplification of the chromosomal band 11q13, increases tumor aggressiveness. In this review, we report on the current knowledge and potential mechanisms of action of cortactin as a critical mediator of cancer cell migration and invasion.
Collapse
Affiliation(s)
- Miao Yin
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Wenqing Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Liguo An
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
70
|
Amin EM, Liu Y, Deng S, Tan KS, Chudgar N, Mayo MW, Sanchez-Vega F, Adusumilli PS, Schultz N, Jones DR. The RNA-editing enzyme ADAR promotes lung adenocarcinoma migration and invasion by stabilizing FAK. Sci Signal 2017; 10:eaah3941. [PMID: 28928239 PMCID: PMC5771642 DOI: 10.1126/scisignal.aah3941] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Large-scale, genome-wide studies report that RNA binding proteins are altered in cancers, but it is unclear how these proteins control tumor progression. We found that the RNA-editing protein ADAR (adenosine deaminase acting on double-stranded RNA) acted as a facilitator of lung adenocarcinoma (LUAD) progression through its ability to stabilize transcripts encoding focal adhesion kinase (FAK). In samples from 802 stage I LUAD patients, increased abundance of ADAR at both the mRNA and protein level correlated with tumor recurrence. Knocking down ADAR in LUAD cells suppressed their mesenchymal properties, migration, and invasion in culture. Analysis of gene expression patterns in LUAD cells identified ADAR-associated enrichment of a subset of genes involved in cell migration pathways; among these, FAK is the most notable gene whose expression was increased in the presence of ADAR. Molecular analyses revealed that ADAR posttranscriptionally increased FAK protein abundance by binding to the FAK transcript and editing a specific intronic site that resulted in the increased stabilization of FAK mRNA. Pharmacological inhibition of FAK blocked ADAR-induced invasiveness of LUAD cells, suggesting a potential therapeutic application for LUAD that has a high abundance of ADAR.
Collapse
Affiliation(s)
- Elianna M Amin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yuan Liu
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Su Deng
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kay See Tan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Neel Chudgar
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marty W Mayo
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Francisco Sanchez-Vega
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Prasad S Adusumilli
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nikolaus Schultz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David R Jones
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
71
|
Bati-Ayaz G, Can A, Pesen-Okvur D. Cellular distribution of invadopodia is regulated by nanometer scale surface protein patterns. Eur J Cell Biol 2017; 96:673-684. [PMID: 28847588 DOI: 10.1016/j.ejcb.2017.08.001] [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: 07/17/2016] [Revised: 06/26/2017] [Accepted: 08/14/2017] [Indexed: 10/25/2022] Open
Abstract
Invadopodia are proteolytic structures formed by cancer cells. It is not known whether their cellular distribution can be regulated by the organization of the extracellular matrix or the organization of the golgi complex or whether they have an adhesion requirement. Here, we used electron beam lithography to fabricate fibronectin (FN) nanodots with isotropic and gradient micrometer scale spacings on K-casein and laminin backgrounds. Investigating cancer cells cultured on protein nanopatterns, we showed that (i) presence of FN nanodots on a K-casein background decreased percent of cells with neutral invadopodia polarization compared to FN control surfaces; (ii) presence of a gradient of FN nanodots on a K-casein background increased percent of cells with negative invadopodia polarization compared to FN control surfaces; (iii) polarization of the golgi complex was similar to that of invadopodia in agreement with a spatial link; (iv) local adhesion did not necessarily appear to be a prerequisite for invadopodia formation.
Collapse
Affiliation(s)
- Gizem Bati-Ayaz
- Izmir Institute of Technology, Graduate Program in Biotechnology and Bioengineering, Turkey
| | - Ali Can
- Izmir Institute of Technology, Graduate Program in Biotechnology and Bioengineering, Turkey
| | - Devrim Pesen-Okvur
- Izmir Institute of Technology, Department of Molecular Biology and Genetics, Turkey.
| |
Collapse
|
72
|
Pang B, Wu N, Guan R, Pang L, Li X, Li S, Tang L, Guo Y, Chen J, Sun D, Sun H, Dai J, Bai J, Ji G, Liu P, Liu A, Wang Q, Xiao S, Fu S, Jin Y. Overexpression of RCC2 Enhances Cell Motility and Promotes Tumor Metastasis in Lung Adenocarcinoma by Inducing Epithelial-Mesenchymal Transition. Clin Cancer Res 2017; 23:5598-5610. [PMID: 28606921 DOI: 10.1158/1078-0432.ccr-16-2909] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 04/25/2017] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Investigate the role of regulator of chromosome condensation 2 (RCC2) on lung adenocarcinoma (LUAD) metastasis.Experimental Design: Clinical specimens were used to assess the impact of RCC2 on LUAD metastasis. Mouse models, cytobiology, and molecular biology assays were performed to elucidate the function and underlying mechanisms of RCC2 in LUAD.Results: RCC2 expression was frequently increased in LUADs (88/122, 72.13%). It was confirmed by analysis of a larger cohort of TCGA RNA-seq data containing 488 LUADs and 58 normal lung tissues (P < 0.001). Importantly, increased level of RCC2 was significantly associated with T status of tumor (P = 0.002), lymph node metastasis (P = 0.004), and advanced clinical stage (P = 0.001). Patients with LUAD with higher expression of RCC2 had shorter overall survival. Cox regression analysis demonstrated that RCC2 was an independent poorer prognostic factor for patients with LUAD. Moreover, forced expression of RCC2 promoted intrapulmonary metastasis in vivo and significantly enhanced LUAD cell migration, invasion, and proliferation in vitro Further study found that RCC2 induced epithelial-mesenchymal transition (EMT) and also stimulated the expression of MMP-2 and MMP-9. In addition, RCC2 was able to activate JNK, while inhibition of JNK suppressed the effect of RCC2 on LUAD cell migration, invasion, EMT, and the expression of MMP-2 and MMP-9.Conclusions: RCC2 plays a pivotal role in LUAD metastasis by inducing EMT via activation of MAPK-JNK signaling. Clin Cancer Res; 23(18); 5598-610. ©2017 AACR.
Collapse
Affiliation(s)
- Bo Pang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Nan Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Rongwei Guan
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Lin Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinlei Li
- Department of Human Anatomy, Harbin Medical University, Harbin, China
| | - Su Li
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Liudi Tang
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Ying Guo
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jialei Chen
- Department of Thoracic Surgery, The Second Affiliated Clinical Hospital, Harbin Medical University, Harbin, China
| | - Donglin Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Haiming Sun
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jialin Dai
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Jing Bai
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Guohua Ji
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Peng Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - An Liu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Qiushi Wang
- Department of Thoracic Surgery, The Second Affiliated Clinical Hospital, Harbin Medical University, Harbin, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Songbin Fu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China. .,Key Laboratory of Medical Genetics (Harbin Medical University), Heilongjiang Higher Education Institutions, Harbin, China
| | - Yan Jin
- Laboratory of Medical Genetics, Harbin Medical University, Harbin, China. .,Key Laboratory of Medical Genetics (Harbin Medical University), Heilongjiang Higher Education Institutions, Harbin, China
| |
Collapse
|
73
|
Gangoda L, Mathivanan S. Cortactin enhances exosome secretion without altering cargo. J Cell Biol 2017; 214:129-31. [PMID: 27432895 PMCID: PMC4949455 DOI: 10.1083/jcb.201606131] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 01/18/2023] Open
Abstract
The role of cortactin, a regulator of late endosomal trafficking, in the biogenesis and secretion of exosomes is poorly understood. In this issue, Sinha et al. (2016. J. Cell Biol.http://dx.doi.org/10.1083/jcb.201601025) elucidate the role of cortactin as a positive regulator of late endosomal docking and exosome secretion.
