1
|
Venkatramani A, Ashtam A, Panda D. EB1 Increases the Dynamics of Tau Droplets and Inhibits Tau Aggregation: Implications in Tauopathies. ACS Chem Neurosci 2024; 15:1219-1233. [PMID: 38445984 DOI: 10.1021/acschemneuro.3c00815] [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] [Indexed: 03/07/2024] Open
Abstract
EB1, a microtubule plus end-tracking protein (+TIP), regulates microtubule dynamics. Recent evidence indicates cross-talk between EB proteins and tau, a microtubule-associated neuronal protein that is important for the growth and stability of microtubules. We investigated the interaction between tau and EB1 and the effect of binding of EB1 on tau function and aggregation. EB1 colocalized with tau in SH-SY5Y cells and coimmunoprecipitated with tau. Further, purified EB1 impaired the ability of adult tau to induce tubulin polymerization in vitro. EB1 bound to tau with a dissociation constant of 2.5 ± 0.7 μM. EB1 reduced heparin-induced tau aggregation with a half-maximal inhibitory concentration of 4.3 ± 0.2 μM, and increased the dynamics of tau in phase-separated droplets. The fluorescence recovery rate in tau droplets increased from 0.02 ± 0.01 to 0.07 ± 0.03 s-1, while the half-time of recovery decreased from 44.5 ± 14 to 13.5 ± 6 s in the presence of 8 μM EB1, suggesting a delay in the transition of tau from the soluble to aggregated form in tau liquid-liquid phase separation. EB1 decreased the rate of aggregation and increased the critical concentration of tau aggregation. Dynamic light scattering, atomic force microscopy, dot blot assays, and SDS-PAGE analysis showed that EB1 inhibited the formation of oligomers and higher-order aggregates of tau. The data suggest a novel role for EB1 as a regulator of tau function and aggregation, and the findings indicated the role of the EB family proteins in neuronal function and neurodegeneration.
Collapse
Affiliation(s)
- Anuradha Venkatramani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Anvesh Ashtam
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
- National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160062, India
| |
Collapse
|
2
|
Fakhri S, Moradi SZ, Faraji F, Kooshki L, Webber K, Bishayee A. Modulation of hypoxia-inducible factor-1 signaling pathways in cancer angiogenesis, invasion, and metastasis by natural compounds: a comprehensive and critical review. Cancer Metastasis Rev 2024; 43:501-574. [PMID: 37792223 DOI: 10.1007/s10555-023-10136-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023]
Abstract
Tumor cells employ multiple signaling mediators to escape the hypoxic condition and trigger angiogenesis and metastasis. As a critical orchestrate of tumorigenic conditions, hypoxia-inducible factor-1 (HIF-1) is responsible for stimulating several target genes and dysregulated pathways in tumor invasion and migration. Therefore, targeting HIF-1 pathway and cross-talked mediators seems to be a novel strategy in cancer prevention and treatment. In recent decades, tremendous efforts have been made to develop multi-targeted therapies to modulate several dysregulated pathways in cancer angiogenesis, invasion, and metastasis. In this line, natural compounds have shown a bright future in combating angiogenic and metastatic conditions. Among the natural secondary metabolites, we have evaluated the critical potential of phenolic compounds, terpenes/terpenoids, alkaloids, sulfur compounds, marine- and microbe-derived agents in the attenuation of HIF-1, and interconnected pathways in fighting tumor-associated angiogenesis and invasion. This is the first comprehensive review on natural constituents as potential regulators of HIF-1 and interconnected pathways against cancer angiogenesis and metastasis. This review aims to reshape the previous strategies in cancer prevention and treatment.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leila Kooshki
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6714415153, Iran
| | - Kassidy Webber
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL, 34211, USA.
