1
|
Gope TK, Pal D, Srivastava AK, Chatterjee B, Bose S, Ain R. ARID3A inhibits colorectal cancer cell stemness and drug-resistance by targeting a multitude of stemness-associated genes. Life Sci 2025; 372:123642. [PMID: 40250751 DOI: 10.1016/j.lfs.2025.123642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/14/2025] [Accepted: 04/11/2025] [Indexed: 04/20/2025]
Abstract
AIMS ARID3A is highly expressed in CRC patients. However, the functional role of ARID3A in CRC remains unexplored. We sought to demonstrate ARID3A function in CRC. MATERIALS AND METHODS ARID3A was knocked-down using lentiviruses harboring shRNA. CRC patients' tissue cDNA array was used to assess expression of ARID3A. Effect of ARID3A on CSC-associated genes was analysed using real-time PCR array. Western-blot analysis and ChIP assay were used to validate the role of ARID3A. Paclitaxel-resistant CSC-enriched cell population was used to assess correlation between ARID3A, stemness and drug resistance potential. Ex vivo findings were corroborated on preclinical mouse model. KEY FINDINGS ARID3A expression was significantly higher throughout CRC stages than normal individuals. ARID3A expression was significantly higher in the aggressive CRC cell line HCT116 compared to HT29, which expressed higher levels of CD44, CD133, and EpCAM, suggesting a reciprocal relationship between ARID3A expression and CRC stemness. Real-time PCR-based stem cell array using ARID3A-knockdown HCT116 cells showed upregulation of 9 cancer stem cell (CSC)-associated genes. ChIP-assay verified binding of ARID3A on transcriptionally active promoter regions of CSC associated genes. ARID3A depletion led to enhanced proliferation, anchorage-independent growth, and ABCG2 upregulation in HCT116 cells. In paclitaxel-resistant HCT116 cells, ARID3A expression was dampened, whereas, CD44 and CD133 increased. ARID3A knockdown accelerated tumor growth and promoted larger tumor formation in nude-mouse xenograft model. Ki67, CD44 and CD133 were highly upregulated in knockdown tumors. SIGNIFICANCE This study demonstrated that ARID3A inhibits CRC stemness, anchorage-independent growth, self-renewal, anti-cancer drug resistance of CRC cells and tumor growth in vivo.
Collapse
Affiliation(s)
- Tamal Kanti Gope
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West-Bengal 700032, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, UP 201002, India
| | - Debankur Pal
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West-Bengal 700032, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, UP 201002, India
| | - Amit Kumar Srivastava
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India
| | - Bilash Chatterjee
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India
| | - Subhankar Bose
- Cancer Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, WB, India
| | - Rupasri Ain
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West-Bengal 700032, India; Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, UP 201002, India.
| |
Collapse
|
2
|
Tao Y, Ji H, Hu W, Jiang G, Yang F, Peng X, Zhang X, Yin Y, Yuan Z, Chen D. SMARCC1 promotes M2 macrophage polarization and reduces ferroptosis in lung cancer by activating FLOT1 transcription. J Mol Med (Berl) 2025; 103:453-467. [PMID: 40108025 DOI: 10.1007/s00109-025-02531-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Grounded on the bioinformatics insights, this study explores the role of flotillin 1 (FLOT1) in modulating macrophage phenotype and immune evasion in lung cancer cells. The bioinformatics analyses revealed positive correlations between FLOT1 expression and infiltration of M2 macrophages, neutrophils, dendritic cells, and CD4 memory T cells. Furthermore, elevated FLOT1 expression was associated with a poor prognosis in lung cancer patients. Analysis of tumor and adjacent non-tumor tissues from 53 lung cancer patients revealed significantly higher immunohistochemical staining of FLOT1 in tumor tissues, showing positive correlation with the staining intensity of PD-L1. Additionally, staining intensities for markers of M2 macrophages (Arg1), CD4 memory T cells (CD4), dendritic cells (CD83), and neutrophils (CD177) were significantly higher in tumor tissues with high FLOT1 levels. Silencing of FLOT1 was induced in two lung cancer cell lines. Co-culturing in conditioned media of the FLOT1-silenced cancer cells led to reduced chemotactic migration and M2 skewing of macrophages in vitro. Using xenograft models, we observed that FLOT1 silencing weakened tumorigenic activity of A549 cells in mice and reduced M2 macrophage infiltration in tumors. SWI/SNF related BAF chromatin remodeling complex subunit C1 (SMARCC1) was identified as a transcription factor that activated FLOT1 transcription by binding to its promoter. Knockdown of SMARCC1 in lung cancer cells similarly reduced the migration and M2 polarization of macrophages as well as weakened tumorigenesis in mice. However, these effects were counteracted by FLOT1 overexpression. Further analysis of the downstream effectors of the SMARCC1/FLOT1 cascade revealed the enrichment of these factors in ferroptosis-related pathways. Mechanistically, SMARCC1 knockdown led to a decreased GSH:GSSG ratio and increased lipid peroxidation in macrophages, while FLOT1 overexpression restored these changes. Transmission electron microscopic observation revealed typical features of ferroptosis-resistant mitochondria following SMARCC1 knockdown, including fragmented or reduced cristae and increased outer membrane integrity. These mitochondrial changes were mitigated by FLOT1 overexpression. In conclusion, SMARCC1 promotes immune evasion in lung cancer by activating FLOT1 transcription. This activation enhances recruitment and M2 polarization of macrophages, and increases PD-L1 expression, reduces ferroptosis. These findings provide valuable insights into the molecular mechanisms of immune evasion and suggest potential therapeutic targets for lung cancer treatment. KEY MESSAGES: • FLOT1 is associated with poor prognosis in lung cancer patients. • Association between FLOT1 and immune cell infiltration in lung cancer. • Silencing FLOT1 inhibits the recruitment of macrophages by lung cancer cells. • SMARCC1 is highly expressed in lung cancer and promotes the transcription of FLOT1. • FLOT1 overexpression rescues the inhibitory effect of SMARCC1 knockdown on M2 macrophage infiltration and activation of Ferroptosis.
Collapse
Affiliation(s)
- Youliang Tao
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Huafeng Ji
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Wensheng Hu
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Guojun Jiang
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Fangding Yang
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Xu Peng
- Department of Orthopedics, The First People's Hospital of Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Xu Zhang
- Department of General Surgery, The First People's Hospital of Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Yuqin Yin
- Department of Nephrology, The First People's Hospital of Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Zhize Yuan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Rd, Shanghai, 200433, China.
| | - Dukai Chen
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China.
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China.
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China.
| |
Collapse
|
3
|
Weinelt N, Wächtershäuser KN, Celik G, Jeiler B, Gollin I, Zein L, Smith S, Andrieux G, Das T, Roedig J, Feist L, Rotter B, Boerries M, Pampaloni F, van Wijk SJL. LUBAC-mediated M1 Ub regulates necroptosis by segregating the cellular distribution of active MLKL. Cell Death Dis 2024; 15:77. [PMID: 38245534 PMCID: PMC10799905 DOI: 10.1038/s41419-024-06447-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/22/2023] [Accepted: 01/05/2024] [Indexed: 01/22/2024]
Abstract
Plasma membrane accumulation of phosphorylated mixed lineage kinase domain-like (MLKL) is a hallmark of necroptosis, leading to membrane rupture and inflammatory cell death. Pro-death functions of MLKL are tightly controlled by several checkpoints, including phosphorylation. Endo- and exocytosis limit MLKL membrane accumulation and counteract necroptosis, but the exact mechanisms remain poorly understood. Here, we identify linear ubiquitin chain assembly complex (LUBAC)-mediated M1 poly-ubiquitination (poly-Ub) as novel checkpoint for necroptosis regulation downstream of activated MLKL in cells of human origin. Loss of LUBAC activity inhibits tumor necrosis factor α (TNFα)-mediated necroptosis, not by affecting necroptotic signaling, but by preventing membrane accumulation of activated MLKL. Finally, we confirm LUBAC-dependent activation of necroptosis in primary human pancreatic organoids. Our findings identify LUBAC as novel regulator of necroptosis which promotes MLKL membrane accumulation in human cells and pioneer primary human organoids to model necroptosis in near-physiological settings.
Collapse
Affiliation(s)
- Nadine Weinelt
- Institute for Experimental Paediatric Haematology and Oncology (EPHO), Goethe University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Kaja Nicole Wächtershäuser
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Biological Sciences (IZN), Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438, Frankfurt am Main, Germany
| | - Gulustan Celik
- Institute for Experimental Paediatric Haematology and Oncology (EPHO), Goethe University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Birte Jeiler
- Institute for Experimental Paediatric Haematology and Oncology (EPHO), Goethe University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Isabelle Gollin
- Institute for Experimental Paediatric Haematology and Oncology (EPHO), Goethe University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Laura Zein
- Institute for Experimental Paediatric Haematology and Oncology (EPHO), Goethe University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Sonja Smith
- Institute for Experimental Paediatric Haematology and Oncology (EPHO), Goethe University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Tonmoy Das
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
| | - Jens Roedig
- Institute for Experimental Paediatric Haematology and Oncology (EPHO), Goethe University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany
| | - Leonard Feist
- GenXPro GmbH, Altenhoeferallee 3, 60438, Frankfurt am Main, Germany
| | - Björn Rotter
- GenXPro GmbH, Altenhoeferallee 3, 60438, Frankfurt am Main, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany
- German Cancer Consortium (DKTK) partner site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Francesco Pampaloni
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Biological Sciences (IZN), Goethe University Frankfurt, Max-von-Laue-Strasse 15, 60438, Frankfurt am Main, Germany
| | - Sjoerd J L van Wijk
- Institute for Experimental Paediatric Haematology and Oncology (EPHO), Goethe University Frankfurt, Komturstrasse 3a, 60528, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) partner site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- University Cancer Centre Frankfurt (UCT), University Hospital Frankfurt, Goethe-University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
4
|
Wisniewski DJ, Liyasova MS, Korrapati S, Zhang X, Ratnayake S, Chen Q, Gilbert SF, Catalano A, Voeller D, Meerzaman D, Guha U, Porat-Shliom N, Annunziata CM, Lipkowitz S. Flotillin-2 regulates epidermal growth factor receptor activation, degradation by Cbl-mediated ubiquitination, and cancer growth. J Biol Chem 2023; 299:102766. [PMID: 36470425 PMCID: PMC9823131 DOI: 10.1016/j.jbc.2022.102766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/08/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) signaling is frequently dysregulated in various cancers. The ubiquitin ligase Casitas B-lineage lymphoma proto-oncogene (Cbl) regulates degradation of activated EGFR through ubiquitination and acts as an adaptor to recruit proteins required for trafficking. Here, we used stable isotope labeling with amino acids in cell culture mass spectrometry to compare Cbl complexes with or without epidermal growth factor (EGF) stimulation. We identified over a hundred novel Cbl interactors, and a secondary siRNA screen found that knockdown of Flotillin-2 (FLOT2) led to increased phosphorylation and degradation of EGFR upon EGF stimulation in HeLa cells. In PC9 and H441 cells, FLOT2 knockdown increased EGF-stimulated EGFR phosphorylation, ubiquitination, and downstream signaling, reversible by EGFR inhibitor erlotinib. CRISPR knockout (KO) of FLOT2 in HeLa cells confirmed EGFR downregulation, increased signaling, and increased dimerization and endosomal trafficking. Furthermore, we determined that FLOT2 interacted with both Cbl and EGFR. EGFR downregulation upon FLOT2 loss was Cbl dependent, as coknockdown of Cbl and Cbl-b restored EGFR levels. In addition, FLOT2 overexpression decreased EGFR signaling and growth. Overexpression of wildtype (WT) FLOT2, but not the soluble G2A FLOT2 mutant, inhibited EGFR phosphorylation upon EGF stimulation in HEK293T cells. FLOT2 loss induced EGFR-dependent proliferation and anchorage-independent growth. Lastly, FLOT2 KO increased tumor formation and tumor volume in nude mice and NSG mice, respectively. Together, these data demonstrated that FLOT2 negatively regulated EGFR activation and dimerization, as well as its subsequent ubiquitination, endosomal trafficking, and degradation, leading to reduced proliferation in vitro and in vivo.