Collapse
Affiliation(s)
- Lahiru Gangoda
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| |
Collapse
|
74
|
Cortactin and phosphorylated cortactin tyr421 and tyr466 expression in supraglottic laryngeal carcinomas and lymph node metastases. Int J Biol Markers 2017; 33:79-86. [DOI: 10.5301/ijbm.5000297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: The most important adverse prognostic factor for laryngeal squamous cell carcinoma (LSCC) is the presence of cervical lymph node metastases. The supraglottic area of the larynx is richly supplied with lymphatics, and 25%-75% of supraglottic carcinomas metastasize in neck lymph nodes. Cortactin is a multidomain protein related to actin cytoskeleton regulation, podosome and lamellipodia formation, integrin signaling, axon guidance and extracellular matrix degradation. Cortactin is involved in metastasis formation because of its role in cell mobility. The present study focused mainly on the role of cortactin and phosphorylated cortactin (residues tyr421 and tyr466) expression and subcellular localization in primary supraglottic LSCCs and their cervical lymph node metastases. Methods: The immunohistochemical expression of cortactin, p-Y466-cortactin and p-Y421-cortactin was assessed in 38 primary supraglottic LSCCs and 10 lymph node metastases. The statistical approach included bootstrapping analysis. Results: Despite a significantly higher expression of cortactin in carcinoma cells than in adjacent normal laryngeal mucosa, no associations emerged between prognosis and the expression of cortactin or its isoforms in supraglottic LSCC. Statistical analysis found cortactin expression higher in less-differentiated LSCCs (p = 0.03). A significant direct correlation was found between cortactin and p-Y466-cortactin levels (p = 0.031), and between p-Y466-cortactin and p-Y421-cortactin levels (p = 0.001). Conclusions: Cortactin expression in carcinoma cells and its known involvement in the EGFR pathway suggest a role for this protein as a target for LSCC therapy. Further prospective studies are needed to investigate the potential of cortactin, p-Y466-cortactin and p-Y421-cortactin expression as markers of response to treatment (particularly EGFR-directed agents) in LSCC.
Collapse
|
75
|
He J, Ma G, Qian J, Zhu Y, Liang M, Yao N, Ding Q, Chen L, Liu X, Xia T, Wang S. Interaction Between Ezrin and Cortactin in Promoting Epithelial to Mesenchymal Transition in Breast Cancer Cells. Med Sci Monit 2017; 23:1583-1596. [PMID: 28364518 PMCID: PMC5386444 DOI: 10.12659/msm.904124] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/14/2017] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Epithelial to mesenchymal transition (EMT) contributes to metastases in various types of tumors, and is also the key step in the breast cancer metastatic cascade. In our previous study, a mouse model containing human-derived normal breast tissue was established and allowed EMT/MET process of human breast cancer cells to be mimicked in a humanized mammary microenvironment. MATERIAL AND METHODS Two-dimensional electrophoresis (2-DE) and mass spectrometry were used to detect different proteins between parental MDA-MB-231 and its variant sub-line obtained from tumors grown in transplanted normal human breast tissue (MDA-MB-231br). We knocked down the ezrin in 2 cell lines (MDA-MB-231 and SUM1315). The migration and invasion ability was assessed. EMT markers were examined by real-time reverse transcription PCR analysis and Western blot analysis. The relationship of ezrin with cortactin was tested by tissue microarray and co-immunoprecipitation. RESULTS Proteomic analysis revealed 81 differentially expressed proteins between parental MDA-MB-231 and MDA-MB-231br. Among these proteins, the expression of ezrin and cortactin and the phosphorylation of ezrin were significantly correlated, accompanied with a group of classic EMT makers. Knockdown of ezrin reversed the expression of EMT markers and downregulated cortactin and EMT transcription factors. Ezrin silencing inhibited tumor cell migration and invasion. Breast cancer tissue microarray and immunohistochemistry showed a significant positive association between ezrin and cortactin. CONCLUSIONS These findings indicate that ezrin is correlated with cortactin in facilitating EMT in breast cancer. The interaction between ezrin and cortactin is a novel mechanism contributing to the EMT process in cancer metastases.
Collapse
Affiliation(s)
- Jing He
- Department of Surgical Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China (mainland)
| | - Ge Ma
- Breast Disease Center, 1st Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Jiayi Qian
- Breast Disease Center, 1st Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Mengdi Liang
- Breast Disease Center, 1st Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Na Yao
- Breast Disease Center, 1st Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Qiang Ding
- Breast Disease Center, 1st Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Lin Chen
- Breast Disease Center, 1st Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Xiaoan Liu
- Breast Disease Center, 1st Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Tiansong Xia
- Breast Disease Center, 1st Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Shui Wang
- Breast Disease Center, 1st Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
76
|
Reconstruction and signal propagation analysis of the Syk signaling network in breast cancer cells. PLoS Comput Biol 2017; 13:e1005432. [PMID: 28306714 PMCID: PMC5376343 DOI: 10.1371/journal.pcbi.1005432] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 03/31/2017] [Accepted: 02/20/2017] [Indexed: 11/19/2022] Open
Abstract
The ability to build in-depth cell signaling networks from vast experimental data is a key objective of computational biology. The spleen tyrosine kinase (Syk) protein, a well-characterized key player in immune cell signaling, was surprisingly first shown by our group to exhibit an onco-suppressive function in mammary epithelial cells and corroborated by many other studies, but the molecular mechanisms of this function remain largely unsolved. Based on existing proteomic data, we report here the generation of an interaction-based network of signaling pathways controlled by Syk in breast cancer cells. Pathway enrichment of the Syk targets previously identified by quantitative phospho-proteomics indicated that Syk is engaged in cell adhesion, motility, growth and death. Using the components and interactions of these pathways, we bootstrapped the reconstruction of a comprehensive network covering Syk signaling in breast cancer cells. To generate in silico hypotheses on Syk signaling propagation, we developed a method allowing to rank paths between Syk and its targets. We first annotated the network according to experimental datasets. We then combined shortest path computation with random walk processes to estimate the importance of individual interactions and selected biologically relevant pathways in the network. Molecular and cell biology experiments allowed to distinguish candidate mechanisms that underlie the impact of Syk on the regulation of cortactin and ezrin, both involved in actin-mediated cell adhesion and motility. The Syk network was further completed with the results of our biological validation experiments. The resulting Syk signaling sub-networks can be explored via an online visualization platform.
Collapse
|
77
|
Pennanen P, Alanne MH, Fazeli E, Deguchi T, Näreoja T, Peltonen S, Peltonen J. Diversity of actin architecture in human osteoclasts: network of curved and branched actin supporting cell shape and intercellular micrometer-level tubes. Mol Cell Biochem 2017; 432:131-139. [PMID: 28293874 PMCID: PMC5532409 DOI: 10.1007/s11010-017-3004-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/04/2017] [Indexed: 12/17/2022]
Abstract
Osteoclasts are multinucleated bone-resorbing cells with a dynamic actin cytoskeleton. Osteoclasts are derived from circulating mononuclear precursors. Confocal and stimulated emission depletion (STED) super-resolution microscopy was used to investigate peripheral blood-derived human osteoclasts cultured on glass surfaces. STED and confocal microscopy demonstrated that the actin was curved and branched, for instance, in the vicinity of membrane ruffles. The overall architecture of the curved actin network extended from the podosomes to the top of the cell. The other novel finding was that a micrometer-level tube containing actin bridged the osteoclasts well above the level of the culture glass. The actin filaments of the tubes originated from the network of curved actin often surrounding a group of nuclei. Furthermore, nuclei were occasionally located inside the tubes. Our findings demonstrated the accumulation of c-Src, cortactin, cofilin, and actin around nuclei suggesting their role in nuclear processes such as the locomotion of nuclei. ARP2/3 labeling was abundant at the substratum level of osteoclasts and in the branched actin network, where it localized to the branching points. We speculate that the actin-containing tubes of osteoclasts may provide a means of transportation of nuclei, e.g., during the fusion of osteoclasts. These novel findings can pave the way for future studies aiming at the elucidation of the differentiation of multinucleated osteoclasts.
Collapse
Affiliation(s)
- Paula Pennanen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Maria Helena Alanne
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Elnaz Fazeli
- Laboratory of Biophysics, Department of Cell Biology and Anatomy and Medicity Research Laboratories, University of Turku, P.O. Box 123, 20521, Turku, Finland
| | - Takahiro Deguchi
- Laboratory of Biophysics, Department of Cell Biology and Anatomy and Medicity Research Laboratories, University of Turku, P.O. Box 123, 20521, Turku, Finland
| | - Tuomas Näreoja
- Laboratory of Biophysics, Department of Cell Biology and Anatomy and Medicity Research Laboratories, University of Turku, P.O. Box 123, 20521, Turku, Finland
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sirkku Peltonen
- Department of Dermatology, University of Turku and Turku University Hospital, PO BOX 52, 20521, Turku, Finland
| | - Juha Peltonen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland.