| |
Collapse
|
3
|
Jia W, Gong X, Ye Z, Li N, Zhan X. Nitroproteomics is instrumental for stratification and targeted treatments of astrocytoma patients: expert recommendations for advanced 3PM approach with improved individual outcomes. EPMA J 2023; 14:673-696. [PMID: 38094577 PMCID: PMC10713973 DOI: 10.1007/s13167-023-00348-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/11/2023] [Indexed: 12/05/2024]
Abstract
Protein tyrosine nitration is a selectively and reversible important post-translational modification, which is closely related to oxidative stress. Astrocytoma is the most common neuroepithelial tumor with heterogeneity and complexity. In the past, the diagnosis of astrocytoma was based on the histological and clinical features, and the treatment methods were nothing more than surgery-assisted radiotherapy and chemotherapy. Obviously, traditional methods short falls an effective treatment for astrocytoma. In late 2021, the World Health Organization (WHO) adopted molecular biomarkers in the comprehensive diagnosis of astrocytoma, such as IDH-mutant and DNA methylation, which enabled the risk stratification, classification, and clinical prognosis prediction of astrocytoma to be more correct. Protein tyrosine nitration is closely related to the pathogenesis of astrocytoma. We hypothesize that nitroproteome is significantly different in astrocytoma relative to controls, which leads to establishment of nitroprotein biomarkers for patient stratification, diagnostics, and prediction of disease stages and severity grade, targeted prevention in secondary care, treatment algorithms tailored to individualized patient profile in the framework of predictive, preventive, and personalized medicine (PPPM; 3P medicine). Nitroproteomics based on gel electrophoresis and tandem mass spectrometry is an effective tool to identify the nitroproteins and effective biomarkers in human astrocytomas, clarifying the biological roles of oxidative/nitrative stress in the pathophysiology of astrocytomas, functional characteristics of nitroproteins in astrocytomas, nitration-mediated signal pathway network, and early diagnosis and treatment of astrocytomas. The results finds that these nitroproteins are enriched in mitotic cell components, which are related to transcription regulation, signal transduction, controlling subcellular organelle events, cell perception, maintaining cell homeostasis, and immune activity. Eleven statistically significant signal pathways are identified in astrocytoma, including remodeling of epithelial adherens junctions, germ cell-sertoli cell junction signaling, 14-3-3-mediated signaling, phagosome maturation, gap junction signaling, axonal guidance signaling, assembly of RNA polymerase III complex, and TREM1 signaling. Furthermore, protein tyrosine nitration is closely associated with the therapeutic effects of protein drugs, and molecular mechanism and drug targets of cancer. It provides valuable data for studying the protein nitration biomarkers, molecular mechanisms, and therapeutic targets of astrocytoma towards PPPM (3P medicine) practice. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00348-y.
Collapse
Affiliation(s)
- Wenshuang Jia
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xiaoxia Gong
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Zhen Ye
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
4
|
Makarov D, Kielkowski P. Chemical Proteomics Reveals Protein Tyrosination Extends Beyond the Alpha-Tubulins in Human Cells. Chembiochem 2022; 23:e202200414. [PMID: 36218090 PMCID: PMC10099736 DOI: 10.1002/cbic.202200414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/10/2022] [Indexed: 01/25/2023]
Abstract
Tubulin detyrosination-tyrosination cycle regulates the stability of microtubules. With respect to α-tubulins, the tyrosination level is maintained by a single tubulin-tyrosine ligase (TTL). However, the precise dynamics and tubulin isoforms which undergo (de)tyrosination in neurons are unknown. Here, we exploit the substrate promiscuity of the TTL to introduce an O-propargyl-l-tyrosine to neuroblastoma cells and neurons. Mass spectrometry-based chemical proteomics in neuroblastoma cells using the O-propargyl-l-tyrosine probe revealed previously discussed tyrosination of TUBA4A, MAPRE1, and other non-tubulin proteins. This finding was further corroborated in differentiating neurons. Together we present the method for tubulin tyrosination profiling in living cells. Our results show that detyrosination-tyrosination is not restricted to α-tubulins with coded C-terminal tyrosine and is thus involved in fine-tuning of the tubulin and non-tubulin proteins during neuronal differentiation.