Collapse
Affiliation(s)
- David J Wisniewski
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Mariya S Liyasova
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Soumya Korrapati
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Xu Zhang
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Shashikala Ratnayake
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - Qingrong Chen
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - Samuel F Gilbert
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Alexis Catalano
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Donna Voeller
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Daoud Meerzaman
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - Udayan Guha
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Natalie Porat-Shliom
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Christina M Annunziata
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
| |
Collapse
|
5
|
Genest M, Comunale F, Planchon D, Govindin P, Noly D, Vacher S, Bièche I, Robert B, Malhotra H, Schoenit A, Tashireva LA, Casas J, Gauthier-Rouvière C, Bodin S. Upregulated flotillins and sphingosine kinase 2 derail AXL vesicular traffic to promote epithelial-mesenchymal transition. J Cell Sci 2022; 135:274986. [PMID: 35394045 DOI: 10.1242/jcs.259178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 02/15/2022] [Indexed: 12/14/2022] Open
Abstract
Altered endocytosis and vesicular trafficking are major players during tumorigenesis. Flotillin overexpression, a feature observed in many invasive tumors and identified as a marker of poor prognosis, induces a deregulated endocytic and trafficking pathway called upregulated flotillin-induced trafficking (UFIT). Here, we found that in non-tumoral mammary epithelial cells, induction of the UFIT pathway promotes epithelial-to-mesenchymal transition (EMT) and accelerates the endocytosis of several transmembrane receptors, including AXL, in flotillin-positive late endosomes. AXL overexpression, frequently observed in cancer cells, is linked to EMT and metastasis formation. In flotillin-overexpressing non-tumoral mammary epithelial cells and in invasive breast carcinoma cells, we found that the UFIT pathway-mediated AXL endocytosis allows its stabilization and depends on sphingosine kinase 2, a lipid kinase recruited in flotillin-rich plasma membrane domains and endosomes. Thus, the deregulation of vesicular trafficking following flotillin upregulation, and through sphingosine kinase 2, emerges as a new mechanism of AXL overexpression and EMT-inducing signaling pathway activation.
Collapse
Affiliation(s)
- Mallory Genest
- CRBM, University of Montpellier, CNRS, 1919 route de Mende, 34293 Montpellier, France
| | - Franck Comunale
- CRBM, University of Montpellier, CNRS, 1919 route de Mende, 34293 Montpellier, France
| | - Damien Planchon
- CRBM, University of Montpellier, CNRS, 1919 route de Mende, 34293 Montpellier, France
| | - Pauline Govindin
- CRBM, University of Montpellier, CNRS, 1919 route de Mende, 34293 Montpellier, France
| | - Dune Noly
- CRBM, University of Montpellier, CNRS, 1919 route de Mende, 34293 Montpellier, France
| | - Sophie Vacher
- Department of Genetics, Institut Curie, Paris 75005, France
| | - Ivan Bièche
- Department of Genetics, Institut Curie, Paris 75005, France
| | - Bruno Robert
- IRCM, Campus Val d'Aurelle, 208 avenue des Apothicaires, 34298 Montpellier, France
| | - Himanshu Malhotra
- CRBM, University of Montpellier, CNRS, 1919 route de Mende, 34293 Montpellier, France
| | - Andreas Schoenit
- CRBM, University of Montpellier, CNRS, 1919 route de Mende, 34293 Montpellier, France
| | - Liubov A Tashireva
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634050, Russia
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC), Spanish Council for Scientific Research (CSIC), 08034 Barcelona, Spain.,Liver and Digestive Diseases Networking Biomedical Research Centre (CIBER-EHD), 28029 Madrid, Spain
| | | | - Stéphane Bodin
- CRBM, University of Montpellier, CNRS, 1919 route de Mende, 34293 Montpellier, France
| |
Collapse
|
6
|
Liang K, Mei S, Gao X, Peng S, Zhan J. Dynamics of Endocytosis and Degradation of Antibody-Drug Conjugate T-DM1 in HER2 Positive Cancer Cells. Drug Des Devel Ther 2022; 15:5135-5150. [PMID: 34992350 PMCID: PMC8713712 DOI: 10.2147/dddt.s344052] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/14/2021] [Indexed: 12/29/2022] Open
Abstract
Purpose T-DM1 is an antibody–drug conjugate (ADC) consisting of trastuzumab and DM1 linked together. T-DM1 binds to human epidermal growth factor receptor-2 (HER2) in tumors and then triggers the endocytosis of T-DM1 and release of payload. Therefore, endocytosis efficacy is considered as a critical step for the initiation of T-DM1 therapy; however, the endocytosis mechanism of T-DM1 remains poorly understood. Meanwhile, HER2 is regarded as an internalization-resistant receptor, which hinders the endocytosis and effectiveness of T-DM1. The present study is to explore the T-DM1 endocytosis pathway, which may provide insights into the internalization mechanism of ADCs and help to improve efficacy. Methods Confocal microscopy and flow cytometry were used to analyse T-DM1 intracellular trafficking and endocytosis efficiency, while Western blot assay was performed to detect T-DM1 degradation. Results We found that intracellular T-DM1 was increased to 50% within 12 h. T-DM1 was colocalized with cholera toxin B (CTxB), a lipid raft marker, within 2 h and then degraded in lysosome. Upon overexpression of caveolin-1 (CAV-1) and utilization of caveolae/lipid-raft disruptors, we found that temporal CAV-1 upregulation significantly facilitated T-DM1 endocytosis and degradation, whereas nystatin and lovastatin disrupted caveolae/lipid-raft structure and inhibited T-DM1 degradation. We demonstrate that T-DM1 internalizes through the lipid raft-mediated endocytosis in a CAV-1 dependent manner, rather than through the clathrin-mediated endocytosis in HER2-positive cancer cells. Conclusion Our findings suggest that modulation of the caveolae/lipid-raft mediated endocytosis may be a possible option for improving the clinical therapeutic effect of T-DM1 because it plays a key role in regulating T-DM1 internalization.
Collapse
Affiliation(s)
- Keying Liang
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Shengsheng Mei
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Xiangzheng Gao
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Shanshan Peng
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Jinbiao Zhan
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou, 310058, People's Republic of China
| |
Collapse
|
7
|
Jeong J, Shin JH, Li W, Hong JY, Lim J, Hwang JY, Chung JJ, Yan Q, Liu Y, Choi J, Wysolmerski J. MAL2 mediates the formation of stable HER2 signaling complexes within lipid raft-rich membrane protrusions in breast cancer cells. Cell Rep 2021; 37:110160. [PMID: 34965434 PMCID: PMC8762588 DOI: 10.1016/j.celrep.2021.110160] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/16/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
The lipid raft-resident protein, MAL2, has been implicated as contributing to the pathogenesis of several malignancies, including breast cancer, but the underlying mechanism for its effects on tumorigenesis is unknown. Here, we show that MAL2-mediated lipid raft formation leads to HER2 plasma membrane retention and enhanced HER2 signaling in breast cancer cells. We demonstrate physical interactions between HER2 and MAL2 in lipid rafts using proximity ligation assays. Super-resolution structured illumination microscopy imaging displays the structural organization of the HER2/Ezrin/NHERF1/PMCA2 protein complex. Formation of this protein complex maintains low intracellular calcium concentrations in the vicinity of the plasma membrane. HER2/MAL2 protein interactions in lipid rafts are enhanced in trastuzumab-resistant breast cancer cells. Our findings suggest that MAL2 is crucial for lipid raft formation, HER2 signaling, and HER2 membrane stability in breast cancer cells, suggesting MAL2 as a potential therapeutic target. Jeong et al. show that the formation of MAL2-mediated lipid raft-rich membrane protrusions is crucial for HER2 signaling in breast cancer cells. MAL2 is required for the formation of HER2/Ezrin/NHERF1/PMCA2 protein complexes. Formation of these protein complexes leads to a low calcium environment in the plasma membrane
Collapse
|
8
|
Hammood M, Craig AW, Leyton JV. Impact of Endocytosis Mechanisms for the Receptors Targeted by the Currently Approved Antibody-Drug Conjugates (ADCs)-A Necessity for Future ADC Research and Development. Pharmaceuticals (Basel) 2021; 14:ph14070674. [PMID: 34358100 PMCID: PMC8308841 DOI: 10.3390/ph14070674] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
Biologically-based therapies increasingly rely on the endocytic cycle of internalization and exocytosis of target receptors for cancer therapies. However, receptor trafficking pathways (endosomal sorting (recycling, lysosome localization) and lateral membrane movement) are often dysfunctional in cancer. Antibody-drug conjugates (ADCs) have revitalized the concept of targeted chemotherapy by coupling inhibitory antibodies to cytotoxic payloads. Significant advances in ADC technology and format, and target biology have hastened the FDA approval of nine ADCs (four since 2019). Although the links between aberrant endocytic machinery and cancer are emerging, the impact of dysregulated internalization processes of ADC targets and response rates or resistance have not been well studied. This is despite the reliance on ADC uptake and trafficking to lysosomes for linker cleavage and payload release. In this review, we describe what is known about all the target antigens for the currently approved ADCs. Specifically, internalization efficiency and relevant intracellular sorting activities are described for each receptor under normal processes, and when complexed to an ADC. In addition, we discuss aberrant endocytic processes that have been directly linked to preclinical ADC resistance mechanisms. The implications of endocytosis in regard to therapeutic effectiveness in the clinic are also described. Unexpectedly, information on endocytosis is scarce (absent for two receptors). Moreover, much of what is known about endocytosis is not in the context of receptor-ADC/antibody complexes. This review provides a deeper understanding of the pertinent principles of receptor endocytosis for the currently approved ADCs.
Collapse
Affiliation(s)
- Manar Hammood
- Departément de Medécine Nucléaire et Radiobiologie, Faculté de Medécine et des Sciences de la Santé, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Andrew W. Craig
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Jeffrey V. Leyton
- Departément de Medécine Nucléaire et Radiobiologie, Faculté de Medécine et des Sciences de la Santé, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
- Centre d’Imagerie Moleculaire, Centre de Recherche, CHUS, Sherbrooke, QC J1H 5N4, Canada
- Correspondence: ; Tel.: +1-819-346-1110
| |
Collapse
|
9
|
Santamaria S, Gagliani MC, Bellese G, Marconi S, Lechiara A, Dameri M, Aiello C, Tagliatti E, Castagnola P, Cortese K. Imaging of Endocytic Trafficking and Extracellular Vesicles Released Under Neratinib Treatment in ERBB2 + Breast Cancer Cells. J Histochem Cytochem 2021; 69:461-473. [PMID: 34126793 PMCID: PMC8246527 DOI: 10.1369/00221554211026297] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancers (BCa) with ERBB2 amplification show rapid tumor growth, increased disease progression, and lower survival rate. Deregulated intracellular trafficking and extracellular vesicle (EVs) release are mechanisms that support cancer progression and resistance to treatments. Neratinib (NE) is a Food and Drug Administration-approved pan-ERBB inhibitor employed for the treatment of ERBB2+ BCa that blocks signaling and causes survival inhibition. However, the effects of NE on ERBB2 internalization, its trafficking to multivesicular bodies (MVBs), and the release of EVs that originate from these organelles remain poorly studied. By confocal and electron microscopy, we observed that low nanomolar doses of NE induced a modest ERBB2 internalization along with an increase of clathrin-mediated endocytosis and of the CD63+ MVB compartment in SKBR-3 cells. Furthermore, we showed in the culture supernatant two distinct EV subsets, based on their size and ERBB2 positivity: small (30-100 nm) ERBB2- EVs and large (>100 nm) ERBB2+ EVs. In particular, we found that NE increased the overall release of EVs, which displayed a reduced ERBB2 positivity compared with controls. Taken together, these results provide novel insight into the effects of NE on ERBB2+ BCa cells that may lead to a reduction of ERBB2 potentially transferred to distant target cells by EVs.
Collapse
Affiliation(s)
- Sara Santamaria
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Lab, Università di Genova, Genova, Italy
| | - Maria Cristina Gagliani
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Lab, Università di Genova, Genova, Italy
| | - Grazia Bellese
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Lab, Università di Genova, Genova, Italy
| | - Silvia Marconi
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Lab, Università di Genova, Genova, Italy
| | - Anastasia Lechiara
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Lab, Università di Genova, Genova, Italy
| | - Martina Dameri
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Lab, Università di Genova, Genova, Italy
| | - Cinzia Aiello
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Erica Tagliatti
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Katia Cortese
- DIMES, Department of Experimental Medicine, Cellular Electron Microscopy Lab, Università di Genova, Genova, Italy
| |
Collapse
|
10
|
von Zastrow M, Sorkin A. Mechanisms for Regulating and Organizing Receptor Signaling by Endocytosis. Annu Rev Biochem 2021; 90:709-737. [PMID: 33606955 DOI: 10.1146/annurev-biochem-081820-092427] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intricate relationships between endocytosis and cellular signaling, first recognized nearly 40 years ago through the study of tyrosine kinase growth factor receptors, are now known to exist for multiple receptor classes and to affect myriad physiological and developmental processes. This review summarizes our present understanding of how endocytosis orchestrates cellular signaling networks, with an emphasis on mechanistic underpinnings and focusing on two receptor classes-tyrosine kinase and G protein-coupled receptors-that have been investigated in particular detail. Together, these examples provide a useful survey of the current consensus, uncertainties, and controversies in this rapidly advancing area of cell biology.