| |
Collapse
|
78
|
Jeannot P, Nowosad A, Perchey RT, Callot C, Bennana E, Katsube T, Mayeux P, Guillonneau F, Manenti S, Besson A. p27 Kip1 promotes invadopodia turnover and invasion through the regulation of the PAK1/Cortactin pathway. eLife 2017; 6. [PMID: 28287395 PMCID: PMC5388532 DOI: 10.7554/elife.22207] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 03/09/2017] [Indexed: 12/29/2022] Open
Abstract
p27Kip1 (p27) is a cyclin-CDK inhibitor and negative regulator of cell proliferation. p27 also controls other cellular processes including migration and cytoplasmic p27 can act as an oncogene. Furthermore, cytoplasmic p27 promotes invasion and metastasis, in part by promoting epithelial to mesenchymal transition. Herein, we find that p27 promotes cell invasion by binding to and regulating the activity of Cortactin, a critical regulator of invadopodia formation. p27 localizes to invadopodia and limits their number and activity. p27 promotes the interaction of Cortactin with PAK1. In turn, PAK1 promotes invadopodia turnover by phosphorylating Cortactin, and expression of Cortactin mutants for PAK-targeted sites abolishes p27’s effect on invadopodia dynamics. Thus, in absence of p27, cells exhibit increased invadopodia stability due to impaired PAK1-Cortactin interaction, but their invasive capacity is reduced compared to wild-type cells. Overall, we find that p27 directly promotes cell invasion by facilitating invadopodia turnover via the Rac1/PAK1/Cortactin pathway. DOI:http://dx.doi.org/10.7554/eLife.22207.001 When animals develop from embryos to adults, or try to heal wounds later in life, their cells have to move. Moving means that the cells must invade into their surroundings, a dense network of proteins called the extracellular matrix. The cell first attaches to the extracellular matrix; degrades it; and then moves into the newly opened space. Cells have developed specialized structures called invadosomes to enable all these steps. Invadosomes are never static, they first assemble where cells interact with extracellular matrix, they then release proteins that loosen the matrix, and finally disassemble again to allow cells to move. Invadosomes in cancer cells often become overactive, and can allow the tumor cells to spread throughout the body. A lot of different proteins are involved in controlling how and when cells move. p27 is a well-known protein usually found in a cell’s nucleus along with the cell’s DNA. Inside the nucleus, p27 suppresses tumor growth by stopping cells from dividing. However, often in cancer cells p27 moves outside of the cell’s nucleus where it contributes to cell movement via an unknown mechanism. To answer how p27 controls cell invasion, Jeannot et al. used a biochemical technique to uncover which proteins p27 binds to when it is outside of the nucleus. One of its interaction partners was called Cortactin. This protein is known to be an important building block of invadosomes, and is involved in both the assembly and disassembly of these structures. In further experiments, Jeannot studied mouse cells with or without p27 and human cancer cells that can be grown in the laboratory. The experiments revealed that p27 promotes an enzyme called PAK1 to also bind to Cortactin. PAK1 then modified Cortactin, causing whole invadosomes to disassemble, which in turn allowed cells to de-attach from the matrix and move forward. In contrast, cells lacking p27 had more stable invadosomes, attached more strongly to the matrix and were better at degrading it, but could not invade as well as cells with p27. Overall these experiments showed a new way that p27 promotes cell invasion. The next steps will include finding out exactly how the modification of Cortactin causes the invadosomes to disassemble. Furthermore, it will be important to study whether forcing p27 back into the nucleus can reduce the spread of cancer cells in the body. DOI:http://dx.doi.org/10.7554/eLife.22207.002
Collapse
Affiliation(s)
- Pauline Jeannot
- INSERM UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,CNRS ERL5294, Toulouse, France
| | - Ada Nowosad
- INSERM UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,CNRS ERL5294, Toulouse, France
| | - Renaud T Perchey
- INSERM UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,CNRS ERL5294, Toulouse, France
| | - Caroline Callot
- INSERM UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,CNRS ERL5294, Toulouse, France
| | - Evangeline Bennana
- 3P5 proteomics facility of the Université Paris Descartes, Inserm U1016 Institut Cochin, Sorbonne Paris Cité, Paris, France
| | | | - Patrick Mayeux
- 3P5 proteomics facility of the Université Paris Descartes, Inserm U1016 Institut Cochin, Sorbonne Paris Cité, Paris, France
| | - François Guillonneau
- 3P5 proteomics facility of the Université Paris Descartes, Inserm U1016 Institut Cochin, Sorbonne Paris Cité, Paris, France
| | - Stéphane Manenti
- INSERM UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,CNRS ERL5294, Toulouse, France
| | - Arnaud Besson
- INSERM UMR1037, Cancer Research Center of Toulouse, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France.,CNRS ERL5294, Toulouse, France
| |
Collapse
|
79
|
Liu YC, Ho HC, Lee MR, Yeh CM, Tseng HC, Lin YC, Chung JG. Cortactin is a prognostic marker for oral squamous cell carcinoma and its overexpression is involved in oral carcinogenesis. ENVIRONMENTAL TOXICOLOGY 2017; 32:799-812. [PMID: 27148699 DOI: 10.1002/tox.22280] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 04/13/2016] [Accepted: 04/17/2016] [Indexed: 06/05/2023]
Abstract
EMS1 (chromosome eleven, band q13, mammary tumor and squamous cell carcinoma-associated gene 1) gene amplification and the concomitant cortactin overexpression have been reported to associate with poor prognosis and tumor metastasis. In this study, we examined cortactin expression by immunohistochemistry in human oral tumors and murine tongue tumors which were induced by the carcinogen 4-nitroquinoline 1-oxide (4-NQO). The immunostaining results show over- to moderate expression of cortactin in 85% (104/122) of oral squamous cell carcinoma (OSCC) tissues and in all 15 leukoplakia tissues examined. Further, statistical analysis indicates that cortactin overexpression appears to be a predictor for shorter survival and poorer prognosis in OSCC patients. In an animal model, cortactin is shown to upregulate in infiltrating squamous cell carcinoma, papilloma, and epithelia with squamous hyperplasia, indicating that cortactin induction is an early event during oral carcinogenesis. It is suggested that cortactin expression is mediated in the progression of pre-malignancy to papilloma, based on earlier cortactin induction in pre-malignancy preceding cyclin D1 in papilloma. In conclusion, cortactin overexpression is frequently observed in human OSCC and mouse tongue tumors. Thus, cortactin may have an important role in the development of oral tumors in human and mice. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 799-812, 2017.
Collapse
Affiliation(s)
- Yu-Ching Liu
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Heng-Chien Ho
- Departments of Biochemistry, China Medical University, Taichung, Taiwan
| | - Miau-Rong Lee
- Departments of Biochemistry, China Medical University, Taichung, Taiwan
| | - Chung-Min Yeh
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsien-Chang Tseng
- Department of Otolaryngology, China Medical University Hospital, Taichung, Taiwan
| | - Yung-Chang Lin
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Jing-Gung Chung
- Departments of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
- Department of Biotechnology, Asia University, Taichung, 413, Taiwan
| |
Collapse
|
80
|
Cortactin and phosphorylated cortactin tyr 466 expression in temporal bone carcinoma. Am J Otolaryngol 2017; 38:208-212. [PMID: 28131549 DOI: 10.1016/j.amjoto.2017.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/17/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE Cortactin is a multidomain protein engaged in several cellular mechanisms involving actin assembly and cytoskeletal arrangement. Cortactin overexpression in several malignancies has been associated with increased cell migration, invasion, and metastatic potential. Cortactin needs to be activated by tyrosine or serine/threonine phosphorylation. The role of cortactin and phosphorylated cortactin (residue tyr466) was investigated in temporal bone squamous cell carcinoma (TBSCC). MATERIALS AND METHODS Immunohistochemical expression of cortactin and phosphorylated cortactin (residue tyr466) was assessed in 27 consecutively-operated TBSCCs. RESULTS Several clinicopathological variables correlated with recurrence (pT stage, dura mater involvement), and disease-free survival (DFS) (cT stage, pT stage, pN status, dura mater involvement). Twenty-three of 24 immunohistochemically evaluable TBSCCs were cortactin-positive. Median cortactin expression was 75.0%. Cortactin reaction in the cytoplasm was more intense in carcinoma cells than in normal adjacent tissue. Recurrence and DFS rates did not correlate with cortactin and phosphorylated cortactin (residue tyr466) expression in TBSCC specimens. CONCLUSIONS Cortactin upregulation in TBSCC supports the conviction that inhibiting cortactin functions could have selective effects on this malignancy. Multi-institutional studies should further investigate the role of cortactin and phosphorylated cortactin in TBSCC, and their potential clinical application in integrated treatment modalities.