Collapse
Affiliation(s)
- Dmytro Makarov
- LMU München, Department of Chemistry, Institute for Chemical Epigenetics - Munich (ICEM), Würmtalstrasse 201, 81375, Munich, Germany
| | - Pavel Kielkowski
- LMU München, Department of Chemistry, Institute for Chemical Epigenetics - Munich (ICEM), Würmtalstrasse 201, 81375, Munich, Germany
| |
Collapse
|
5
|
Bär J, Popp Y, Bucher M, Mikhaylova M. Direct and indirect effects of tubulin post-translational modifications on microtubule stability: Insights and regulations. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119241. [PMID: 35181405 DOI: 10.1016/j.bbamcr.2022.119241] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/17/2022]
Abstract
Microtubules (MTs) mediate various cellular functions such as structural support, chromosome segregation, and intracellular transport. To achieve this, the pivotal properties of MTs have to be changeable and tightly controlled. This is enabled by a high variety of tubulin posttranslational modifications, which influence MT properties directly, via altering the MT lattice structurally, or indirectly by changing MT interaction partners. Here, the distinction between these direct and indirect effects of MT PTMs are exemplified by acetylation of the luminal α-tubulin K40 resulting in decreased rigidity of MTs, and by MT detyrosination which decreases interaction with depolymerizing proteins, thus causing more stable MTs. We discuss how these PTMs are reversed and regulated, e.g. on the level of enzyme transcription, localization, and activity via various signalling pathways including the conventional calcium-dependent proteases calpains and how advances in microscopy techniques and development of live-sensors facilitate the understanding of MT PTM interaction and effects.
Collapse
Affiliation(s)
- Julia Bär
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany; Guest Group "Neuronal Protein Transport", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| | - Yannes Popp
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany; Guest Group "Neuronal Protein Transport", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| | - Michael Bucher
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany; Guest Group "Neuronal Protein Transport", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| | - Marina Mikhaylova
- RG Optobiology, Institute of Biology, Humboldt Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany; Guest Group "Neuronal Protein Transport", Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| |
Collapse
|
6
|
Perez T, Bergès R, Maccario H, Oddoux S, Honoré S. Low concentrations of vorinostat decrease EB1 expression in GBM cells and affect microtubule dynamics, cell survival and migration. Oncotarget 2021; 12:304-315. [PMID: 33659042 PMCID: PMC7899546 DOI: 10.18632/oncotarget.27892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/01/2021] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiform (GBM) is the most frequent primitive brain tumor with a high recurrence and mortality. Histone deacetylase inhibitors (HDACi) have evoked great interest because they are able to change transcriptomic profiles to promote tumor cell death but also induce side effects due to the lack of selectivity. We show in this paper new anticancer properties and mechanisms of action of low concentrations of vorinostat on various GBM cells which acts by affecting microtubule cytoskeleton in a non-histone 3 (H3) manner. Indeed, vorinostat induces tubulin acetylation and detyrosination, affects EB stabilizing cap on microtubule plus ends and suppresses microtubule dynamic instability. We previously identified EB1 overexpression as a marker of bad prognostic in GBM. Interestingly, we show for the first time to our knowledge, a strong decrease of EB1 expression in GBM cells by a drug. Altogether, our results suggest that low dose vorinostat, which is more selective for HDAC6 inhibition, could therefore represent an interesting therapeutic option for GBM especially in patients with EB1 overexpressing tumor with lower expected side effects. A validation of our hypothesis is needed during future clinical trials with this drug in GBM.
Collapse
Affiliation(s)
- Thomas Perez
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France.,APHM, Hôpital de la Timone, Service Pharmacie, Marseille, France
| | - Raphaël Bergès
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Hélène Maccario
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Sarah Oddoux
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France
| | - Stéphane Honoré
- Aix-Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France.,APHM, Hôpital de la Timone, Service Pharmacie, Marseille, France
| |
Collapse
|
7
|
EB1-dependent long survival of glioblastoma-grafted mice with the oral tubulin-binder BAL101553 is associated with inhibition of tumor angiogenesis. Oncotarget 2020; 11:759-774. [PMID: 32165998 PMCID: PMC7055546 DOI: 10.18632/oncotarget.27374] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/06/2019] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma (GBM) are aggressive brain tumors with limited treatment options. Cancer stem-like cells (CSLCs) contribute to GBM invasiveness, representing promising targets. BAL101553, a prodrug of BAL27862, is a novel small molecule tubulin-binding agent, promoting tumor cell death through spindle assembly checkpoint activation, which is currently in Phase 1/2a in advanced solid tumor patients including GBM. This study aimed to evaluate long-term daily oral BAL101553 treatment of mice orthotopically grafted with GBM CSLCs (GBM6) according to EB1 expression-level, and to decipher its mechanism of action on GBM stem cells. Oral treatment with BAL101553 for 100 days provoked a large EB1 expression level-dependent survival benefit, together with a decrease in tumor growth and brain invasion. Formation of vascular structures by the fluorescent GBM6-GFP-sh0 cells, mimicking endothelial vascular networks, was observed in the brains of control grafted mice. Following BAL101553 treatment, vessels were no longer detectable, suggesting inhibition of the endothelial trans-differentiation of GBM stem cells. In vitro, BAL27862 treatment resulted in a switch to the endothelial-like phenotype of GBM6 towards an astrocytic phenotype. Moreover, the drug inhibited secretion of VEGF, thus preventing normal endothelial cell migration activated by CSLCs. The decrease in VEGF secretion was confirmed in a human GBM explant following drug treatment. Altogether, our data first confirm the potential of EB1 expression as a response-predictive biomarker of BAL101553 in GBM we previously published and add new insights in BAL101553 long-term action by counteracting CSLCs mediated tumor angiogenesis. Our results strongly support BAL101553 clinical studies in GBM patients.