Collapse
Affiliation(s)
- Mark von Zastrow
- Department of Psychiatry, University of California, San Francisco, California 94143, USA;
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, USA;
| |
Collapse
|
11
|
Abstract
Flotillins 1 and 2 are two ubiquitous, highly conserved homologous proteins that assemble to form heterotetramers at the cytoplasmic face of the plasma membrane in cholesterol- and sphingolipid-enriched domains. Flotillin heterotetramers can assemble into large oligomers to form molecular scaffolds that regulate the clustering of at the plasma membrane and activity of several receptors. Moreover, flotillins are upregulated in many invasive carcinomas and also in sarcoma, and this is associated with poor prognosis and metastasis formation. When upregulated, flotillins promote plasma membrane invagination and induce an endocytic pathway that allows the targeting of cargo proteins in the late endosomal compartment in which flotillins accumulate. These late endosomes are not degradative, and participate in the recycling and secretion of protein cargos. The cargos of this Upregulated Flotillin–Induced Trafficking (UFIT) pathway include molecules involved in signaling, adhesion, and extracellular matrix remodeling, thus favoring the acquisition of an invasive cellular behavior leading to metastasis formation. Thus, flotillin presence from the plasma membrane to the late endosomal compartment influences the activity, and even modifies the trafficking and fate of key protein cargos, favoring the development of diseases, for instance tumors. This review summarizes the current knowledge on flotillins and their role in cancer development focusing on their function in cellular membrane remodeling and vesicular trafficking regulation.
Collapse
|
12
|
Clathrin- and dynamin-dependent endocytosis limits canonical NF-κB signaling triggered by lymphotoxin β receptor. Cell Commun Signal 2020; 18:176. [PMID: 33148272 PMCID: PMC7640449 DOI: 10.1186/s12964-020-00664-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/18/2020] [Indexed: 02/08/2023] Open
Abstract
Background Lymphotoxin β receptor (LTβR) is a member of tumor necrosis factor receptor (TNFR) superfamily which regulates the immune response. At the cellular level, upon ligand binding, the receptor activates the pro-inflammatory NF-κB and AP-1 pathways. Yet, the intracellular distribution of LTβR, the routes of its endocytosis and their connection to the signaling activation are not characterized. Here, we investigated the contribution of LTβR internalization to its signaling potential. Methods Intracellular localization of LTβR in unstimulated and stimulated cells was analyzed by confocal microscopy. Endocytosis impairment was achieved through siRNA- or CRISPR/Cas9-mediated depletion, or chemical inhibition of proteins regulating endocytic routes. The activation of LTβR-induced signaling was examined. The levels of effector proteins of the canonical and non-canonical branches of the NF-κB pathway, and the phosphorylation of JNK, Akt, ERK1/2, STAT1 and STAT3 involved in diverse signaling cascades, were measured by Western blotting. A transcriptional response to LTβR stimulation was assessed by qRT-PCR analysis. Results We demonstrated that LTβR was predominantly present on endocytic vesicles and the Golgi apparatus. The ligand-bound pool of the receptor localized to endosomes and was trafficked towards lysosomes for degradation. Depletion of regulators of different endocytic routes (clathrin-mediated, dynamin-dependent or clathrin-independent) resulted in the impairment of LTβR internalization, indicating that this receptor uses multiple entry pathways. Cells deprived of clathrin and dynamins exhibited enhanced activation of canonical NF-κB signaling represented by increased degradation of IκBα inhibitor and elevated expression of LTβR target genes. We also demonstrated that clathrin and dynamin deficiency reduced to some extent LTβR-triggered activation of the non-canonical branch of the NF-κB pathway. Conclusions Our work shows that the impairment of clathrin- and dynamin-dependent internalization amplifies a cellular response to LTβR stimulation. We postulate that receptor internalization restricts responsiveness of the cell to subthreshold stimuli. Video Abstract
Graphical abstract ![]()
Supplementary information Supplementary information accompanies this paper at 10.1186/s12964-020-00664-0.
Collapse
|
13
|
Abstract
Several studies have demonstrated interactions between the two leaflets in membrane bilayers and the importance of specific lipid species for such interaction and membrane function. We here discuss these investigations with a focus on the sphingolipid and cholesterol-rich lipid membrane domains called lipid rafts, including the small flask-shaped invaginations called caveolae, and the importance of such membrane structures in cell biology and cancer. We discuss the possible interactions between the very long-chain sphingolipids in the outer leaflet of the plasma membrane and the phosphatidylserine species PS 18:0/18:1 in the inner leaflet and the importance of cholesterol for such interactions. We challenge the view that lipid rafts contain a large fraction of lipids with two saturated fatty acyl groups and argue that it is important in future studies of membrane models to use asymmetric membrane bilayers with lipid species commonly found in cellular membranes. We also discuss the need for more quantitative lipidomic studies in order to understand membrane function and structure in general, and the importance of lipid rafts in biological systems. Finally, we discuss cancer-related changes in lipid rafts and lipid composition, with a special focus on changes in glycosphingolipids and the possibility of using lipid therapy for cancer treatment.
Collapse
Affiliation(s)
- Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway
| | - Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Ullernchausséen 70, 0379, Oslo, Norway.
- Department of Biosciences, University of Oslo, 0316, Oslo, Norway.
| |
Collapse
|
14
|
Flotillins: At the Intersection of Protein S-Palmitoylation and Lipid-Mediated Signaling. Int J Mol Sci 2020; 21:ijms21072283. [PMID: 32225034 PMCID: PMC7177705 DOI: 10.3390/ijms21072283] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
Flotillin-1 and flotillin-2 are ubiquitously expressed, membrane-associated proteins involved in multifarious cellular events from cell signaling, endocytosis, and protein trafficking to gene expression. They also contribute to oncogenic signaling. Flotillins bind the cytosolic leaflet of the plasma membrane and endomembranes and, upon hetero-oligomerization, serve as scaffolds facilitating the assembly of multiprotein complexes at the membrane-cytosol interface. Additional functions unique to flotillin-1 have been discovered recently. The membrane-binding of flotillins is regulated by S-palmitoylation and N-myristoylation, hydrophobic interactions involving specific regions of the polypeptide chain and, to some extent, also by their oligomerization. All these factors endow flotillins with an ability to associate with the sphingolipid/cholesterol-rich plasma membrane domains called rafts. In this review, we focus on the critical input of lipids to the regulation of the flotillin association with rafts and thereby to their functioning. In particular, we discuss how the recent developments in the field of protein S-palmitoylation have contributed to the understanding of flotillin1/2-mediated processes, including endocytosis, and of those dependent exclusively on flotillin-1. We also emphasize that flotillins affect directly or indirectly the cellular levels of lipids involved in diverse signaling cascades, including sphingosine-1-phosphate and PI(4,5)P2. The mutual relations between flotillins and distinct lipids are key to the regulation of their involvement in numerous cellular processes.
Collapse
|
15
|
Indira Chandran V, Månsson AS, Barbachowska M, Cerezo-Magaña M, Nodin B, Joshi B, Koppada N, Saad OM, Gluz O, Isaksson K, Borgquist S, Jirström K, Nabi IR, Jernström H, Belting M. Hypoxia Attenuates Trastuzumab Uptake and Trastuzumab-Emtansine (T-DM1) Cytotoxicity through Redistribution of Phosphorylated Caveolin-1. Mol Cancer Res 2020; 18:644-656. [PMID: 31900313 DOI: 10.1158/1541-7786.mcr-19-0856] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/28/2019] [Accepted: 12/20/2019] [Indexed: 11/16/2022]
Abstract
The antibody-drug conjugate trastuzumab-emtansine (T-DM1) offers an additional treatment option for patients with HER2-amplified tumors. However, primary and acquired resistance is a limiting factor in a significant subset of patients. Hypoxia, a hallmark of cancer, regulates the trafficking of several receptor proteins with potential implications for tumor targeting. Here, we have investigated how hypoxic conditions may regulate T-DM1 treatment efficacy in breast cancer. The therapeutic effect of T-DM1 and its metabolites was evaluated in conjunction with biochemical, flow cytometry, and high-resolution imaging studies to elucidate the functional and mechanistic aspects of hypoxic regulation. HER2 and caveolin-1 expression was investigated in a well-annotated breast cancer cohort. We find that hypoxia fosters relative resistance to T-DM1 in HER2+ cells (SKBR3 and BT474). This effect was not a result of deregulated HER2 expression or resistance to emtansine and its metabolites. Instead, we show that hypoxia-induced translocation of caveolin-1 from cytoplasmic vesicles to the plasma membrane contributes to deficient trastuzumab internalization and T-DM1 chemosensitivity. Caveolin-1 depletion mimicked the hypoxic situation, indicating that vesicular caveolin-1 is indispensable for trastuzumab uptake and T-DM1 cytotoxicity. In vitro studies suggested that HER2 and caveolin-1 are not coregulated, which was supported by IHC analysis in patient tumors. We find that phosphorylation-deficient caveolin-1 inhibits trastuzumab internalization and T-DM1 cytotoxicity, suggesting a specific role for caveolin-1 phosphorylation in HER2 trafficking. IMPLICATIONS: Together, our data for the first time identify hypoxic regulation of caveolin-1 as a resistance mechanism to T-DM1 with potential implications for individualized treatment of breast cancer.
Collapse
Affiliation(s)
- Vineesh Indira Chandran
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden.
| | - Ann-Sofie Månsson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Magdalena Barbachowska
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Myriam Cerezo-Magaña
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Björn Nodin
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Bharat Joshi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Neelima Koppada
- BioAnalytical Sciences-ADT, gRED, Genentech, South San Francisco, California
| | - Ola M Saad
- BioAnalytical Sciences-ADT, gRED, Genentech, South San Francisco, California
| | - Oleg Gluz
- West German Study Group, Moenchengladbach, Germany
| | - Karolin Isaksson
- Division of Surgery, Lund University, Skåne University Hospital, Lund, Central Hospital, Kristianstad, Sweden
| | - Signe Borgquist
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
- Department of Oncology, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
| | - Karin Jirström
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Ivan Robert Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Helena Jernström
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Mattias Belting
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden.
- Department of Hematology, Oncology and Radiophysics, Skåne University Hospital, Lund, Sweden
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
Baig N, Li Z, Lu J, Chen H, Yu S, Li T, Niu Z, Niu J. Clinical significance and comparison of flotillin 1 expression in left and right colon cancer. Oncol Lett 2019; 18:997-1004. [PMID: 31423159 PMCID: PMC6607386 DOI: 10.3892/ol.2019.10401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 09/28/2018] [Indexed: 12/29/2022] Open
Abstract
Flotillin 1 (FLOT1) is increasingly implicated in various types of cancer, and has been reported to influence tumorigenesis and cancer progression, leading to poor prognosis for survival time; however, its expression in colorectal cancer (CRC) and its influence on various clinicopathological parameters of this disease remain unknown. In the present study, FLOT1 expression and its effect on different clinicopathological parameters were assessed immunohistochemically and histologically in 81 CRC and 81 non-tumorous colon tissue samples. The immunohistochemical staining was scored semi-quantitatively. The association of FLOT1 expression with various parameters and its effect on overall survival time was also assessed. FLOT1 was upregulated in the CRC tissue, with increased expression in the right colon tissue samples compared with those of left colon. Increased FLOT1 expression in CRC tissue samples was associated with tumor volume, differentiation, tumor grade and poor overall survival time. In the right colon tissue samples in particular, there was a notable association with tumor volume and grade, indicating its effect on proliferation and tumor stage at this site. A multivariate Cox regression hazard analysis revealed that only tumor grade and differentiation were the independent predictors of overall survival time in patients with CRC. Together, the results of the present study suggest that FLOT1 serves important functions in the proliferation and progression of CRC, contributes to decreased survival time, and may serve as a novel therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Nadia Baig
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zequn Li
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Jing Lu
- Department of Chemotherapy, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Haibin Chen
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Songyang Yu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Tianen Li
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhaojian Niu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Jun Niu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
17
|
Sugiyama MG, Fairn GD, Antonescu CN. Akt-ing Up Just About Everywhere: Compartment-Specific Akt Activation and Function in Receptor Tyrosine Kinase Signaling. Front Cell Dev Biol 2019; 7:70. [PMID: 31131274 PMCID: PMC6509475 DOI: 10.3389/fcell.2019.00070] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
The serine/threonine kinase Akt is a master regulator of many diverse cellular functions, including survival, growth, metabolism, migration, and differentiation. Receptor tyrosine kinases are critical regulators of Akt, as a result of activation of phosphatidylinositol-3-kinase (PI3K) signaling leading to Akt activation upon receptor stimulation. The signaling axis formed by receptor tyrosine kinases, PI3K and Akt, as well as the vast range of downstream substrates is thus central to control of cell physiology in many different contexts and tissues. This axis must be tightly regulated, as disruption of PI3K-Akt signaling underlies the pathology of many diseases such as cancer and diabetes. This sophisticated regulation of PI3K-Akt signaling is due in part to the spatial and temporal compartmentalization of Akt activation and function, including in specific nanoscale domains of the plasma membrane as well as in specific intracellular membrane compartments. Here, we review the evidence for localized activation of PI3K-Akt signaling by receptor tyrosine kinases in various specific cellular compartments, as well as that of compartment-specific functions of Akt leading to control of several fundamental cellular processes. This spatial and temporal control of Akt activation and function occurs by a large number of parallel molecular mechanisms that are central to regulation of cell physiology.