Collapse
|
81
|
Cortactin Is a Regulator of Activity-Dependent Synaptic Plasticity Controlled by Wingless. J Neurosci 2017; 37:2203-2215. [PMID: 28123080 DOI: 10.1523/jneurosci.1375-16.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 12/05/2016] [Accepted: 01/17/2017] [Indexed: 01/07/2023] Open
Abstract
Major signaling molecules initially characterized as key early developmental regulators are also essential for the plasticity of the nervous system. Previously, the Wingless (Wg)/Wnt pathway was shown to underlie the structural and electrophysiological changes during activity-dependent synaptic plasticity at the Drosophila neuromuscular junction. A challenge remains to understand how this signal mediates the cellular changes underlying this plasticity. Here, we focus on the actin regulator Cortactin, a major organizer of protrusion, membrane mobility, and invasiveness, and define its new role in synaptic plasticity. We show that Cortactin is present presynaptically and postsynaptically at the Drosophila NMJ and that it is a presynaptic regulator of rapid activity-dependent modifications in synaptic structure. Furthermore, animals lacking presynaptic Cortactin show a decrease in spontaneous release frequency, and presynaptic Cortactin is necessary for the rapid potentiation of spontaneous release frequency that takes place during activity-dependent plasticity. Most interestingly, Cortactin levels increase at stimulated synaptic terminals and this increase requires neuronal activity, de novo transcription and depends on Wg/Wnt expression. Because it is not simply the presence of Cortactin in the presynaptic terminal but its increase that is necessary for the full range of activity-dependent plasticity, we conclude that it probably plays a direct and important role in the regulation of this process.SIGNIFICANCE STATEMENT In the nervous system, changes in activity that lead to modifications in synaptic structure and function are referred to as synaptic plasticity and are thought to be the basis of learning and memory. The secreted Wingless/Wnt molecule is a potent regulator of synaptic plasticity in both vertebrates and invertebrates. Understanding the molecular mechanisms that underlie these plastic changes is a major gap in our knowledge. Here, we identify a presynaptic effector molecule of the Wingless/Wnt signal, Cortactin. We show that this molecule is a potent regulator of modifications in synaptic structure and is necessary for the electrophysiological changes taking place during synaptic plasticity.
Collapse
|
82
|
Role of Akt2 in regulation of metastasis suppressor 1 expression and colorectal cancer metastasis. Oncogene 2017; 36:3104-3118. [PMID: 28068324 DOI: 10.1038/onc.2016.460] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/27/2016] [Accepted: 10/31/2016] [Indexed: 12/15/2022]
Abstract
Survival signaling is critical for the metastatic program of cancer cells. The current study investigated the role of Akt survival proteins in colorectal cancer (CRC) metastasis and explored potential mechanisms of Akt-mediated metastasis regulation. Using an orthotopic implantation model in mice, which uniquely recapitulates the entire multistep process of CRC metastasis, combined with an inducible system of short hairpin RNA-mediated Akt isoform knockdown in human CRC cells, our studies confirm a role of Akt2 in CRC cell dissemination to distant organs in vivo. Akt2 deficiency profoundly inhibited the development of liver lesions in mice, whereas Akt1 had no effect under the experimental conditions used in the study. Array analysis of human metastatic genes identified the scaffolding protein metastasis suppressor 1 (MTSS1) as a novel Akt2-regulated gene. Inducible loss of Akt2 in CRC cells robustly upregulated MTSS1 at the messenger RNA and protein level, and the accumulated protein was functionally active as shown by its ability to engage an MTSS1-Src-cortactin inhibitory axis. MTSS1 expression led to a marked reduction in levels of functional cortacin (pcortactin Y421), an actin nucleation-promoting factor that has a crucial role in cancer cell invasion and metastasis. MTSS1 was also shown to mediate suppressive effects of Akt2 deficiency on CRC cell viability, survival, migration and actin polymerization in vitro. The relevance of these findings to human CRC is supported by analysis of The Cancer Genome Atlas (TCGA) and NCBI GEO data sets, which demonstrated inverse changes in expression of Akt2 and MTSS1 during CRC progression. Taken together, the data identify MTSS1 as a new Akt2-regulated gene, and point to suppression of MTSS1 as a key step in the metastasis-promoting effects of Akt2 in CRC cells.
Collapse
|
83
|
Xu A, Li X, Li S, Sun L, Wu S, Zhang B, Huang J. A novel role for 14-kDa phosphohistidine phosphatase in lamellipodia formation. Cell Adh Migr 2017; 11:488-495. [PMID: 27924678 DOI: 10.1080/19336918.2016.1268319] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cell migration involves dynamic regulation of the actin cytoskeleton, which exhibits rapid actin polymerization at the leading edge of migrating cells. This process relies on regulated recruitment of actin nucleators and actin-binding proteins to the leading edge to polymerize new actin filaments. Many of these proteins have been identified, including the actin-related protein (Arp) 2/3 complex, which has emerged as the core player in the initiation of actin polymerization. However, the functional coordination of these proteins is unclear. Previously, we have demonstrated that the 14-kDa phosphohistidine phosphatase (PHP14) is involved in cell migration regulation and affects actin cytoskeleton reorganization. Here, we show that PHP14 may regulate actin remodeling directly and play an important role in dynamic regulation of the actin cytoskeleton. We observed a colocalization of PHP14 with Arp3 and F-actin at the leading edge of migrating cells. Moreover, PHP14 was recruited to the actin remodeling sites in parallel with Arp3 during lamellipodia formation. Furthermore, PHP14 knockdown impaired Arp3 localization at the leading edge of lamellipodia, as well as lamellipodia formation. Most importantly, we found that PHP14 was a novel F-actin-binding protein, displaying an Arp2/3-dependent localization to the leading edge. Collectively, our results indicated a crucial role for PHP14 in the dynamic regulation of the actin cytoskeleton and cell migration.
Collapse
Affiliation(s)
- Anjian Xu
- a Experimental Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China.,b National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University , Beijing , China
| | - Xiaojin Li
- a Experimental Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China.,b National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University , Beijing , China
| | - Siwen Li
- a Experimental Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China
| | - Lan Sun
- c Department of Pathology , Beijing Friendship Hospital, Capital Medical University , Beijing , China
| | - Shanna Wu
- d Clinical Laboratory Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China
| | - Bei Zhang
- a Experimental Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China.,b National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University , Beijing , China
| | - Jian Huang
- a Experimental Center, Beijing Friendship Hospital, Capital Medical University , Beijing , China.,b National Clinical Research Center for Digestive Disease, Beijing Friendship Hospital, Capital Medical University , Beijing , China
| |
Collapse
|
84
|
Abstract
Schizophrenia is a severe psychiatric disorder that is characterized by a wide array of symptoms and a complex neuropathology. A well-characterized neurobiological feature of schizophrenia is abnormal synaptic plasticity, although the mechanisms underlying this are not fully understood. Numerous studies have demonstrated a link between proper functioning of the cytoskeleton and synaptic plasticity. The actin-related protein-2/3 (Arp2/3) complex is responsible for the nucleation of new actin filaments and elongation of existing actin filaments and is thus crucial to cytoskeletal dynamics, especially actin polymerization and organization. To determine whether the Arp2/3 complex is abnormally expressed in schizophrenia, we measured the protein expression of Arp2 and Arp3, as well as Arp2/3 complex binding partners and associated proteins including cortactin, neuronal-Wiskott-Aldrich syndrome protein (WASP), WASP-family verprolin homologous protein 1 (WAVE1), and Abelson interactor 1 (Abi1) in the superior temporal gyrus of paired schizophrenia and comparison participants. No changes were found in Arp2, Arp3, neuronal-WASP, WAVE1, or Abi1. However, all three isoforms of cortactin were decreased in schizophrenia. Specifically, the 62 kDa isoform was decreased by 43%; the 71 kDa isoform was decreased by 32%; and the 58 kDa isoform was decreased by 35%. Cortactin regulates branching of filamentous actin through its binding and activation of the Arp2/3 complex, and it is thus critical to the formation of stable actin networks. These findings contribute to a growing body of evidence implicating altered cytoskeletal dynamics in schizophrenia.
Collapse
|
85
|
Annexin A2 is critical for blood-testis barrier integrity and spermatid disengagement in the mammalian testis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:527-545. [PMID: 27974247 DOI: 10.1016/j.bbamcr.2016.12.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 12/06/2016] [Accepted: 12/09/2016] [Indexed: 01/08/2023]
Abstract
Throughout spermatogenesis, two important processes occur at late stage VIII of the seminiferous epithelial cycle in the rat testis: preleptotene spermatocytes commence entry into the adluminal compartment and step 19 spermatids release from the seminiferous epithelium. Presently, it is not clear how these processes, which involve extensive restructuring of unique Sertoli-Sertoli and Sertoli-germ cell junctions, are mediated. We aimed to determine whether annexin A2 (ANXA2), a Ca2+-dependent and phospholipid-binding protein, participates in cell junction dynamics. To address this, in vitro and in vivo RNA interference studies were performed on prepubertal Sertoli cells and adult rat testes. The endpoints of Anxa2 knockdown were determined by immunoblotting, morphological analyses, fluorescent immunostaining, and barrier integrity assays. In the testis, ANXA2 localized to the Sertoli cell stalk, with specific staining at the blood-testis barrier and the concave (ventral) surface of elongated spermatids. ANXA2 also bound actin when testis lysates were used for immunoprecipitation. Anxa2 knockdown was found to disrupt the Sertoli cell/blood-testis barrier in vitro and in vivo. The disruption in barrier function was substantiated by changes in the localization of claudin-11, zona occludens-1, N-cadherin, and β-catenin. Furthermore, Anxa2 knockdown resulted in spermiation defects caused by a dysfunction of tubulobulbar complexes, testis-specific actin-rich ultrastructures that internalize remnant cell junction components prior to spermiation. Additionally, there were changes in the localization of several tubulobulbar complex component proteins, including actin-related protein 3, cortactin, and dynamin I/II. Our results indicate that ANXA2 is critical for the integrity of the blood-testis barrier and the timely release of spermatids.