Collapse
|
8
|
Blanco C, Morales D, Mogollones I, Vergara‐Jaque A, Vargas C, Álvarez A, Riquelme D, Leiva‐Salcedo E, González W, Morales D, Maureira D, Aldunate I, Cáceres M, Varela D, Cerda O. EB1‐ and EB2‐dependent anterograde trafficking of TRPM4 regulates focal adhesion turnover and cell invasion. FASEB J 2019; 33:9434-9452. [DOI: 10.1096/fj.201900136r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Constanza Blanco
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Danna Morales
- Program of Physiology and Biophysics Institute of Biomedical Sciences Faculty of Medicine Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Ignacio Mogollones
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Ariela Vergara‐Jaque
- Program of Physiology and Biophysics Institute of Biomedical Sciences Faculty of Medicine Universidad de Chile Santiago Chile
- Multidisciplinary Scientific Nucleus Universidad de Talca Talca Chile
- Center for Bioinformatics and Molecular Simulation Universidad de Talca Talca Chile
| | - Carla Vargas
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Alhejandra Álvarez
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Denise Riquelme
- Department of Biology Faculty of Chemistry and Biology Universidad de Santiago de Chile Santiago Chile
| | - Elías Leiva‐Salcedo
- Department of Biology Faculty of Chemistry and Biology Universidad de Santiago de Chile Santiago Chile
| | - Wendy González
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
- Center for Bioinformatics and Molecular Simulation Universidad de Talca Talca Chile
| | - Diego Morales
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Diego Maureira
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Ismael Aldunate
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
| | - Mónica Cáceres
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
- The Wound Repair Treatment, and Health (WoRTH) Initiative Santiago Chile
| | - Diego Varela
- Program of Physiology and Biophysics Institute of Biomedical Sciences Faculty of Medicine Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
- The Wound Repair Treatment, and Health (WoRTH) Initiative Santiago Chile
| |
Collapse
|
9
|
Berges R, Denicolai E, Tchoghandjian A, Baeza-Kallee N, Honore S, Figarella-Branger D, Braguer D. Proscillaridin A exerts anti-tumor effects through GSK3β activation and alteration of microtubule dynamics in glioblastoma. Cell Death Dis 2018; 9:984. [PMID: 30250248 PMCID: PMC6155148 DOI: 10.1038/s41419-018-1018-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/26/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is characterized by highly aggressive growth and invasive behavior. Due to the highly lethal nature of GBM, new therapies are urgently needed and repositioning of existing drugs is a promising approach. We have previously shown the activity of Proscillaridin A (ProA), a cardiac glycoside inhibitor of the Na(+)/K(+) ATPase (NKA) pump, against proliferation and migration of GBM cell lines. ProA inhibited tumor growth in vivo and increased mice survival after orthotopic grafting of GBM cells. This study aims to decipher the mechanism of action of ProA in GBM tumor and stem-like cells. ProA displayed cytotoxic activity on tumor and stem-like cells grown in 2D and 3D culture, but not on healthy cells as astrocytes or oligodendrocytes. Even at sub-cytotoxic concentration, ProA impaired cell migration and disturbed EB1 accumulation at microtubule (MT) plus-ends and MT dynamics instability. ProA activates GSK3β downstream of NKA inhibition, leading to EB1 phosphorylation on S155 and T166, EB1 comet length shortening and MT dynamics alteration, and finally inhibition of cell migration and cytotoxicity. Similar results were observed with digoxin. Therefore, we disclosed here a novel pathway by which ProA and digoxin modulate MT-governed functions in GBM tumor and stem-like cells. Altogether, our results support ProA and digoxin as potent candidates for drug repositioning in GBM.