Collapse
Affiliation(s)
- Michael G. Sugiyama
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| | - Gregory D. Fairn
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Costin N. Antonescu
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
18
|
Zhang J, Li Q, Wu Y, Wang D, Xu L, Zhang Y, Wang S, Wang T, Liu F, Zaky MY, Hou S, Liu S, Zou K, Lei H, Zou L, Zhang Y, Liu H. Cholesterol content in cell membrane maintains surface levels of ErbB2 and confers a therapeutic vulnerability in ErbB2-positive breast cancer. Cell Commun Signal 2019; 17:15. [PMID: 30786890 PMCID: PMC6383291 DOI: 10.1186/s12964-019-0328-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/13/2019] [Indexed: 11/30/2022] Open
Abstract
Background ErbB2 overexpression identifies a subset of breast cancer as ErbB2-positive and is frequently associated with poor clinical outcomes. As a membrane-embedded receptor tyrosine kinase, cell surface levels of ErbB2 are regulated dynamically by membrane physical properties. The present study aims to investigate the influence of membrane cholesterol contents on ErbB2 status and cellular responses to its tyrosine kinase inhibitors. Methods The cholesterol abundance was examined in ErbB2-positive breast cancer cells using filipin staining. Cellular ErbB2 localizations were investigated by immunofluorescence with altered membrane cholesterol contents. The inhibitory effects of the cholesterol-lowering drug lovastatin were assessed using cell proliferation, apoptosis, immunoblotting and immunofluorescence assays. The synergistic effects of lovastatin with the ErbB2 inhibitor lapatinib were evaluated using an ErbB2-positive breast cancer xenograft mouse model. Results Membrane cholesterol contents positively correlated with cell surface distribution of ErbB2 through increasing the rigidity and decreasing the fluidity of cell membranes. Reduction in cholesterol abundance assisted the internalization and degradation of ErbB2. The cholesterol-lowering drug lovastatin significantly potentiated the inhibitory effects of ErbB2 kinase inhibitors, accompanied with enhanced ErbB2 endocytosis. Lovastatin also synergized with lapatinib to strongly suppress the in vivo growth of ErbB2-positive breast cancer xenografts. Conclusion The cell surface distribution of ErbB2 was closely regulated by membrane physical properties governed by cholesterol contents. The cholesterol-lowering medications can hence be exploited for potential combinatorial therapies with ErbB2 kinase inhibitors in the clinical treatment of ErbB2-positive breast cancer.
Collapse
Affiliation(s)
- Jinrui Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Qiong Li
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yueguang Wu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Duchuang Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Lu Xu
- The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yang Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Shanshan Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Taishu Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Fang Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Mohamed Y Zaky
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,Molecular Physiology Division, Department of Zoology, Faculty of Science, Beni-Suef University, Beni Suef, Egypt
| | - Shuai Hou
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Shuyan Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Kun Zou
- Department of Radiotherapy Oncology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Haixin Lei
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Lijuan Zou
- The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yingqiu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| | - Han Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China. .,Cancer Biotherapy & Translational Medicine Center of Liaoning Province, Dalian Medical University, Dalian, China.
| |
Collapse
|
19
|
Christians A, Poisel E, Hartmann C, von Deimling A, Pusch S. Characterization of the epithelial membrane protein 3 interaction network reveals a potential functional link to mitogenic signal transduction regulation. Int J Cancer 2019; 145:461-473. [PMID: 30614533 DOI: 10.1002/ijc.32107] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/07/2018] [Accepted: 12/20/2018] [Indexed: 12/18/2022]
Abstract
Epithelial Membrane Protein 3 (EMP3), a 4-transmembrane glycoprotein, first gained attention as a putative tumor suppressor. Accumulating evidence, however, points to a more tumor promotive function of EMP3. The biological function of EMP3 remains largely unclear. To elucidate more of EMP3's interaction network, we performed a Yeast-Two-Hybrid (Y2H) screening, followed by validation of candidate interactors by Biomolecular Fluorescence Complementation (BiFC) and Proximity Ligation Assay (PLA). Furthermore, we generated stable EMP3 knockdown cell lines and measured cell proliferation, migration and sensitivity to apoptosis induction as well as the expression and activation levels of important signal pathway components. The Y2H screening yielded 10 novel interactions of EMP3, eight of which could also be detected by BiFC and PLA interaction assays. All newly discovered interaction partners are involved in signaling or trafficking regulation. Most notably, FLOT1 and HTATIP2 have well described roles in the regulation of EGFR signaling. In addition, knockdown of EMP3 resulted in reduced levels of p-AKT, p-ERK and p-EGFR, attenuated cell proliferation and migration and sensitized cells to apoptosis induction by TRAIL and Staurosporine. Based on these observations we hypothesize that EMP3 might be involved in the regulation of receptor-tyrosine-kinase mediated mitogenic signaling.
Collapse
Affiliation(s)
- Arne Christians
- Department of Neuropathology, Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Eric Poisel
- Department of Neuropathology, Institute of Pathology, Ruprecht-Karls-University Heidelberg, Germany.,German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Hartmann
- Department of Neuropathology, Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Ruprecht-Karls-University Heidelberg, Germany.,German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Pusch
- Department of Neuropathology, Institute of Pathology, Ruprecht-Karls-University Heidelberg, Germany.,German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
20
|
Dong Z, Cheng F, Yang Y, Zhang F, Chen G, Liu D. Expression and functional analysis of flotillins in Dugesia japonica. Exp Cell Res 2019; 374:76-84. [DOI: 10.1016/j.yexcr.2018.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 12/20/2022]
|
21
|
Liu XX, Liu WD, Wang L, Zhu B, Shi X, Peng ZX, Zhu HC, Liu XD, Zhong MZ, Xie D, Zeng MS, Ren CP. Roles of flotillins in tumors. J Zhejiang Univ Sci B 2018; 19:171-182. [PMID: 29504311 DOI: 10.1631/jzus.b1700102] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The identification and use of molecular biomarkers have greatly improved the diagnosis and treatment of malignant tumors. However, a much deeper understanding of oncogenic proteins is needed for the benefit to cancer patients. The lipid raft marker proteins, flotillin-1 and flotillin-2, were first found in goldfish retinal ganglion cells during axon regeneration. They have since been found in a variety of cells, mainly on the inner surface of cell membranes, and not only act as a skeleton to provide a platform for protein-protein interactions, but also are involved in signal transduction, nerve regeneration, endocytosis, and lymphocyte activation. Previous studies have shown that flotillins are closely associated with tumor development, invasion, and metastasis. In this article, we review the functions of flotillins in relevant cell processes, their underlying mechanisms of action in a variety of tumors, and their potential applications to tumor molecular diagnosis and targeted therapy.
Collapse
Affiliation(s)
- Xu-Xu Liu
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Central South University, Changsha 410078, China
| | - Wei-Dong Liu
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Central South University, Changsha 410078, China
| | - Lei Wang
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Central South University, Changsha 410078, China
| | - Bin Zhu
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Central South University, Changsha 410078, China
| | - Xiao Shi
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Central South University, Changsha 410078, China
| | - Zi-Xuan Peng
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Central South University, Changsha 410078, China
| | - He-Cheng Zhu
- Changsha Kexin Cancer Hospital, Changsha 410205, China
| | - Xing-Dong Liu
- Changsha Kexin Cancer Hospital, Changsha 410205, China
| | - Mei-Zuo Zhong
- Changsha Kexin Cancer Hospital, Changsha 410205, China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Cai-Ping Ren
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, Central South University, Changsha 410078, China
| |
Collapse
|
22
|
Planchon D, Rios Morris E, Genest M, Comunale F, Vacher S, Bièche I, Denisov EV, Tashireva LA, Perelmuter VM, Linder S, Chavrier P, Bodin S, Gauthier-Rouvière C. MT1-MMP targeting to endolysosomes is mediated by upregulation of flotillins. J Cell Sci 2018; 131:jcs.218925. [PMID: 30111578 DOI: 10.1242/jcs.218925] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/21/2018] [Indexed: 12/31/2022] Open
Abstract
Tumor cell invasion and metastasis formation are the major cause of death in cancer patients. These processes rely on extracellular matrix (ECM) degradation mediated by organelles termed invadopodia, to which the transmembrane matrix metalloproteinase MT1-MMP (also known as MMP14) is delivered from its reservoir, the RAB7-containing endolysosomes. How MT1-MMP is targeted to endolysosomes remains to be elucidated. Flotillin-1 and -2 are upregulated in many invasive cancers. Here, we show that flotillin upregulation triggers a general mechanism, common to carcinoma and sarcoma, which promotes RAB5-dependent MT1-MMP endocytosis and its delivery to RAB7-positive endolysosomal reservoirs. Conversely, flotillin knockdown in invasive cancer cells greatly reduces MT1-MMP accumulation in endolysosomes, its subsequent exocytosis at invadopodia, ECM degradation and cell invasion. Our results demonstrate that flotillin upregulation is necessary and sufficient to promote epithelial and mesenchymal cancer cell invasion and ECM degradation by controlling MT1-MMP endocytosis and delivery to the endolysosomal recycling compartment.
Collapse
Affiliation(s)
- Damien Planchon
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Eduardo Rios Morris
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Mallory Genest
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Franck Comunale
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | - Sophie Vacher
- Department of Genetics, Institut Curie, 75005 Paris, France
| | - Ivan Bièche
- Department of Genetics, Institut Curie, 75005 Paris, France
| | - Evgeny V Denisov
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk 634050, Russia.,Tomsk State University, Tomsk 634050, Russia
| | - Lubov A Tashireva
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk 634050, Russia
| | - Vladimir M Perelmuter
- Cancer Research Institute, Tomsk National Research Medical Center, Tomsk 634050, Russia
| | - Stefan Linder
- Institut für Medizinische Mikrobiologie, Virologie und Hygiene, University Medical Center Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Philippe Chavrier
- Cell Dynamics and Compartmentalization Unit, Institut Curie, 75005 Paris, France
| | - Stéphane Bodin
- CRBM, Univ Montpellier, CNRS, France, 1919 Route de Mende, 34293 Montpellier, France
| | | |
Collapse
|
23
|
Dietrich M, Malik MS, Nikolaysen F, Skeie M, Stang E. Protein kinase C mediated internalization of ErbB2 is independent of clathrin, ubiquitination and Hsp90 dissociation. Exp Cell Res 2018; 371:139-150. [PMID: 30098331 DOI: 10.1016/j.yexcr.2018.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
Abstract
Overexpression of ErbB2 is frequent in cancer and understanding the mechanisms which regulate its expression is important. ErbB2 is considered endocytosis resistant. It has no identified ligand, but upon heterodimerization it is a potent mediator of proliferative signaling. A recent study established a role for protein kinase C (PKC) in internalization and recycling of ErbB2. We have now further investigated the molecular mechanisms involved in PKC-mediated downregulation of ErbB2. We confirm that PMA-induced PKC activation causes ErbB2 internalization, but while the Hsp90 inhibitor 17-AAG induced ErbB2 degradation, PMA had no such effect. When combined with 17-AAG, PMA had additive effect on ErbB2 internalization indicating that Hsp90 inhibition and PKC activation induce internalization by alternative mechanisms. We confirm that while 17-AAG-induced internalization was clathrin-mediated, PMA-induced internalization was clathrin independent. This difference may be explained by while both 17-AAG and PMA reduced the constitutive tyrosine phosphorylation of ErbB2, only 17-AAG induced Hsp90 dissociation, Hsp70 recruitment and ubiquitination of ErbB2. Importantly, since PMA induced internalization of ErbB2, but not dissociation of Hsp90, Hsp90 does not per se retain ErbB2 at the plasma membrane. The morphology of the compartment into which receptors are sorted upon PKC activation has not previously been identified. By immuno-electron microscopy, we show that PMA sorts ErbB2 into a complex tubulovesicular or cisternal organelle resembling a previously described endocytic recycling compartment.