Collapse
|
86
|
Watkins RJ, Imruetaicharoenchoke W, Read ML, Sharma N, Poole VL, Gentilin E, Bansal S, Bosseboeuf E, Fletcher R, Nieto HR, Mallick U, Hackshaw A, Mehanna H, Boelaert K, Smith VE, McCabe CJ. Pro-invasive Effect of Proto-oncogene PBF Is Modulated by an Interaction with Cortactin. J Clin Endocrinol Metab 2016; 101:4551-4563. [PMID: 27603901 PMCID: PMC5155689 DOI: 10.1210/jc.2016-1932] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Metastatic disease is responsible for the majority of endocrine cancer deaths. New therapeutic targets are urgently needed to improve patient survival rates. OBJECTIVE The proto-oncogene PTTG1-binding factor (PBF/PTTG1IP) is overexpressed in multiple endocrine cancers and circumstantially associated with tumor aggressiveness. This study aimed to understand the role of PBF in tumor cell invasion and identify possible routes to inhibit its action. Design, Setting, Patients, and Interventions: Thyroid, breast, and colorectal cells were transfected with PBF and cultured for in vitro analysis. PBF and cortactin (CTTN) expression was determined in differentiated thyroid cancer and The Cancer Genome Atlas RNA-seq data. PRIMARY OUTCOME MEASURE Pro-invasive effects of PBF were evaluated by 2D Boyden chamber, 3D organotypic, and proximity ligation assays. RESULTS Our study identified that PBF and CTTN physically interact and co-localize, and that this occurs at the cell periphery, particularly at the leading edge of migrating cancer cells. Critically, PBF induces potent cellular invasion and migration in thyroid and breast cancer cells, which is entirely abrogated in the absence of CTTN. Importantly, we found that CTTN is over-expressed in differentiated thyroid cancer, particularly in patients with regional lymph node metastasis, which significantly correlates with elevated PBF expression. Mutation of PBF (Y174A) or pharmacological intervention modulates the PBF: CTTN interaction and attenuates the invasive properties of cancer cells. CONCLUSION Our results demonstrate a unique role for PBF in regulating CTTN function to promote endocrine cell invasion and migration, as well as identify a new targetable interaction to block tumor cell movement.
Collapse
Affiliation(s)
- Rachel J Watkins
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Waraporn Imruetaicharoenchoke
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Martin L Read
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Neil Sharma
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Vikki L Poole
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Erica Gentilin
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Sukhchain Bansal
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Emy Bosseboeuf
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Rachel Fletcher
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Hannah R Nieto
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Ujjal Mallick
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Allan Hackshaw
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Hisham Mehanna
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Kristien Boelaert
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Vicki E Smith
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Christopher J McCabe
- Institute of Metabolism and Systems Research (R.J.W., W.I., M.L.R., N.S., V.L.P., S.B., R.F., H.R.N., K.B., V.E.S., C.J.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom; Department of Surgery, Faculty of Medicine (W.I.), Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Section of Endocrinology and Internal Medicine (E.G.), University of Ferrara, 44121 Ferrara, Italy; STIM Laboratory (E.B.), University of Poitiers, 86073 Poitiers Cedex 9, France; Northern Centre for Cancer Care (U.M.), Freeman Hospital, Newcastle upon Tyne NE7 7DN, United Kingdom; Cancer Research United Kingdom & UCL Cancer Trials Centre (A.H.), University College London, London WC1E 6BT, United Kingdom; and Institute of Cancer and Genomic Sciences (H.M.), University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
87
|
Wu H, Cheng X, Ji X, He Y, Jing X, Wu H, Zhao R. Cortactin contributes to the tumorigenicity of colorectal cancer by promoting cell proliferation. Oncol Rep 2016; 36:3497-3503. [PMID: 27805253 DOI: 10.3892/or.2016.5207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/25/2016] [Indexed: 11/06/2022] Open
Abstract
Cortactin is a scaffolding protein that regulates Arp2/3-mediated actin polymerization. We showed in a previous study that cortactin was highly expressed in human stage II-III colorectal cancer (CRC) tissues. In the present study, using colony formation and CCK-8 assays, we showed that overexpression of cortactin accelerated the proliferation of CRC cells. Flow cytometric assays revealed that cortactin promoted G1/S phase cell cycle transition. Later, we constructed the phosphorylation mutation of cortactin at the Tyr421 residue. Colony formation and CCK-8 assays showed that cortactin/Tyr421A lost its ability to promote cell proliferation. Western blot analysis indicated that cortactin activated cyclin D1, but not cortactin/Tyr421A. Further study in nude mice revealed that there was a greater decrease in both tumor volume and tumor weight in animals injected with SW480/cortactin/Tyr421A cells than in those injected with SW480/cortactin/WT cells. Thus, the present study demonstrates that the cortactin Tyr421 residue is required to promote cell proliferation both in vitro and in vivo.
Collapse
Affiliation(s)
- Huo Wu
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Xi Cheng
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Xiaopin Ji
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Yonggang He
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Xiaoqian Jing
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Haoxuan Wu
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Ren Zhao
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
88
|
Qi Y, Dai Y, Gui S. Protein tyrosine phosphatase PTPRB regulates Src phosphorylation and tumour progression in NSCLC. Clin Exp Pharmacol Physiol 2016; 43:1004-12. [PMID: 27314562 DOI: 10.1111/1440-1681.12610] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Yinliang Qi
- Department of Respiratory Disease; Anhui Medical University; Hefei Anhui China
- General Department of Hyperbaric Oxygen; The Second People's Hospital of Hefei; Hefei Anhui China
| | - Yuanchang Dai
- General Department of Hyperbaric Oxygen; The Second People's Hospital of Hefei; Hefei Anhui China
| | - Shuyu Gui
- Department of Respiratory Disease; Anhui Medical University; Hefei Anhui China
| |
Collapse
|
89
|
Elevated hydrostatic pressure enhances the motility and enlarges the size of the lung cancer cells through aquaporin upregulation mediated by caveolin-1 and ERK1/2 signaling. Oncogene 2016; 36:863-874. [DOI: 10.1038/onc.2016.255] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 05/25/2016] [Accepted: 06/13/2016] [Indexed: 02/07/2023]
|
90
|
Chen DY, Husain M. Caspase-mediated degradation of host cortactin that promotes influenza A virus infection in epithelial cells. Virology 2016; 497:146-156. [PMID: 27471953 DOI: 10.1016/j.virol.2016.07.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/18/2016] [Accepted: 07/18/2016] [Indexed: 01/09/2023]
Abstract
Influenza A virus (IAV) is well-known to exploit host factors to its advantage. Here, we report that IAV exploits host cortactin, an actin filament-stabilising protein for infection in epithelial cells. By using RNA interference-mediated knockdown and overexpression approach, we demonstrate that cortactin promotes IAV infection. However, cortactin polypeptide undergoes the degradation during late IAV infection. By perturbing the lysosome and proteasome, two main compartments governing the degradation of mammalian proteins, we demonstrate that a lysosome-associated apoptotic pathway mediates the degradation of cortactin in IAV-infected cells. However, we could not detect cleaved cortactin fragments by western blotting using the antibodies recognising either N-terminal/Central or C-terminal cortactin regions, which suggested the presence of multiple caspase cleavage sites. Indeed, CaspDB, a recently-described database predicted up to 35 caspase cleavage motifs present across cortactin polypeptide. The data presented indicate that host cortactin potentially has a dual but contrasting role during IAV infection.