Collapse
Affiliation(s)
- Raphael Berges
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Emilie Denicolai
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | | | | | - Stephane Honore
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | | | - Diane Braguer
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.
| |
Collapse
|
10
|
Dentesano YM, Ditamo Y, Hansen C, Arce CA, Bisig CG. Post‐translational incorporation of 3,4‐dihydroxyphenylalanine into the C terminus of α‐tubulin in living cells. FEBS J 2018; 285:1064-1078. [DOI: 10.1111/febs.14386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/07/2017] [Accepted: 01/11/2018] [Indexed: 01/18/2023]
Affiliation(s)
- Yanela M. Dentesano
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC) UNC‐CONICET Departamento de Química Biológica Facultad de Ciencias Químicas Universidad Nacional de Córdoba Argentina
| | - Yanina Ditamo
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC) UNC‐CONICET Departamento de Química Biológica Facultad de Ciencias Químicas Universidad Nacional de Córdoba Argentina
| | | | - Carlos A. Arce
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC) UNC‐CONICET Departamento de Química Biológica Facultad de Ciencias Químicas Universidad Nacional de Córdoba Argentina
| | - Carlos Gaston Bisig
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC) UNC‐CONICET Departamento de Química Biológica Facultad de Ciencias Químicas Universidad Nacional de Córdoba Argentina
| |
Collapse
|
11
|
Aillaud C, Bosc C, Peris L, Bosson A, Heemeryck P, Van Dijk J, Le Friec J, Boulan B, Vossier F, Sanman LE, Syed S, Amara N, Couté Y, Lafanechère L, Denarier E, Delphin C, Pelletier L, Humbert S, Bogyo M, Andrieux A, Rogowski K, Moutin MJ. Vasohibins/SVBP are tubulin carboxypeptidases (TCPs) that regulate neuron differentiation. Science 2017; 358:1448-1453. [PMID: 29146868 DOI: 10.1126/science.aao4165] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/03/2017] [Indexed: 12/28/2022]
Abstract
Reversible detyrosination of α-tubulin is crucial to microtubule dynamics and functions, and defects have been implicated in cancer, brain disorganization, and cardiomyopathies. The identity of the tubulin tyrosine carboxypeptidase (TCP) responsible for detyrosination has remained unclear. We used chemical proteomics with a potent irreversible inhibitor to show that the major brain TCP is a complex of vasohibin-1 (VASH1) with the small vasohibin binding protein (SVBP). VASH1 and its homolog VASH2, when complexed with SVBP, exhibited robust and specific Tyr/Phe carboxypeptidase activity on microtubules. Knockdown of vasohibins or SVBP and/or inhibitor addition in cultured neurons reduced detyrosinated α-tubulin levels and caused severe differentiation defects. Furthermore, knockdown of vasohibins disrupted neuronal migration in developing mouse neocortex. Thus, vasohibin/SVBP complexes represent long-sought TCP enzymes.