Collapse
Affiliation(s)
- Markus Dietrich
- Department of Pathology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | - Marianne Skeie
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Stang
- Department of Pathology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
24
|
Sandvig K, Kavaliauskiene S, Skotland T. Clathrin-independent endocytosis: an increasing degree of complexity. Histochem Cell Biol 2018; 150:107-118. [PMID: 29774430 PMCID: PMC6096564 DOI: 10.1007/s00418-018-1678-5] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2018] [Indexed: 11/03/2022]
Abstract
This article aims at providing an update on the complexity of clathrin-independent endocytosis. It is now almost 30 years since we first wrote a review about its existence; at that time many people believed that with the exception of macropinocytosis, which will only be briefly mentioned in this review, all uptake could be accounted for by clathrin-dependent endocytosis. Now it is generally accepted that there are different clathrin-independent mechanisms, some of them regulated by ligands and membrane lipid composition. They can be both dynamin-dependent and -independent, meaning that the uptake cannot be accounted for by caveolae and other dynamin-dependent processes such as tubular structures that can be induced by toxins, e.g. Shiga toxin, or the fast endophilin mediated endocytosis recently described. Caveolae seem to be mostly quite stable structures with other functions than endocytosis, but evidence suggests that they may have cell-type dependent functions. Although several groups have been working on endocytic mechanisms for years, and new advanced methods have improved our ability to study mechanistic details, there are still a number of important questions we need to address, such as: How many endocytic mechanisms does a cell have? How quantitatively important are they? What about the complexity in polarized cells where clathrin-independent endocytosis is differentially regulated on the apical and basolateral poles? These questions are not easy to answer since one and the same molecule may contribute to more than one process, and manipulating one mechanism can affect another. Also, several inhibitors of endocytic processes commonly used turn out to be less specific than originally thought. We will here describe the current view of clathrin-independent endocytic processes and the challenges in studying them.
Collapse
Affiliation(s)
- Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379, Oslo, Norway.
- Department of Molecular Biosciences, University of Oslo, 0316, Oslo, Norway.
| | - Simona Kavaliauskiene
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379, Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0379, Oslo, Norway
| |
Collapse
|
25
|
Deng Y, Ge P, Tian T, Dai C, Wang M, Lin S, Liu K, Zheng Y, Xu P, Zhou L, Hao Q, Dai Z. Prognostic value of flotillins (flotillin-1 and flotillin-2) in human cancers: A meta-analysis. Clin Chim Acta 2018; 481:90-98. [PMID: 29499201 DOI: 10.1016/j.cca.2018.02.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/15/2017] [Accepted: 02/27/2018] [Indexed: 01/30/2023]
Abstract
Increasing evidence indicates that flotillins which associate with cell infiltration and metastasis are overexpressed in multiple tumors. The prognostic role of flotillins remains controversial. We conducted a comprehensive meta-analysis of published research to investigate the prognostic value of flotillins in patients with cancer. Pooled HRs (hazard ratio) with 95% CIs (confidence interval) were collected to estimate the prognostic value. Twenty-seven studies with 4803 cancer patients were finally identified. The results indicated that: (1) elevated flotillins predicted poorer OS (overall survival) (HR = 2.17, 95% CI 1.87 to 2.52; HR = 1.61, 95% CI 1.44 to 1.81) and DFS (disease-free survival) (HR = 2.41, 95% CI 1.83 to 3.18; HR = 3.01, 95% CI 2.12 to 4.27) in patients with cancer; (2) Subgroup analysis showed that the prognostic value of flotillin-1 on OS and DFS in the investigated tumors were not altered by tumor type (such as digestive system cancers, renal cell cancer, lung cancer, or others), country (China or Canada), cutoff value, detection method, analysis type or paper quality and flotillin-2 overexpression indicates poor OS in human cancers except for nasopharyngeal carcinoma. Flotillins are promising as new biomarkers to predict poor prognosis of patients with tumors. This conclusion needs more clinical studies with different types of cancer to be proven.
Collapse
Affiliation(s)
- Yujiao Deng
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Pengbo Ge
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Binzhou Medical University, Binzhou 256603, China
| | - Tian Tian
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Cong Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Meng Wang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Shuai Lin
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Kang Liu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yi Zheng
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Peng Xu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Linghui Zhou
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Qian Hao
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Zhijun Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
26
|
Shramova EI, Proshkina GM, Deyev SM. The Cause of ErbB2 Receptor Resistance to Downregulation. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2018. [DOI: 10.1134/s1068162018030147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
27
|
Identification of an HSP90 modulated multi-step process for ERBB2 degradation in breast cancer cells. Oncotarget 2018; 7:85411-85429. [PMID: 27863425 PMCID: PMC5356745 DOI: 10.18632/oncotarget.13392] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/28/2016] [Indexed: 12/24/2022] Open
Abstract
The receptor tyrosine kinase ERBB2 interacts with HSP90 and is overexpressed in aggressive breast cancers. Therapeutic HSP90 inhibitors, i.e. Geldanamycin (GA), target ERBB2 to degradation. We have previously shown that HSP90 is responsible for the missorting of recycling ERBB2 to degradation compartments. In this study, we used biochemical, immunofluorescence and electron microscopy techniques to demonstrate that in SKBR3 human breast cancer cells, GA strongly induces polyubiquitination and internalization of the full-length p185-ERBB2, and promotes its cleavage, with the formation of a p116-ERBB2 form in EEA1-positive endosomes (EE). p116-ERBB2 corresponds to a non-ubiquitinated, signaling-impaired, membrane-bound fragment, which is readily sorted to lysosomes and degraded. To define the sequence of events leading to p116-ERBB2 degradation, we first blocked the EE maturation/trafficking to late endosomes/lysosomes with wortmannin, and found an increase in GA-dependent formation of p116-ERBB2; we then inhibited the proteasome activity with MG-132 or lactacystin, and observed an efficient block of p185-ERBB2 cleavage, and its accumulation in EE, suggesting that p185-ERBB2 polyubiquitination is necessary for proteasome-dependent p116-ERBB2 generation occurring in EE. As polyubiquitination has also been implicated in autophagy-mediated degradation of ERBB2 under different experimental conditions, we exploited this possibility and demonstrate that GA strongly inhibits early autophagy, and reduces the levels of the autophagy markers atg5-12 and LC3-II, irrespective of GA-induced ERBB2 polyubiquitination, ruling out a GA-dependent autophagic degradation of ERBB2. In conclusion, we propose that HSP90 inhibition fosters ERBB2 polyubiquitination and proteasome-dependent generation of a non-ubiquitinated and inactive p116-ERBB2 form in EE, which is trafficked from altered EE to lysosomes.
Collapse
|
28
|
Regulation of ErbB2 localization and function in breast cancer cells by ERM proteins. Oncotarget 2018; 7:25443-60. [PMID: 27029001 PMCID: PMC5041916 DOI: 10.18632/oncotarget.8327] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 03/10/2016] [Indexed: 12/20/2022] Open
Abstract
The ERM protein family is implicated in processes such as signal transduction, protein trafficking, cell proliferation and migration. Consequently, dysregulation of ERM proteins has been described to correlate with carcinogenesis of different cancer types. However, the underlying mechanisms are poorly understood. Here, we demonstrate a novel functional interaction between ERM proteins and the ErbB2 receptor tyrosine kinase in breast cancer cells. We show that the ERM proteins ezrin and radixin are associated with ErbB2 receptors at the plasma membrane, and depletion or functional inhibition of ERM proteins destabilizes the interaction of ErbB2 with ErbB3, Hsp90 and Ebp50. Accompanied by the dissociation of this protein complex, binding of ErbB2 to the ubiquitin-ligase c-Cbl is increased, and ErbB2 becomes dephosphorylated, ubiquitinated and internalized. Furthermore, signaling via Akt- and Erk-mediated pathways is impaired upon ERM inhibition. Finally, interference with ERM functionality leads to receptor degradation and reduced cellular levels of ErbB2 and ErbB3 receptors in breast cancer cells.
Collapse
|
29
|
Liu R, Yang G, Zhou MH, He Y, Mei YA, Ding Y. Flotillin-1 downregulates K(+) current by directly coupling with Kv2.1 subunit. Protein Cell 2017; 7:455-60. [PMID: 27221754 PMCID: PMC4887332 DOI: 10.1007/s13238-016-0276-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Rui Liu
- School of Life Sciences, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200438, China
| | - Guang Yang
- School of Life Sciences, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200438, China
| | - Meng-Hua Zhou
- School of Life Sciences, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200438, China
| | - Yu He
- School of Life Sciences, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200438, China
| | - Yan-Ai Mei
- School of Life Sciences, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200438, China.
| | - Yu Ding
- School of Life Sciences, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
30
|
Alaimo S, Marceca GP, Ferro A, Pulvirenti A. Detecting Disease Specific Pathway Substructures through an Integrated Systems Biology Approach. Noncoding RNA 2017; 3:ncrna3020020. [PMID: 29657291 PMCID: PMC5831934 DOI: 10.3390/ncrna3020020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 03/28/2017] [Accepted: 04/10/2017] [Indexed: 12/14/2022] Open
Abstract
In the era of network medicine, pathway analysis methods play a central role in the prediction of phenotype from high throughput experiments. In this paper, we present a network-based systems biology approach capable of extracting disease-perturbed subpathways within pathway networks in connection with expression data taken from The Cancer Genome Atlas (TCGA). Our system extends pathways with missing regulatory elements, such as microRNAs, and their interactions with genes. The framework enables the extraction, visualization, and analysis of statistically significant disease-specific subpathways through an easy to use web interface. Our analysis shows that the methodology is able to fill the gap in current techniques, allowing a more comprehensive analysis of the phenomena underlying disease states.
Collapse
Affiliation(s)
- Salvatore Alaimo
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, c/o Dipartimento di Matematica e Informatica, Viale A. Doria 6, 95125 Catania, Italy.
| | - Gioacchino Paolo Marceca
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, c/o Dipartimento di Matematica e Informatica, Viale A. Doria 6, 95125 Catania, Italy.
| | - Alfredo Ferro
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, c/o Dipartimento di Matematica e Informatica, Viale A. Doria 6, 95125 Catania, Italy.
| | - Alfredo Pulvirenti
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, c/o Dipartimento di Matematica e Informatica, Viale A. Doria 6, 95125 Catania, Italy.
| |
Collapse
|
31
|
Ou YX, Liu FT, Chen FY, Zhu ZM. Prognostic value of Flotillin-1 expression in patients with solid tumors. Oncotarget 2017; 8:52665-52677. [PMID: 28881760 PMCID: PMC5581059 DOI: 10.18632/oncotarget.17075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/16/2017] [Indexed: 12/01/2022] Open
Abstract
Background In numerous studies, Flotillin-1 was reported to be involved in tumor progression, indicating prognosis in various types of cancer. However, the results were inconsistent. Results A total of 2473 patients from 13 articles were included. The results indicated that: (1) Patients detected with high expression level of Flotillin-1 protein had a significantly shorter OS (HR =1.64; 95%CI: 1.39-1.88), statistical significance was also observed in subgroup meta-analyses stratified by the cancer type, nationality, detecting method, cutoff value, analysis type, sample size and publication date. (2) Patients with high Flotillin-1 protein expression level had a poorer DFS (HR = 2.49; 95%CI: 1.64-3.35), a worse RFS(HR = 3.26; 95%CI: 1.10-5.43) and a potential shorter PFS(HR = 1.84; 95%CI: 0.81-2.87). (3) The pooled odds ratios (ORs) showed that increased Flotillin-1 level was also related to lymph node metastasis (OR =6.30; 95% CI: 3.15-12.59), distant metastasis (OR =6.02; 95% CI: 1.50-24.06) and more advanced TNM stage (OR =4.69; 95% CI: 2.74-8.03). Materials and methods A comprehensive retrieval was performed in multiple databases, including PubMed, Embase, Web of Science and CNKI. The relevant articles were screened for investigating the association between increased Flotillin-1 expression level and prognosis. Additionally, clinicopathological features data was also extracted from these studies. Conclusions High expression level of Flotillin-1 protein was correlated with poorer clinical outcome. It might serve as a prognostic biomarker and a potential predictive factor of clinicopathology in various tumors. Further well-designed clinical studies should be performed to verify the clinical utility of Flotillin-1 in human solid tumors.