Collapse
Affiliation(s)
- Da-Yuan Chen
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Matloob Husain
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
91
|
Adams G, Zhou J, Wang W, Wu H, Quan J, Liu Y, Xia P, Wang Z, Zhou S, Jiang J, Mo F, Zhuang X, Thomas K, Hill DL, Aikhionbare FO, He P, Liu X, Ding X, Yao X. The Microtubule Plus End Tracking Protein TIP150 Interacts with Cortactin to Steer Directional Cell Migration. J Biol Chem 2016; 291:20692-706. [PMID: 27451391 DOI: 10.1074/jbc.m116.732719] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Indexed: 02/05/2023] Open
Abstract
Cell migration is orchestrated by dynamic interactions of microtubules with the plasma membrane cortex. How these interactions facilitate these dynamic processes is still being actively investigated. TIP150 is a newly characterized microtubule plus end tracking protein essential for mitosis and entosis (Ward, T., Wang, M., Liu, X., Wang, Z., Xia, P., Chu, Y., Wang, X., Liu, L., Jiang, K., Yu, H., Yan, M., Wang, J., Hill, D. L., Huang, Y., Zhu, T., and Yao, X. (2013) Regulation of a dynamic interaction between two microtubule-binding proteins, EB1 and TIP150, by the mitotic p300/CBP-associated factor (PCAF) orchestrates kinetochore microtubule plasticity and chromosome stability during mitosis. J. Biol. Chem. 288, 15771-15785; Xia, P., Zhou, J., Song, X., Wu, B., Liu, X., Li, D., Zhang, S., Wang, Z., Yu, H., Ward, T., Zhang, J., Li, Y., Wang, X., Chen, Y., Guo, Z., and Yao, X. (2014) Aurora A orchestrates entosis by regulating a dynamic MCAK-TIP150 interaction. J. Mol. Cell Biol. 6, 240-254). Here we show that TIP150 links dynamic microtubules to steer cell migration by interacting with cortactin. Mechanistically, TIP150 binds to cortactin via its C-terminal tail. Interestingly, the C-terminal TIP150 proline-rich region (CT150) binds to the Src homology 3 domain of cortactin specifically, and such an interaction is negatively regulated by EGF-elicited tyrosine phosphorylation of cortactin. Importantly, suppression of TIP150 or overexpression of phospho-mimicking cortactin inhibits polarized cell migration. In addition, CT150 disrupts the biochemical interaction between TIP150 and cortactin in vitro, and perturbation of the TIP150-cortactin interaction in vivo using a membrane-permeable TAT-CT150 peptide results in an inhibition of directional cell migration. We reason that a dynamic TIP150-cortactin interaction orchestrates directional cell migration via coupling dynamic microtubule plus ends to the cortical cytoskeleton.
Collapse
Affiliation(s)
- Gregory Adams
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China, the Departments of Physiology and
| | - Jiajia Zhou
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Wenwen Wang
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China, the Departments of Physiology and
| | - Huihui Wu
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China, the Departments of Physiology and
| | - Jie Quan
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yingying Liu
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China, the Departments of Physiology and
| | - Peng Xia
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zhikai Wang
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China, the Departments of Physiology and
| | - Shu Zhou
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jiying Jiang
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Fei Mo
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiaoxuan Zhuang
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Kelwyn Thomas
- Medicine and Neurobiology, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Donald L Hill
- the Comprehensive Cancer Center, University of Alabama, Birmingham, Alabama 35294, and
| | - Felix O Aikhionbare
- Medicine and Neurobiology, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Ping He
- the Departments of Physiology and the Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Xing Liu
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China, the Departments of Physiology and
| | - Xia Ding
- From the BUCM-MSM Joint Research Group for Cellular Dynamics, BUCM School of Basic Medical Sciences, and Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, University of Science and Technology of China, Hefei, Anhui 230026, China,
| | | |
Collapse
|
92
|
Baghel KS, Tewari BN, Shrivastava R, Malik SA, Lone MUD, Jain NK, Tripathi C, Kanchan RK, Dixit S, Singh K, Mitra K, Negi MPS, Srivastava M, Misra S, Bhatt MLB, Bhadauria S. Macrophages promote matrix protrusive and invasive function of breast cancer cells via MIP-1β dependent upregulation of MYO3A gene in breast cancer cells. Oncoimmunology 2016; 5:e1196299. [PMID: 27622050 DOI: 10.1080/2162402x.2016.1196299] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/20/2022] Open
Abstract
The potential of a tumor cell to metastasize profoundly depends on its microenvironment, or "niche" interactions with local components. Tumor-associated-macrophages (TAMs) are the most abundant subpopulation of tumor stroma and represent a key component of tumor microenvironment. The dynamic interaction of cancer cells with neighboring TAMs actively drive cancer progression and metastatic transformation through intercellular signaling networks that need better elucidation. Thus, current study was planned for discerning paracrine communication networks operational between TAMs, and breast cancer cells with special reference to cancer cell invasion and dissemination to distant sites. Here, we report role of MIP-1β in enhancing invasive potential of metastatic breast cancer MDA-MB-231 and MDA-MB-468 cells. In addition, the poorly metastatic MCF-7 cells were also rendered invasive by MIP-1β. The MIP-1β-driven cancer cell invasion was dependent on upregulated expression levels of MYO3A gene, which encodes an unconventional myosin super-family protein harboring a kinase domain. Ex ovo study employing Chick-embryo-model and in vivo Syngenic 4T1/BALB/c mice-model further corroborated aforementioned in vitro findings, thereby substantiating their physiological relevance. Concordantly, human breast cancer specimen exhibited significant association between mRNA expression levels of MIP-1β and MYO3A. Both, MIP-1β and MYO3A exhibited positive correlation with MMP9, an established molecular determinant of cancer cell invasion. Higher expression of these genes correlated with poor survival of breast cancer patients. Collectively, these results point toward so far undisclosed MIP-1β/MYO3A axis being operational during metastasis, wherein macrophage-derived MIP-1β potentiated cancer cell invasion and metastasis via up regulation of MYO3A gene within cancer cells. Our study exposes opportunities for devising potential anti-metastatic strategies for efficient clinical management of breast cancer.
Collapse
Affiliation(s)
- Khemraj Singh Baghel
- Division of Toxicology, Central Drug Research Institute (CSIR) , Lucknow, Uttar Pradesh, India
| | - Brij Nath Tewari
- Department of Surgical Oncology, King George Medical University , Lucknow, Uttar Pradesh, India
| | - Richa Shrivastava
- Division of Toxicology, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, (AcSIR), New Delhi, India
| | - Showkat Ahmad Malik
- Division of Toxicology, Central Drug Research Institute (CSIR) , Lucknow, Uttar Pradesh, India
| | - Mehraj U-Din Lone
- Division of Toxicology, Central Drug Research Institute (CSIR) , Lucknow, Uttar Pradesh, India
| | - Nem Kumar Jain
- Division of Toxicology, Central Drug Research Institute (CSIR) , Lucknow, Uttar Pradesh, India
| | - Chakrapani Tripathi
- Division of Toxicology, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, (AcSIR), New Delhi, India
| | - Ranjana Kumari Kanchan
- Division of Toxicology, Central Drug Research Institute (CSIR) , Lucknow, Uttar Pradesh, India
| | - Sameer Dixit
- Division of Plant Molecular Biology and Genetic Engineering, National Botanical Research Institute (CSIR) , Lucknow, Uttar Pradesh, India
| | - Kavita Singh
- Electron Microscopy Unit, Sophisticated Analytical Instrumentation Facility, Central Drug Research Institute (CSIR) , Lucknow, Uttar Pradesh, India
| | - Kalyan Mitra
- Academy of Scientific and Innovative Research, (AcSIR), New Delhi, India; Electron Microscopy Unit, Sophisticated Analytical Instrumentation Facility, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh, India
| | - Mahendra Pal Singh Negi
- Academy of Scientific and Innovative Research, (AcSIR), New Delhi, India; Division of Clinical and Experimental Medicine, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh, India
| | - Mukesh Srivastava
- Academy of Scientific and Innovative Research, (AcSIR), New Delhi, India; Division of Clinical and Experimental Medicine, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh, India
| | - Sanjeev Misra
- Department of Surgical Oncology, King George Medical University , Lucknow, Uttar Pradesh, India
| | - Madan Lal Brahma Bhatt
- Department of Radiotherapy, King George Medical University , Lucknow, Uttar Pradesh, India
| | - Smrati Bhadauria
- Division of Toxicology, Central Drug Research Institute (CSIR), Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, (AcSIR), New Delhi, India
| |
Collapse
|
93
|
Novel role of cortactin in G protein-coupled receptor agonist-induced nuclear export and degradation of p21Cip1. Sci Rep 2016; 6:28687. [PMID: 27363897 PMCID: PMC4929470 DOI: 10.1038/srep28687] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/08/2016] [Indexed: 12/16/2022] Open
Abstract
Monocyte chemotactic protein 1 (MCP1) stimulates phosphorylation of cortactin on Y421 and Y446 residues in a time-dependent manner and phosphorylation at Y446 but not Y421 residue is required for MCP1-induced CDK-interacting protein 1 (p21Cip1) nuclear export and degradation in facilitating human aortic smooth muscle cell (HASMC) proliferation. In addition, MCP1-induced cortactin tyrosine phosphorylation, p21Cip1 degradation and HASMC proliferation are dependent on Fyn activation. Upstream to Fyn, MCP1 stimulated C-C chemokine receptor type 2 (CCR2) and Gi/o and inhibition of either one of these molecules using their specific antagonists or inhibitors attenuated MCP1-induced cortactin tyrosine phosphorylation, p21Cip1 degradation and HASMC proliferation. Cortactin phosphorylation at Y446 residue is also required for another G protein-coupled receptor (GPCR) agonist, thrombin-induced p21Cip1 nuclear export and its degradation in promoting HASMC proliferation. Quite interestingly, the receptor tyrosine kinase (RTK) agonist, platelet-derived growth factor-BB (PDGF-BB)-induced p21Cip1 degradation and HASMC proliferation do not require cortactin tyrosine phosphorylation. Together, these findings demonstrate that tyrosine phosphorylation of cortactin at Y446 residue is selective for only GPCR but not RTK agonist-induced nuclear export and proteolytic degradation of p21Cip1 in HASMC proliferation.