Collapse
Affiliation(s)
- Chrystelle Aillaud
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Christophe Bosc
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Leticia Peris
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Anouk Bosson
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Pierre Heemeryck
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Juliette Van Dijk
- Institut de Génétique Humaine (IGH), Université Montpellier, CNRS UMR9002, 34000 Montpellier, France.,Centre de Recherche en Biochimie Macromoléculaire (CRBM), Université Montpellier, CNRS UMR5237, 34000 Montpellier, France
| | - Julien Le Friec
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Benoit Boulan
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Frédérique Vossier
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Laura E Sanman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Salahuddin Syed
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Neri Amara
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yohann Couté
- Institut de Biosciences et Biotechnologies de Grenoble (BIG)-Laboratoire Biologie à Grande Échelle, Université Grenoble Alpes, CEA, INSERM, F-38000 Grenoble, France
| | - Laurence Lafanechère
- Team Regulation and Pharmacology of the Cytoskeleton, Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Eric Denarier
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France.,BIG-Physiopathologie du Cytosquelette, CEA, F-38000 Grenoble, France
| | - Christian Delphin
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Laurent Pelletier
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Sandrine Humbert
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Annie Andrieux
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France. .,Inserm, U1216, F-38000 Grenoble, France.,BIG-Physiopathologie du Cytosquelette, CEA, F-38000 Grenoble, France
| | - Krzysztof Rogowski
- Institut de Génétique Humaine (IGH), Université Montpellier, CNRS UMR9002, 34000 Montpellier, France
| | - Marie-Jo Moutin
- Grenoble Institut des Neurosciences (GIN), Université Grenoble Alpes, F-38000 Grenoble, France.,Inserm, U1216, F-38000 Grenoble, France
| |
Collapse
|
12
|
Liu Y, Visetsouk M, Mynlieff M, Qin H, Lechtreck KF, Yang P. H +- and Na +- elicited rapid changes of the microtubule cytoskeleton in the biflagellated green alga Chlamydomonas. eLife 2017; 6:26002. [PMID: 28875932 PMCID: PMC5779235 DOI: 10.7554/elife.26002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 09/05/2017] [Indexed: 12/27/2022] Open
Abstract
Although microtubules are known for dynamic instability, the dynamicity is considered to be tightly controlled to support a variety of cellular processes. Yet diverse evidence suggests that this is not applicable to Chlamydomonas, a biflagellate fresh water green alga, but intense autofluorescence from photosynthesis pigments has hindered the investigation. By expressing a bright fluorescent reporter protein at the endogenous level, we demonstrate in real time discreet sweeping changes in algal microtubules elicited by rises of intracellular H+ and Na+. These results from this model organism with characteristics of animal and plant cells provide novel explanations regarding how pH may drive cellular processes; how plants may respond to, and perhaps sense stresses; and how organisms with a similar sensitive cytoskeleton may be susceptible to environmental changes.
Collapse
Affiliation(s)
- Yi Liu
- Department of Biological Sciences, Marquette University, Milwaukee, United States
| | - Mike Visetsouk
- Department of Biological Sciences, Marquette University, Milwaukee, United States
| | - Michelle Mynlieff
- Department of Biological Sciences, Marquette University, Milwaukee, United States
| | - Hongmin Qin
- Department of Biology, Texas A&M University, College Station, United States
| | - Karl F Lechtreck
- Department of Cellular Biology, University of Georgia, Athen, United States
| | - Pinfen Yang
- Department of Biological Sciences, Marquette University, Milwaukee, United States
| |
Collapse
|
13
|
White D, Honoré S, Hubert F. Exploring the effect of end-binding proteins and microtubule targeting chemotherapy drugs on microtubule dynamic instability. J Theor Biol 2017. [DOI: 10.1016/j.jtbi.2017.06.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
14
|
Nehlig A, Molina A, Rodrigues-Ferreira S, Honoré S, Nahmias C. Regulation of end-binding protein EB1 in the control of microtubule dynamics. Cell Mol Life Sci 2017; 74:2381-2393. [PMID: 28204846 PMCID: PMC11107513 DOI: 10.1007/s00018-017-2476-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/13/2017] [Accepted: 01/24/2017] [Indexed: 12/14/2022]
Abstract
The regulation of microtubule dynamics is critical to ensure essential cell functions, such as proper segregation of chromosomes during mitosis or cell polarity and migration. End-binding protein 1 (EB1) is a plus-end-tracking protein (+TIP) that accumulates at growing microtubule ends and plays a pivotal role in the regulation of microtubule dynamics. EB1 autonomously binds an extended tubulin-GTP/GDP-Pi structure at growing microtubule ends and acts as a molecular scaffold that recruits a large number of regulatory +TIPs through interaction with CAP-Gly or SxIP motifs. While extensive studies have focused on the structure of EB1-interacting site at microtubule ends and its role as a molecular platform, the mechanisms involved in the negative regulation of EB1 have only started to emerge and remain poorly understood. In this review, we summarize recent studies showing that EB1 association with MT ends is regulated by post-translational modifications and affected by microtubule-targeting agents. We also present recent findings that structural MAPs, that have no tip-tracking activity, physically interact with EB1 to prevent its accumulation at microtubule plus ends. These observations point out a novel concept of "endogenous EB1 antagonists" and emphasize the importance of finely regulating EB1 function at growing microtubule ends.