Collapse
Affiliation(s)
- Yang-Xi Ou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P. R. China
| | - Fang-Teng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P. R. China.,Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, P. R. China
| | - Fang-Ying Chen
- The Health Centers of Fengzhou Town, Quanzhou 36200, Fujian Province, P. R. China
| | - Zheng-Ming Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, Jiangxi Province, P. R. China
| |
Collapse
|
32
|
Jeong J, Kim W, Kim LK, VanHouten J, Wysolmerski JJ. HER2 signaling regulates HER2 localization and membrane retention. PLoS One 2017; 12:e0174849. [PMID: 28369073 PMCID: PMC5378417 DOI: 10.1371/journal.pone.0174849] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/16/2017] [Indexed: 01/03/2023] Open
Abstract
ErbB2/HER2/Neu is a receptor tyrosine kinase that is overexpressed in 25-30% of human breast cancers, usually associated with amplification of the ERBB2 gene. HER2 has no recognized ligands and heterodimers between HER2 and EGFR (ErbB1/HER1) or HER2 and ErbB3/HER3 are important in breast cancer. Unlike other ErbB family members, HER2 is resistant to internalization and degradation, and remains at the cell surface to signal for prolonged periods after it is activated. Although the mechanisms underlying retention of HER2 at the cell surface are not fully understood, prior studies have shown that, in order to avoid internalization, HER2 must interact with the chaperone, HSP90, and the calcium pump, PMCA2, within specific plasma membrane domains that protrude from the cell surface. In this report, we demonstrate that HER2 signaling, itself, is important for the formation and maintenance of membrane protrusions, at least in part, by maintaining PMCA2 expression and preventing increased intracellular calcium concentrations. Partial genetic knockdown of HER2 expression or pharmacologic inhibition of HER2 signaling causes the depletion of membrane protrusions and disruption of the interactions between HER2 and HSP90. This is associated with the ubiquitination of HER2, its internalization with EGFR or HER3, and its degradation. These results suggest a model by which some threshold of HER2 signaling is required for the formation and/or maintenance of multi-protein signaling complexes that reinforce and prolong HER2/EGFR or HER2/HER3 signaling by inhibiting HER2 ubiquitination and internalization.
Collapse
Affiliation(s)
- Jaekwang Jeong
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Wonnam Kim
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Lark Kyun Kim
- Severance Biomedical Science Institute and BK21 PLUS project to Medical Science, Severance Institute for Vascular and Metabolic Research, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joshua VanHouten
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - John J. Wysolmerski
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
33
|
Kim JM, Cha SH, Choi YR, Jou I, Joe EH, Park SM. DJ-1 deficiency impairs glutamate uptake into astrocytes via the regulation of flotillin-1 and caveolin-1 expression. Sci Rep 2016; 6:28823. [PMID: 27346864 PMCID: PMC4922019 DOI: 10.1038/srep28823] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 06/10/2016] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is a common chronic and progressive neurodegenerative disorder. Although the cause of PD is still poorly understood, mutations in many genes including SNCA, parkin, PINK1, LRRK2, and DJ-1 have been identified in the familial forms of PD. It was recently proposed that alterations in lipid rafts may cause the neurodegeneration shown in PD. Here, we observe that DJ-1 deficiency decreased the expression of flotillin-1 (flot-1) and caveolin-1 (cav-1), the main protein components of lipid rafts, in primary astrocytes and MEF cells. As a mechanism, DJ-1 regulated flot-1 stability by direct interaction, however, decreased cav-1 expression may not be a direct effect of DJ-1, but rather as a result of decreased flot-1 expression. Dysregulation of flot-1 and cav-1 by DJ-1 deficiency caused an alteration in the cellular cholesterol level, membrane fluidity, and alteration in lipid rafts-dependent endocytosis. Moreover, DJ-1 deficiency impaired glutamate uptake into astrocytes, a major function of astrocytes in the maintenance of CNS homeostasis, by altering EAAT2 expression. This study will be helpful to understand the role of DJ-1 in the pathogenesis of PD, and the modulation of lipid rafts through the regulation of flot-1 or cav-1 may be a novel therapeutic target for PD.
Collapse
Affiliation(s)
- Jin-Mo Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Seon-Heui Cha
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Yu Ree Choi
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Ilo Jou
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Eun-Hye Joe
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon, Korea.,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
34
|
Tao Y, Messer JS, Goss KH, Hart J, Bissonnette M, Chang EB. Hsp70 exerts oncogenic activity in the Apc mutant Min mouse model. Carcinogenesis 2016; 37:731-739. [PMID: 27207671 DOI: 10.1093/carcin/bgw056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 04/29/2016] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) develops from colonic epithelial cells that lose expression of key tumor suppressor genes and/or gain expression of proproliferative and antiapoptotic genes like heat shock protein 70 (Hsp70). Heat shock protein 70 is overexpressed in CRC, but it is not known whether this is in response to the proteotoxic stress induced by transformation, or if it contributes to the process of transformation itself. Here, using the Apc (Min/+) mouse model of CRC, we show that Hsp70 regulates mitogenic signaling in intestinal epithelial cells through stabilization of proteins involved in the receptor tyrosine kinase (RTK) and WNT signaling pathways. Loss of Hsp70 reduced tumor size with decreased proliferation and increased tumor cell death. Hsp70 loss also led to decreased expression of ErbB2, Akt, ERK and β-catenin along with decreased β-catenin transcriptional activity as measured by c-myc and axin2 expression. Upregulation of RTK or WNT signals are frequent oncogenic events in CRC and many other cancers. Thus, in addition to the role of Hsp70 in cell-survival after transformation, Hsp70 stabilization of β-catenin, Akt, ERK and ErbB2 are predicted to contribute to transformation. This has important implications not only for understanding the pathophysiology of these cancers, but also for treatment since anti-EGFR antibodies are in clinical use for CRC and EGFR is a major ErbB2 heterodimeric partner. Targeting Hsp70, therefore, might provide an alternative or complementary strategy for achieving better outcomes for CRC and other related cancer types.
Collapse
Affiliation(s)
| | | | | | - John Hart
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
35
|
Flotillin-2 promotes nasopharyngeal carcinoma metastasis and is necessary for the epithelial-mesenchymal transition induced by transforming growth factor-β. Oncotarget 2016; 6:9781-93. [PMID: 25909165 PMCID: PMC4496397 DOI: 10.18632/oncotarget.3382] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 02/14/2015] [Indexed: 12/02/2022] Open
Abstract
Transforming growth factor-β (TGF-β) promotes cancer metastasis via the epithelial-mesenchymal transition (EMT) but the underlying mechanisms in nasopharyngeal carcinoma (NPC) remain unclear. Flotillin-2 (Flot2), a specialized lipid raft domain in cellular membrane, was reported to promote cancer metastasis. Recently, in neuropathy, it was also suggested that Flot2 was involved in Src activation, which is known as the downstream signal of TGF-β. Therefore, we intended to find out the relationship between Flot2 and TGF-β in the process of nasopharyngeal carcinoma (NPC) metastasis. In this study, we found that Flot2 expression level positively correlated with the cancer stage in NPC tissues. Elevated Flot2 in tumor tissue was an independent prognostic marker, and higher Flot2 expression level showed shorter overall survival time in 181 NPC patients. In NPC cells, silencing Flot2 reversed the metastatic effect induced by TGF-β. Moreover, TGF-β-induced Src phosphorylation was significantly inhibited by Flot2 knocking down. As the consequence of Flot2 inhibition, the expression of the epithelial biomarker E-cadherin was upregulated, while the mesenchymal marker vimentin and signaling transducer β-catenin was suppressed. In conclusions, Flot2 is an indispensable member for TGF-β signaling, which is essential for the EMT process in NPC metastasis. Suppressing Flot2 may be a novel way against TGF-β-induced EMT.
Collapse
|
36
|
Luo JY, Fu ZY, Maimaiti A, Zhou Y, Yang YN, Yu ZX, Chen BD, Liu F, Ma YT. Flotillin-2 Gene Is Associated with Coronary Artery Disease in Chinese Han Population. Genet Test Mol Biomarkers 2015; 19:679-83. [PMID: 26556629 DOI: 10.1089/gtmb.2015.0121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Flotillin-2, an important protein of vesicular endocytosis, is commonly used as a marker protein for lipid microdomains. It plays an essential role in cellular cholesterol uptake and biliary cholesterol reabsorption. Excessive cholesterol intake could cause dyslipidemia, which is a major risk factor of coronary artery disease (CAD). AIMS To investigate the association between the human flotillin-2 gene polymorphism and CAD in the Chinese Han population. MATERIALS AND METHODS Three single-nucleotide polymorphisms (SNPs; rs10205, rs3816848 and rs8081659) of the flotillin-2 gene were genotyped by real-time polymerase chain reaction in 307 CAD patients and 441 control subjects. RESULTS The genotypic distribution of these three SNPs was significantly different between CAD patients and control subjects (all p < 0.05). There were significant differences in the plasma levels of total cholesterol (TC) among different genotypes in the CAD group and control group. For rs3816848, CAD patients with the GG genotype had a higher level of TC than those with an AG or AA genotype (p < 0.001). For rs8081659, CAD patients with TT genotype had a higher level of TC than those with a CT or CC genotype (p < 0.001). Multiple logistic regression analysis showed that the GG genotype of rs3816848 was an independent risk factor for CAD (odds ratio [OR] = 1.786; 95% CI = 1.099-2.902; p = 0.019). CONCLUSION There was a strong association between polymorphisms of flotillin-2 gene and CAD in the Chinese Han population. Persons with the GG genotype of rs3816848 may have a higher risk of CAD. Moreover, the plasma levels of TC were significantly different among the different genotypes of the rs3816848 and rs8081659 SNPs in the CAD group as well as the control group.
Collapse
Affiliation(s)
- Jun-Yi Luo
- 1 Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University , Urumqi, China
| | - Zhen-Yan Fu
- 1 Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University , Urumqi, China
| | - Ailifeire Maimaiti
- 1 Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University , Urumqi, China
| | - Yun Zhou
- 1 Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University , Urumqi, China
| | - Yi-Ning Yang
- 1 Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University , Urumqi, China
| | - Zi-Xiang Yu
- 1 Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University , Urumqi, China
| | - Bang-Dang Chen
- 2 Key Laboratory of Cardiovascular Disease Research of Xinjiang , Urumqi, China
| | - Fen Liu
- 2 Key Laboratory of Cardiovascular Disease Research of Xinjiang , Urumqi, China
| | - Yi-Tong Ma
- 1 Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University , Urumqi, China
| |
Collapse
|
37
|
Jang D, Kwon H, Jeong K, Lee J, Pak Y. Essential role of flotillin-1 palmitoylation in the intracellular localization and signaling function of IGF-1 receptor. J Cell Sci 2015; 128:2179-2190. [PMID: 25908865 DOI: 10.1242/jcs.169409] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/14/2015] [Indexed: 12/21/2022] Open
Abstract
Here, we explored flotillin-1-mediated regulation of insulin-like growth factor-1 (IGF-1) signaling. Flotillin-1-deficient cells exhibited a reduction in the activation of IGF-1 receptor (IGF-1R), ERK1/2 and Akt pathways, and the transcriptional activation of Elk-1 and the proliferation in response to IGF-1 were reduced in these cells. We found that IGF-1-independent flotillin-1 palmitoylation at Cys34 in the endoplasmic reticulum (ER) was required for the ER exit and the plasma membrane localization of flotillin-1 and IGF-1R. IGF-1-dependent depalmitoylation and repalmitoylation of flotillin-1 sustained tyrosine kinase activation of the plasma-membrane-targeted IGF-1R. Dysfunction and blocking the turnover of flotillin-1 palmitoylation abrogated cancer cell proliferation after IGF-1R signaling activation. Our data show that flotillin-1 palmitoylation is a new mechanism by which the intracellular localization and activation of IGF-1R are controlled.
Collapse
Affiliation(s)
- Donghwan Jang
- Division of Life Science, Graduate School of Applied Life Science (BK21 Plus Program), PMBBRC, Gyeongsang National University, Jinju 660-701, Korea
| | - Hayeong Kwon
- Division of Life Science, Graduate School of Applied Life Science (BK21 Plus Program), PMBBRC, Gyeongsang National University, Jinju 660-701, Korea
| | - Kyuho Jeong
- Division of Life Science, Graduate School of Applied Life Science (BK21 Plus Program), PMBBRC, Gyeongsang National University, Jinju 660-701, Korea
| | - Jaewoong Lee
- Division of Life Science, Graduate School of Applied Life Science (BK21 Plus Program), PMBBRC, Gyeongsang National University, Jinju 660-701, Korea
| | - Yunbae Pak
- Division of Life Science, Graduate School of Applied Life Science (BK21 Plus Program), PMBBRC, Gyeongsang National University, Jinju 660-701, Korea
| |
Collapse
|
38
|
Bodin S, Planchon D, Rios Morris E, Comunale F, Gauthier-Rouvière C. Flotillins in intercellular adhesion - from cellular physiology to human diseases. J Cell Sci 2014; 127:5139-47. [PMID: 25413346 DOI: 10.1242/jcs.159764] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Flotillin 1 and 2 are ubiquitous and highly conserved proteins. They were initially discovered in 1997 as being associated with specific caveolin-independent cholesterol- and glycosphingolipid-enriched membrane microdomains and as being expressed during axon regeneration. Flotillins have a role in a large number of physiopathological processes, mainly through their function in membrane receptor clustering and in the regulation of clathrin-independent endocytosis. In this Commentary, we summarize the research performed so far on the role of flotillins in cell-cell adhesion. Recent studies have demonstrated that flotillins directly regulate the formation of cadherin complexes. Indeed, flotillin microdomains are required for the dynamic association and stabilization of cadherins at cell-cell junctions and also for cadherin signaling. Moreover, because flotillins regulate endocytosis and also the actin cytoskeleton, they could have an indirect role in the assembly and stabilization of cadherin complexes. Because it has also recently been shown that flotillins are overexpressed during neurodegenerative diseases and in human cancers, where their upregulation is associated with metastasis formation and poor prognosis, understanding to what extent flotillin upregulation participates in the development of such pathologies is thus of particular interest, as well as how, at the molecular level, it might affect cell adhesion processes.