Collapse
|
94
|
García Ponce A, Citalán Madrid AF, Vargas Robles H, Chánez Paredes S, Nava P, Betanzos A, Zarbock A, Rottner K, Vestweber D, Schnoor M. Loss of cortactin causes endothelial barrier dysfunction via disturbed adrenomedullin secretion and actomyosin contractility. Sci Rep 2016; 6:29003. [PMID: 27357373 PMCID: PMC4928053 DOI: 10.1038/srep29003] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 06/13/2016] [Indexed: 12/28/2022] Open
Abstract
Changes in vascular permeability occur during inflammation and the actin cytoskeleton plays a crucial role in regulating endothelial cell contacts and permeability. We demonstrated recently that the actin-binding protein cortactin regulates vascular permeability via Rap1. However, it is unknown if the actin cytoskeleton contributes to increased vascular permeability without cortactin. As we consistently observed more actin fibres in cortactin-depleted endothelial cells, we hypothesised that cortactin depletion results in increased stress fibre contractility and endothelial barrier destabilisation. Analysing the contractile machinery, we found increased ROCK1 protein levels in cortactin-depleted endothelium. Concomitantly, myosin light chain phosphorylation was increased while cofilin, mDia and ERM were unaffected. Secretion of the barrier-stabilising hormone adrenomedullin, which activates Rap1 and counteracts actomyosin contractility, was reduced in plasma from cortactin-deficient mice and in supernatants of cortactin-depleted endothelium. Importantly, adrenomedullin administration and ROCK1 inhibition reduced actomyosin contractility and rescued the effect on permeability provoked by cortactin deficiency in vitro and in vivo. Our data suggest a new role for cortactin in controlling actomyosin contractility with consequences for endothelial barrier integrity.
Collapse
Affiliation(s)
- Alexander García Ponce
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Alí F Citalán Madrid
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Hilda Vargas Robles
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Sandra Chánez Paredes
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Porfirio Nava
- Department for Physiology, Biophysics and Neurosciences, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Abigail Betanzos
- Department for Infectomics and Molecular Pathogenesis, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Clinic of Münster, 48149 Münster, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, TU Braunschweig, 38106 Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Dietmar Vestweber
- Department for Vascular Cell Biology, Max-Planck-Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Michael Schnoor
- Department for Molecular Biomedicine, Center of Research and Advanced Studies (CINVESTAV-IPN), 07360 Mexico-City, Mexico
| |
Collapse
|
95
|
Yamada H, Takeda T, Michiue H, Abe T, Takei K. Actin bundling by dynamin 2 and cortactin is implicated in cell migration by stabilizing filopodia in human non-small cell lung carcinoma cells. Int J Oncol 2016; 49:877-86. [PMID: 27572123 PMCID: PMC4948956 DOI: 10.3892/ijo.2016.3592] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/25/2016] [Indexed: 11/06/2022] Open
Abstract
The endocytic protein dynamin participates in the formation of actin-based membrane protrusions such as podosomes, pseudopodia, and invadopodia, which facilitate cancer cell migration, invasion, and metastasis. However, the role of dynamin in the formation of actin-based membrane protrusions at the leading edge of cancer cells is unclear. In this study, we demonstrate that the ubiquitously expressed dynamin 2 isoform facilitates cell migration by stabilizing F-actin bundles in filopodia of the lung cancer cell line H1299. Pharmacological inhibition of dynamin 2 decreased cell migration and filopodial formation. Furthermore, dynamin 2 and cortactin mostly colocalized along F-actin bundles in filopodia of serum-stimulated H1299 cells by immunofluorescent and immunoelectron microscopy. Knockdown of dynamin 2 or cortactin inhibited the formation of filopodia in serum-stimulated H1299 cells, concomitant with a loss of F-actin bundles. Expression of wild-type cortactin rescued the punctate-like localization of dynamin 2 and filopodial formation. The incubation of dynamin 2 and cortactin with F-actin induced the formation of long and thick actin bundles, with these proteins colocalizing at F-actin bundles. A depolymerization assay revealed that dynamin 2 and cortactin increased the stability of F-actin bundles. These results indicate that dynamin 2 and cortactin participate in cell migration by stabilizing F-actin bundles in filopodia. Taken together, these findings suggest that dynamin might be a possible molecular target for anticancer therapy.
Collapse
Affiliation(s)
- Hiroshi Yamada
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Tetsuya Takeda
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Hiroyuki Michiue
- Department of Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Tadashi Abe
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Kohji Takei
- Department of Neuroscience, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
96
|
The Phosphorylation and Distribution of Cortactin Downstream of Integrin α9β1 Affects Cancer Cell Behaviour. Sci Rep 2016; 6:28529. [PMID: 27339664 PMCID: PMC4919783 DOI: 10.1038/srep28529] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 06/06/2016] [Indexed: 11/25/2022] Open
Abstract
Integrins, a family of heterodimeric adhesion receptors are implicated in cell migration, development and cancer progression. They can adopt conformations that reflect their activation states and thereby impact adhesion strength and migration. Integrins in an intermediate activation state may be optimal for migration and we have shown previously that fully activated integrin α9β1 corresponds with less migratory behaviour in melanoma cells. Here, we aimed to identify components associated with the activation status of α9β1. Using cancer cell lines with naturally occuring high levels of this integrin, activation by α9β1-specific ligands led to upregulation of fibronectin matrix assembly and tyrosine phosphorylation of cortactin on tyrosine 470 (Y470). Specifically, cortactin phosphorylated on Y470, but not Y421, redistributed together with α9β1 to focal adhesions where active β1 integrin also localises, upon integrin activation. This was commensurate with reduced migration. The localisation and phosphorylation of cortactin Y470 was regulated by Yes kinase and PTEN phosphatase. Cortactin levels influenced fibronectin matrix assembly and active β1 integrin on the cell surface, being inversely correlated with migratory behaviour. This study underlines the complex interplay between cortactin and α9β1 integrin that regulates cell-extracellular matrix interactions.
Collapse
|
97
|
Wu Q, Nadesalingam J, Moodley S, Bai X, Liu M. XB130 translocation to microfilamentous structures mediates NNK-induced migration of human bronchial epithelial cells. Oncotarget 2016; 6:18050-65. [PMID: 25980441 PMCID: PMC4627235 DOI: 10.18632/oncotarget.3777] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/09/2015] [Indexed: 01/02/2023] Open
Abstract
Cigarette smoking contributes to the pathogenesis of chronic obstructive pulmonary disease and lung cancer. Nicotine-derived nitrosamine ketone (NNK) is the most potent carcinogen among cigarette smoking components, and is known to enhance migration of cancer cells. However, the effect of NNK on normal human bronchial epithelial cells is not well studied. XB130 is a member of actin filament associated protein family and is involved in cell morphology changes, cytoskeletal rearrangement and outgrowth formation, as well as cell migration. We hypothesized that XB130 mediates NNK-induced migration of normal human bronchial epithelial cells. Our results showed that, after NNK stimulation, XB130 was translocated to the cell periphery and enriched in cell motility-associated structures, such as lamellipodia, in normal human bronchial epithelial BEAS2B cells. Moreover, overexpression of XB130 significantly enhanced NNK-induced migration, which requires both the N- and C-termini of XB130. Overexpression of XB130 enhanced NNK-induced protein tyrosine phosphorylation and promoted matrix metalloproteinase-14 translocation to cell motility-associated cellular structures after NNK stimulation. XB130-mediated NNK-induced cell migration may contribute to airway epithelial repair; however, it may also be involved in cigarette smoking-related chronic obstructive pulmonary disease and lung cancer.