Collapse
Affiliation(s)
- Stéphane Bodin
- Equipe Labellisée Ligue Contre le Cancer, Universités Montpellier 2 et 1, CRBM, CNRS, UMR 5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Damien Planchon
- Equipe Labellisée Ligue Contre le Cancer, Universités Montpellier 2 et 1, CRBM, CNRS, UMR 5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Eduardo Rios Morris
- Equipe Labellisée Ligue Contre le Cancer, Universités Montpellier 2 et 1, CRBM, CNRS, UMR 5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Franck Comunale
- Equipe Labellisée Ligue Contre le Cancer, Universités Montpellier 2 et 1, CRBM, CNRS, UMR 5237, 1919 Route de Mende, 34293 Montpellier, France
| | - Cécile Gauthier-Rouvière
- Equipe Labellisée Ligue Contre le Cancer, Universités Montpellier 2 et 1, CRBM, CNRS, UMR 5237, 1919 Route de Mende, 34293 Montpellier, France
| |
Collapse
|
39
|
Abstract
ErbB2 (v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2), a receptor tyrosine kinase of the ErbB family, is overexpressed in around 25% of breast cancers. In addition to forming a heterodimer with other ErbB receptors in response to ligand stimulation, ErbB2 can be activated in a ligand-independent manner. We report here that Erbin, an ErbB2-interacting protein that was thought to act as an antitumor factor, is specifically expressed in mammary luminal epithelial cells and facilitates ErbB2-dependent proliferation of breast cancer cells and tumorigenesis in MMTV-neu transgenic mice. Disruption of their interaction decreases ErbB2-dependent proliferation, and deletion of the PDZ domain in Erbin hinders ErbB2-dependent tumor development in MMTV-neu mice. Mechanistically, Erbin forms a complex with ErbB2, promotes its interaction with the chaperon protein HSP90, and thus prevents its degradation. Finally, ErbB2 and Erbin expression correlates in human breast tumor tissues. Together, these observations establish Erbin as an ErbB2 regulator for breast tumor formation and progression.
Collapse
|
40
|
Bertelsen V, Stang E. The Mysterious Ways of ErbB2/HER2 Trafficking. MEMBRANES 2014; 4:424-46. [PMID: 25102001 PMCID: PMC4194043 DOI: 10.3390/membranes4030424] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/01/2014] [Accepted: 07/22/2014] [Indexed: 12/14/2022]
Abstract
The EGFR- or ErbB-family of receptor tyrosine kinases consists of EGFR/ErbB1, ErbB2/HER2, ErbB3/HER3 and ErbB4/HER4. Receptor activation and downstream signaling are generally initiated upon ligand-induced receptor homo- or heterodimerization at the plasma membrane, and endocytosis and intracellular membrane transport are crucial for regulation of the signaling outcome. Among the receptors, ErbB2 is special in several ways. Unlike the others, ErbB2 has no known ligand, but is still the favored dimerization partner. Furthermore, while the other receptors are down-regulated either constitutively or upon ligand-binding, ErbB2 is resistant to down-regulation, and also inhibits down-regulation of its partner upon heterodimerization. The reason(s) why ErbB2 is resistant to down-regulation are the subject of debate. Contrary to other ErbB-proteins, mature ErbB2 needs Hsp90 as chaperone. Several data suggest that Hsp90 is an important regulator of factors like ErbB2 stability, dimerization and/or signaling. Hsp90 inhibitors induce degradation of ErbB2, but whether Hsp90 directly makes ErbB2 endocytosis resistant is unclear. Exposure to anti-ErbB2 antibodies can also induce down-regulation of ErbB2. Down-regulation induced by Hsp90 inhibitors or antibodies does at least partly involve internalization and endosomal sorting to lysosomes for degradation, but also retrograde trafficking to the nucleus has been reported. In this review, we will discuss different molecular mechanisms suggested to be important for making ErbB2 resistant to down-regulation, and review how membrane trafficking is involved when down-regulation and/or relocalization of ErbB2 is induced.
Collapse
Affiliation(s)
- Vibeke Bertelsen
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Post Box 4950 Nydalen, 0424 Oslo, Norway.
| | - Espen Stang
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Post Box 4950 Nydalen, 0424 Oslo, Norway.
| |
Collapse
|
41
|
Zhang Y, Li J, Song Y, Chen F, Pei Y, Yao F. Flotillin-1 expression in human clear-cell renal cell carcinoma is associated with cancer progression and poor patient survival. Mol Med Rep 2014; 10:860-6. [PMID: 24913320 DOI: 10.3892/mmr.2014.2310] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 05/19/2014] [Indexed: 11/06/2022] Open
Abstract
The present study was designed to elucidate the expression levels and the proliferative effect of flotillin-1, an integral membrane protein encoded by the FLOT1 gene, in human clear-cell renal cell carcinoma (RCC). Flotillin has been implicated in other types of cancer, but the role of flotillin in RCC has not been established. Immunohistochemistry and western blotting were used to determine FLOT1 protein expression levels in RCC samples from 182 patients who underwent nephrectomy. FLOT1 mRNA expression levels were analyzed using reverse-transcription (RT) and RT-quantitative polymerase chain reaction (PCR). The association between FLOT1 expression levels in the tumor samples and patient survival time was examined using Kaplan‑Meier analysis. To demonstrate the proliferative effect of FLOT1 on RCC cells, a FLOT1 vector was transfected into four RCC cell lines and FLOT1 expression was inhibited using small interfering RNA. The proliferative ability of the RCC cells was investigated using a WST-1 assay and xenograft experiments with BALB/C nude mice. The results demonstrated that FLOT1 expression levels were significantly higher in RCC cell samples from patients than in healthy renal tissue, and the expression levels were associated with tumor stage, size and histological grade. In addition, FLOT1 significantly enhanced the proliferation of RCC cell lines in vitro and in vivo. These findings suggest that FLOT1, which is upregulated in RCC, is involved in RCC cell proliferation, tumorigenesis and progression. Therefore, FLOT1 is an independent prognostic marker and therapeutic target for patients with clear-cell RCC.
Collapse
Affiliation(s)
- Yiyan Zhang
- Department of Nephrology, First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Jijun Li
- Department of Nephrology, First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Yan Song
- Department of Nephrology, First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Fengkun Chen
- Department of Nephrology, First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Yin Pei
- Department of Nephrology, First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| | - Fenghua Yao
- Department of Nephrology, First Affiliated Hospital of Chinese PLA General Hospital, Beijing 100048, P.R. China
| |
Collapse
|
42
|
Guan Y, Song H, Zhang G, Ai X. Overexpression of flotillin-1 is involved in proliferation and recurrence of bladder transitional cell carcinoma. Oncol Rep 2014; 32:748-54. [PMID: 24890092 DOI: 10.3892/or.2014.3221] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/02/2014] [Indexed: 11/06/2022] Open
Abstract
Flotillin-1 (FLOT1) is known to have a role in tumorigenesis; however, the effect of FLOT1 on proliferation and recurrence of human transitional cell carcinoma (TCC) is unclear. Samples from 156 TCC patients and 142 patients undergoing open bladder surgery for indications other than TCC were used in the present study. FLOT1 protein expression was determined by immunohistochemistry and western blot analysis, and mRNA expression was detected by RT-PCR and real-time PCR. A FLOT1-expressing pcDEF3 vector was stably transfected into 4 TCC cell lines and FLOT1 expression was decreased by RNAi. Proliferative analysis of TCC cells was detected by the WST-1 assay and a xenograft model using BALB/C nude mice. The association between FLOT1 expression and TCC recurrence was also analyzed by adhesion, migration and invasion assays. FLOT1 expression in TCC was significantly overexpressed compared to normal urothelial tissue, and the level of FLOT1 expression was significantly correlated with tumor size, pathologic grade, clinical stage and recurrence. In addition, FLOT1 significantly increased the proliferative ability of TCC cells in vitro and in vivo. TCC cells with a high level of FLOT1 expression exhibited a higher level of adhesion, migration and invasion. FLOT1 expression was shown to be upregulated in human TCC. These findings suggest that FLOT1 plays an important role in the proliferation and recurrence of TCC and that silencing FLOT1 expression might be a novel therapeutic strategy.
Collapse
Affiliation(s)
- Yawei Guan
- Department of Urinary Surgery, Beijing Military Region General Hospital, Beijing 100700, P.R. China
| | - Haiyan Song
- Department of Blood Transfusion, The 309th Hospital of Chinese PLA, Beijing 100091, P.R. China
| | - Guohui Zhang
- Department of Urinary Surgery, Beijing Military Region General Hospital, Beijing 100700, P.R. China
| | - Xing Ai
- Department of Urinary Surgery, Beijing Military Region General Hospital, Beijing 100700, P.R. China
| |
Collapse
|
43
|
Tomiyama A, Uekita T, Kamata R, Sasaki K, Takita J, Ohira M, Nakagawara A, Kitanaka C, Mori K, Yamaguchi H, Sakai R. Flotillin-1 regulates oncogenic signaling in neuroblastoma cells by regulating ALK membrane association. Cancer Res 2014; 74:3790-801. [PMID: 24830726 DOI: 10.1158/0008-5472.can-14-0241] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neuroblastomas harbor mutations in the nonreceptor anaplastic lymphoma kinase (ALK) in 8% to 9% of cases where they serve as oncogenic drivers. Strategies to reduce ALK activity offer clinical interest based on initial findings with ALK kinase inhibitors. In this study, we characterized phosphotyrosine-containing proteins associated with ALK to gain mechanistic insights in this setting. Flotillin-1 (FLOT1), a plasma membrane protein involved in endocytosis, was identified as a binding partner of ALK. RNAi-mediated attenuation of FLOT1 expression in neuroblastoma cells caused ALK dissociation from endosomes along with membrane accumulation of ALK, thereby triggering activation of ALK and downstream effector signals. These features enhanced the malignant properties of neuroblastoma cells in vitro and in vivo. Conversely, oncogenic ALK mutants showed less binding affinity to FLOT1 than wild-type ALK. Clinically, lower expression levels of FLOT1 were documented in highly malignant subgroups of human neuroblastoma specimens. Taken together, our findings suggest that attenuation of FLOT1-ALK binding drives malignant phenotypes of neuroblastoma by activating ALK signaling.
Collapse
Affiliation(s)
- Arata Tomiyama
- Authors' Affiliations: Division of Metastasis and Invasion Signaling, National Cancer Center Research Institute; Department of Neurosurgery, National Defense Medical College, Saitama
| | - Takamasa Uekita
- Authors' Affiliations: Division of Metastasis and Invasion Signaling, National Cancer Center Research Institute; Department of Applied Chemistry, National Defense Academy, Kanagawa
| | - Reiko Kamata
- Authors' Affiliations: Division of Metastasis and Invasion Signaling, National Cancer Center Research Institute
| | - Kazuki Sasaki
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center Research Institute, Osaka
| | - Junko Takita
- Department of Cell Therapy and Transplantation Medicine, Graduate School of medicine, The University of Tokyo, Tokyo
| | | | - Akira Nakagawara
- Biochemistry and Innovative Cancer, Chiba Cancer Center Research Institute, Chiba; and
| | - Chifumi Kitanaka
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata, Japan
| | - Kentaro Mori
- Department of Neurosurgery, National Defense Medical College, Saitama
| | - Hideki Yamaguchi
- Authors' Affiliations: Division of Metastasis and Invasion Signaling, National Cancer Center Research Institute
| | - Ryuichi Sakai
- Authors' Affiliations: Division of Metastasis and Invasion Signaling, National Cancer Center Research Institute;
| |
Collapse
|
44
|
Asp N, Pust S, Sandvig K. Flotillin depletion affects ErbB protein levels in different human breast cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1987-96. [PMID: 24747692 DOI: 10.1016/j.bbamcr.2014.04.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 04/08/2014] [Accepted: 04/10/2014] [Indexed: 10/25/2022]
Abstract
The ErbB3 receptor is an important regulator of cell growth and carcinogenesis. Among breast cancer patients, up to 50-70% have ErbB3 overexpression and 20-30% show overexpressed or amplified ErbB2. ErbB3 has also been implicated in the development of resistance to several drugs used against cancers driven by ErbB1 or ErbB2. One of the main challenges in ErbB-targeting therapy is to inactivate signaling mediated by ErbB2-ErbB3 oncogenic receptor complexes. We analyzed the regulatory role of flotillins on ErbB3 levels and ErbB2-ErbB3 complexes in SKBR3, MCF7 and MDA-MB-134-VI human breast cancer cells. Recently, we described a mechanism for interfering with ErbB2 signaling in breast cancer and demonstrated a molecular complex of flotillin scaffolding proteins with ErbB2 and Hsp90. In the present study, flotillins were found to be in a molecular complex with ErbB3, even in cells without the presence of ErbB2 or other ErbB receptors. Depletion of either flotillin-1 or flotillin-2 resulted in downregulation of ErbB3 and a selective reduction of ErbB2-ErbB3 receptor complexes. Moreover, flotillin-2 depletion resulted in reduced activation of Akt and MAPK signaling cascades, and as a functional consequence of flotillin depletion, breast cancer cells showed an impaired cell migration. Altogether, we provide data demonstrating a novel and functional role of flotillins in the regulation of ErbB protein levels and stabilization of ErbB2-ErbB3 receptor complexes. Thus, flotillins are crucial regulators for oncogenic ErbB function and potential targets for cancer treatment.