Collapse
Affiliation(s)
- Qifei Wu
- Latner Thoracic Surgery Research Laboratories, University Health Network, Toronto General Research Institute, Toronto, Ontario, Canada.,Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jeya Nadesalingam
- Latner Thoracic Surgery Research Laboratories, University Health Network, Toronto General Research Institute, Toronto, Ontario, Canada
| | - Serisha Moodley
- Latner Thoracic Surgery Research Laboratories, University Health Network, Toronto General Research Institute, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Xiaohui Bai
- Latner Thoracic Surgery Research Laboratories, University Health Network, Toronto General Research Institute, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, University Health Network, Toronto General Research Institute, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
98
|
Identification of a shootin1 isoform expressed in peripheral tissues. Cell Tissue Res 2016; 366:75-87. [PMID: 27177867 DOI: 10.1007/s00441-016-2415-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/20/2016] [Indexed: 10/24/2022]
Abstract
Shootin1 is a brain-specific cytoplasmic protein involved in neuronal polarity formation and axon outgrowth. It accumulates at the leading edge of axonal growth cones, where it mediates the mechanical coupling between F-actin retrograde flow and cell adhesions as a clutch molecule, thereby producing force for axon outgrowth. In this study, we report a novel splicing isoform of shootin1 which is expressed not only in the brain but also in peripheral tissues. We have renamed the brain-specific shootin1 as shootin1a and termed the novel isoform as shootin1b. Immunoblot and immunohistochemical analyses with a shootin1b-specific antibody revealed that shootin1b is distributed in various mouse tissues including the lung, liver, stomach, intestines, spleen, pancreas, kidney and skin. Interestingly, shootin1b immunoreactivity was widely detected in epithelial cells that constitute simple and stratified epithelia; in some cells, it colocalized with E-cadherin and cortactin at cell-cell contact sites. Shootin1b also localized in dendritic cells in the spleen. These results suggest that shootin1b may function in various peripheral tissues including epithelial cells.
Collapse
|
99
|
Daulat AM, Bertucci F, Audebert S, Sergé A, Finetti P, Josselin E, Castellano R, Birnbaum D, Angers S, Borg JP. PRICKLE1 Contributes to Cancer Cell Dissemination through Its Interaction with mTORC2. Dev Cell 2016; 37:311-325. [PMID: 27184734 DOI: 10.1016/j.devcel.2016.04.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 03/15/2016] [Accepted: 04/18/2016] [Indexed: 12/21/2022]
Abstract
Components of the evolutionarily conserved developmental planar cell polarity (PCP) pathway were recently described to play a prominent role in cancer cell dissemination. However, the molecular mechanisms by which PCP molecules drive the spread of cancer cells remain largely unknown. PRICKLE1 encodes a PCP protein bound to the promigratory serine/threonine kinase MINK1. We identify RICTOR, a member of the mTORC2 complex, as a PRICKLE1-binding partner and show that the integrity of the PRICKLE1-MINK1-RICTOR complex is required for activation of AKT, regulation of focal adhesions, and cancer cell migration. Disruption of the PRICKLE1-RICTOR interaction results in a strong impairment of breast cancer cell dissemination in xenograft assays. Finally, we show that upregulation of PRICKLE1 in basal breast cancers, a subtype characterized by high metastatic potential, is associated with poor metastasis-free survival.
Collapse
Affiliation(s)
- Avais M Daulat
- Inserm, U1068, CRCM, Cell Polarity, Cell Signalling and Cancer "Equipe labellisée Ligue Contre le Cancer", Marseille 13009, France; Institut Paoli-Calmettes, Marseille 13009, France; Aix-Marseille Université, UM 105, Marseille 13284, France; CNRS, UMR7258, CRCM, Marseille 13009, France
| | - François Bertucci
- Institut Paoli-Calmettes, Marseille 13009, France; Aix-Marseille Université, UM 105, Marseille 13284, France; CNRS, UMR7258, CRCM, Marseille 13009, France; Inserm, U1068, CRCM, Molecular Oncology "Equipe labellisée Ligue Contre le Cancer", Marseille 13009, France
| | - Stéphane Audebert
- Inserm, U1068, CRCM, Cell Polarity, Cell Signalling and Cancer "Equipe labellisée Ligue Contre le Cancer", Marseille 13009, France; Institut Paoli-Calmettes, Marseille 13009, France; Aix-Marseille Université, UM 105, Marseille 13284, France; CNRS, UMR7258, CRCM, Marseille 13009, France
| | - Arnauld Sergé
- Institut Paoli-Calmettes, Marseille 13009, France; Aix-Marseille Université, UM 105, Marseille 13284, France; CNRS, UMR7258, CRCM, Marseille 13009, France; Inserm, U1068, CRCM, Leuko/Stromal Interactions, Marseille 13009, France
| | - Pascal Finetti
- Institut Paoli-Calmettes, Marseille 13009, France; Aix-Marseille Université, UM 105, Marseille 13284, France; CNRS, UMR7258, CRCM, Marseille 13009, France; Inserm, U1068, CRCM, Molecular Oncology "Equipe labellisée Ligue Contre le Cancer", Marseille 13009, France
| | - Emmanuelle Josselin
- Institut Paoli-Calmettes, Marseille 13009, France; Aix-Marseille Université, UM 105, Marseille 13284, France; CNRS, UMR7258, CRCM, Marseille 13009, France; Inserm, U1068, CRCM, TrGET Platform, Marseille 13009, France
| | - Rémy Castellano
- Institut Paoli-Calmettes, Marseille 13009, France; Aix-Marseille Université, UM 105, Marseille 13284, France; CNRS, UMR7258, CRCM, Marseille 13009, France; Inserm, U1068, CRCM, TrGET Platform, Marseille 13009, France
| | - Daniel Birnbaum
- Institut Paoli-Calmettes, Marseille 13009, France; Aix-Marseille Université, UM 105, Marseille 13284, France; CNRS, UMR7258, CRCM, Marseille 13009, France; Inserm, U1068, CRCM, Molecular Oncology "Equipe labellisée Ligue Contre le Cancer", Marseille 13009, France
| | - Stéphane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S3M2, Canada; Department of Biochemistry, Faculty of Medicine, University of Toronto, ON M5S1A8, Canada
| | - Jean-Paul Borg
- Inserm, U1068, CRCM, Cell Polarity, Cell Signalling and Cancer "Equipe labellisée Ligue Contre le Cancer", Marseille 13009, France; Institut Paoli-Calmettes, Marseille 13009, France; Aix-Marseille Université, UM 105, Marseille 13284, France; CNRS, UMR7258, CRCM, Marseille 13009, France.
| |
Collapse
|
100
|
Sánchez A, Urrego D, Pardo LA. Cyclic expression of the voltage-gated potassium channel KV10.1 promotes disassembly of the primary cilium. EMBO Rep 2016; 17:708-23. [PMID: 27113750 PMCID: PMC5341513 DOI: 10.15252/embr.201541082] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 03/03/2016] [Accepted: 03/09/2016] [Indexed: 12/19/2022] Open
Abstract
The primary cilium, critical for morphogenic and growth factor signaling, is assembled upon cell cycle exit, but the links between ciliogenesis and cell cycle progression are unclear. KV10.1 is a voltage-gated potassium channel frequently overexpressed in tumors. We have previously reported that expression of KV10.1 is temporally restricted to a time period immediately prior to mitosis in healthy cells. Here, we provide microscopical and biochemical evidence that KV10.1 localizes to the centrosome and the primary cilium and promotes ciliary disassembly. Interference with KV10.1 ciliary localization abolishes not only the effects on ciliary disassembly, but also KV10.1-induced tumor progression in vivo Conversely, upon knockdown of KV10.1, ciliary disassembly is impaired, proliferation is delayed, and proliferating cells show prominent primary cilia. Thus, modulation of ciliogenesis by KV10.1 can explain the influence of KV10.1 expression on the proliferation of normal cells and is likely to be a major mechanism underlying its tumorigenic effects.
Collapse
Affiliation(s)
- Araceli Sánchez
- Oncophysiology Group, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| | - Diana Urrego
- Oncophysiology Group, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| | - Luis A Pardo
- Oncophysiology Group, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|