Collapse
Affiliation(s)
- Nagham Asp
- Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Sascha Pust
- Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway.
| | - Kirsten Sandvig
- Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway; Department of Molecular Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
45
|
Phuyal S, Hessvik NP, Skotland T, Sandvig K, Llorente A. Regulation of exosome release by glycosphingolipids and flotillins. FEBS J 2014; 281:2214-27. [PMID: 24605801 DOI: 10.1111/febs.12775] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 02/06/2014] [Accepted: 03/04/2014] [Indexed: 12/14/2022]
Abstract
Exosomes are released by cells after fusion of multivesicular bodies with the plasma membrane. The molecular mechanism of this process is still unclear. We investigated the role of sphingolipids and flotillins, which constitute a raft-associated family of proteins, in the release of exosomes. Interestingly, our results show that dl-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol, an inhibitor of glucosylceramide synthase, seemed to affect the composition of exosomes released from PC-3 cells. However, the inhibition of ceramide formation from the de novo pathway by fumonisin B1 did not affect exosome secretion. Moreover, in contrast to findings obtained with other cell lines published so far, inhibition of neutral sphingomyelinase 2, an enzyme that catalyzes the formation of ceramide from sphingomyelin, did not inhibit the secretion of exosomes in PC-3 cells. Finally, small interfering RNA-mediated downregulation of flotillin-1 and flotillin-2 did not significantly change the levels of released exosomes as such, but seemed to affect the composition of exosomes. In conclusion, our results reveal the involvement of glycosphingolipids and flotillins in the release of exosomes from PC-3 cells, and indicate that the role of ceramide in exosome formation may be cell-dependent.
Collapse
Affiliation(s)
- Santosh Phuyal
- Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Norway; Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Norway
| | | | | | | | | |
Collapse
|
46
|
Flotillins in receptor tyrosine kinase signaling and cancer. Cells 2014; 3:129-49. [PMID: 24709906 PMCID: PMC3980747 DOI: 10.3390/cells3010129] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 02/11/2014] [Accepted: 02/12/2014] [Indexed: 01/23/2023] Open
Abstract
Flotillins are highly conserved proteins that localize into specific cholesterol rich microdomains in cellular membranes. They have been shown to be associated with, for example, various signaling pathways, cell adhesion, membrane trafficking and axonal growth. Recent findings have revealed that flotillins are frequently overexpressed in various types of human cancers. We here review the suggested functions of flotillins during receptor tyrosine kinase signaling and in cancer. Although flotillins have been implicated as putative cancer therapy targets, we here show that great caution is required since flotillin ablation may result in effects that increase instead of decrease the activity of specific signaling pathways. On the other hand, as flotillin overexpression appears to be related with metastasis formation in certain cancers, we also discuss the implications of these findings for future therapy aspects.
Collapse
|
47
|
Lee TH, McKleroy W, Khalifeh-Soltani A, Sakuma S, Lazarev S, Riento K, Nishimura SL, Nichols BJ, Atabai K. Functional genomic screen identifies novel mediators of collagen uptake. Mol Biol Cell 2014; 25:583-93. [PMID: 24403604 PMCID: PMC3937085 DOI: 10.1091/mbc.e13-07-0382] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Tissue fibrosis occurs when matrix production outpaces matrix degradation. Degradation of collagen, the main component of fibrotic tissue, is mediated through an extracellular proteolytic pathway and intracellular pathway of cellular uptake and lysosomal digestion. Recent studies demonstrate that disruption of the intracellular pathways can exacerbate fibrosis. These pathways are poorly characterized. Here we identify novel mediators of the intracellular pathway of collagen turnover through a genome-wide RNA interference screen in Drosophila S2 cells. Screening of 7505 Drosophila genes conserved among metazoans identified 22 genes that were required for efficient internalization of type I collagen. These included proteins involved in vesicle transport, the actin cytoskeleton, and signal transduction. We show further that the flotillin genes have a conserved and central role in collagen uptake in Drosophila and human cells. Short hairpin RNA-mediated silencing of flotillins in human monocyte and fibroblasts impaired collagen uptake by promoting lysosomal degradation of the endocytic collagen receptors uPARAP/Endo180 and mannose receptor. These data provide an initial characterization of intracellular pathways of collagen turnover and identify the flotillin genes as critical regulators of this process. A better understanding of these pathways may lead to novel therapies that reduce fibrosis by increasing collagen turnover.
Collapse
Affiliation(s)
- Ting-Hein Lee
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158 Department of Medicine, University of California, San Francisco, San Francisco, CA 94158 Lung Biology Center, University of California, San Francisco, San Francisco, CA 94158 Department of Pathology, University of California, San Francisco, San Francisco, CA 94158 MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kurrle N, Völlner F, Eming R, Hertl M, Banning A, Tikkanen R. Flotillins directly interact with γ-catenin and regulate epithelial cell-cell adhesion. PLoS One 2013; 8:e84393. [PMID: 24391950 PMCID: PMC3877284 DOI: 10.1371/journal.pone.0084393] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/14/2013] [Indexed: 12/23/2022] Open
Abstract
Flotillin-1 and flotillin-2 are two homologous, membrane raft associated proteins. Although it has been reported that flotillins are involved in cell adhesion processes and play a role during breast cancer progression, thus making them interesting future therapeutic targets, their precise function has not been well elucidated. The present study investigates the function of these proteins in cell-cell adhesion in non-malignant cells. We have used the non-malignant epithelial MCF10A cells to study the interaction network of flotillins within cell-cell adhesion complexes. RNA interference was used to examine the effect of flotillins on the structure of adherens junctions and on the association of core proteins, such as E-cadherin, with membrane rafts. We here show that the cadherin proteins of the adherens junction associate with flotillin-2 in MCF10A cells and in various human cell lines. In vitro, flotillin-1 and flotillin-2 directly interact with γ-catenin which is so far the only protein known to be present both in the adherens junction and the desmosome. Mapping of the interaction domain within the γ-catenin sequence identified the Armadillo domains 6-8, especially ARM domain 7, to be important for the association with flotillins. Furthermore, depletion of flotillins significantly influenced the morphology of the adherens junction in human epithelial MCF10A cells and altered the association of E-cadherin and γ-catenin with membrane rafts. Taken together, these observations suggest a functional role for flotillins, especially flotillin-2, in cell-cell adhesion in non-malignant epithelial cells.
Collapse
Affiliation(s)
- Nina Kurrle
- Institute of Biochemistry, Medical Faculty, Justus Liebig University, Giessen, Germany
| | - Frauke Völlner
- Institute of Biochemistry, Medical Faculty, Justus Liebig University, Giessen, Germany
| | - Rüdiger Eming
- Department of Dermatology and Allergology, Phillips University, Marburg, Germany
| | - Michael Hertl
- Department of Dermatology and Allergology, Phillips University, Marburg, Germany
| | - Antje Banning
- Institute of Biochemistry, Medical Faculty, Justus Liebig University, Giessen, Germany
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, Justus Liebig University, Giessen, Germany
- * E-mail:
| |
Collapse
|
49
|
Kurrle N, Ockenga W, Meister M, Völlner F, Kühne S, John BA, Banning A, Tikkanen R. Phosphatidylinositol 3-Kinase dependent upregulation of the epidermal growth factor receptor upon Flotillin-1 depletion in breast cancer cells. BMC Cancer 2013; 13:575. [PMID: 24304721 PMCID: PMC4235219 DOI: 10.1186/1471-2407-13-575] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 11/29/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Flotillin-1 and flotillin-2 are two homologous and ubiquitously expressed proteins that are involved in signal transduction and membrane trafficking. Recent studies have reported that flotillins promote breast cancer progression, thus making them interesting targets for breast cancer treatment. In the present study, we have investigated the underlying molecular mechanisms of flotillins in breast cancer. METHODS Human adenocarcinoma MCF7 breast cancer cells were stably depleted of flotillins by means of lentivirus mediated short hairpin RNAs. Western blotting, immunofluorescence and quantitative real-time PCR were used to analyze the expression of proteins of the epidermal growth factor receptor (EGFR) family. Western blotting was used to investigate the effect of EGFR stimulation or inhibition as well as phosphatidylinositol 3-kinase (PI3K) inhibition on mitogen activated protein kinase (MAPK) signaling. Rescue experiments were performed by stable transfection of RNA intereference resistant flotillin proteins. RESULTS We here show that stable knockdown of flotillin-1 in MCF7 cells resulted in upregulation of EGFR mRNA and protein expression and hyperactivation of MAPK signaling, whereas ErbB2 and ErbB3 expression were not affected. Treatment of the flotillin knockdown cells with an EGFR inhibitor reduced the MAPK signaling, demonstrating that the increased EGFR expression and activity is the cause of the increased signaling. Stable ectopic expression of flotillins in the knockdown cells reduced the increased EGFR expression, demonstrating a direct causal relationship between flotillin-1 expression and EGFR amount. Furthermore, the upregulation of EGFR was dependent on the PI3K signaling pathway which is constitutively active in MCF7 cells, and PI3K inhibition resulted in reduced EGFR expression. CONCLUSIONS This study demonstrates that flotillins may not be suitable as cancer therapy targets in cells that carry certain other oncogenic mutations such as PI3K activating mutations, as unexpected effects are prone to emerge upon flotillin knockdown which may even facilitate cancer cell growth and proliferation.
Collapse
Affiliation(s)
- Nina Kurrle
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Wymke Ockenga
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Melanie Meister
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Frauke Völlner
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Sina Kühne
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Bincy A John
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Antje Banning
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| |
Collapse
|
50
|
Wang X, Yang Q, Guo L, Li XH, Zhao XH, Song LB, Lin HX. Flotillin-2 is associated with breast cancer progression and poor survival outcomes. J Transl Med 2013; 11:190. [PMID: 23945257 PMCID: PMC3765773 DOI: 10.1186/1479-5876-11-190] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/13/2013] [Indexed: 12/16/2022] Open
Abstract
Background Flotillin-2 (FLOT2) has been implicated in several signaling pathways in tumor cells. Our study aimed to investigate the expression pattern and clinicopathological significance of FLOT2 in patients with breast cancer. Methods The expression level of FLOT2 in normal breast epithelial cells, breast cancer cell lines, and four breast cancer biopsies paired with adjacent noncancerous tissues were quantified using real-time RT-PCR and Western blotting. FLOT2 protein expression was analyzed in 171 archived paraffin-embedded breast cancer samples using immunohistochemistry (IHC). Statistical analyses were performed to evaluate the clinicopathological significance of FLOT2 expression. Results FLOT2 was significantly upregulated in breast cancer cell lines and tissue samples compared with normal cells and adjacent noncancerous breast tissues, respectively. IHC analysis revealed high expression levels of FLOT2 in 82 of 171 (48.0%) breast cancer specimens. Statistical analysis revealed that FLOT2 expression was significantly correlated with clinical stage (P < 0.001), T classification (P < 0.001), M classification (P < 0.001), histological differentiation (P = 0.005) and ErbB2 expression (P = 0.003). Patients with higher levels of FLOT2 expression had a shorter overall survival duration than patients with lower FLOT2 expression levels. Multivariate analysis suggested that FLOT2 expression was an independent prognostic marker for survival in patients with breast cancer. Conclusions The current results demonstrated that high FLOT2 protein expression was associated with poor outcomes in patients with breast cancer. FLOT2 could be used as a prognostic biomarker for breast cancer progression.
Collapse
Affiliation(s)
- Xi Wang
- State Key Laboratory of Oncology in South China, Guangzhou 510060, PR China.
| | | | | | | | | | | | | |
Collapse
|