Collapse
Affiliation(s)
- Anne Nehlig
- Inserm U981, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
- University Paris Saclay, 94800, Villejuif, France
| | - Angie Molina
- Inserm U981, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
- University Paris Saclay, 94800, Villejuif, France
- CBD, University of Toulouse-3, Toulouse, France
| | - Sylvie Rodrigues-Ferreira
- Inserm U981, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
- University Paris Saclay, 94800, Villejuif, France
| | - Stéphane Honoré
- Aix Marseille University, Inserm U-911, CRO2, Marseille, France
- Service Pharmacie, CHU Hôpital de La Timone, APHM, Marseille, France
| | - Clara Nahmias
- Inserm U981, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France.
- University Paris Saclay, 94800, Villejuif, France.
| |
Collapse
|
15
|
Genova C, Alama A, Coco S, Rijavec E, Dal Bello MG, Vanni I, Biello F, Barletta G, Rossi G, Grossi F. Vinflunine for the treatment of non-small cell lung cancer. Expert Opin Investig Drugs 2016; 25:1447-1455. [PMID: 27771969 DOI: 10.1080/13543784.2016.1252331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Vinflunine belongs to the class of vinca alkaloids and acts by disrupting the microtubule dynamics during cell cycle; this agent is currently available for previously treated advanced transitional cell carcinoma in Europe. The aim of this invited review is to evaluate the potential role of vinflunine for the treatment of non-small cell lung cancer (NSCLC). Areas covered: The potential role of vinflunine in NSCLC is discussed on the basis of the available data, including full papers and meeting abstracts. Relevant preclinical studies describing the pharmacological properties of vinflunine are also included. The review also summarizes clinical studies, including phase I trials involving NSCLC among other tumors as well as phase II/III trials specifically addressing this malignancy. Additionally, the safety profile and the current regulatory status of vinflunine is discussed. Expert opinion: Vinflunine is active as single agent and as part of platinum-based combinations in NSCLC. It results non-inferior to docetaxel in a randomized phase III trial including previously treated NSCLC patients; additionally, its safety profile is generally considered manageable. Ultimately, further studies are needed to confirm the role of vinflunine in NSCLC, in consideration of the evolving evidence regarding targeted therapies and immune check-point inhibitors.
Collapse
Affiliation(s)
- C Genova
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| | - A Alama
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| | - S Coco
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| | - E Rijavec
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| | - M G Dal Bello
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| | - I Vanni
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| | - F Biello
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| | - G Barletta
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| | - G Rossi
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| | - F Grossi
- a UOS Tumori Polmonari , IRCCS AOU San Martino IST-Istituto Nazionale per la Ricerca sul Cancro , Genoa , Italy
| |
Collapse
|
16
|
Finkenstaedt-Quinn SA, Qiu TA, Shin K, Haynes CL. Super-resolution imaging for monitoring cytoskeleton dynamics. Analyst 2016; 141:5674-5688. [PMID: 27549146 DOI: 10.1039/c6an00731g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cytoskeleton is a key cellular structure that is important in the control of cellular movement, structure, and sensing. To successfully image the individual cytoskeleton components, high resolution and super-resolution fluorescence imaging methods are needed. This review covers the three basic cytoskeletal elements and the relative benefits and drawbacks of fixed versus live cell imaging before moving on to recent studies using high resolution and super-resolution techniques. The techniques covered include the near-diffraction limited imaging methods of confocal microscopy and TIRF microscopy and the super-resolution fluorescence imaging methods of STORM, PALM, and STED.
Collapse
|
17
|
Bergès R, Tchoghandjian A, Honoré S, Estève MA, Figarella-Branger D, Bachmann F, Lane HA, Braguer D. The Novel Tubulin-Binding Checkpoint Activator BAL101553 Inhibits EB1-Dependent Migration and Invasion and Promotes Differentiation of Glioblastoma Stem-like Cells. Mol Cancer Ther 2016; 15:2740-2749. [PMID: 27540016 DOI: 10.1158/1535-7163.mct-16-0252] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/09/2016] [Indexed: 11/16/2022]
Abstract
Glioblastoma patients have limited treatment options. Cancer stem-like cells (CSLC) contribute to glioblastoma invasiveness and repopulation; hence, they represent promising targets for novel therapies. BAL101553 is a prodrug of BAL27862, a novel microtubule-destabilizing agent inhibiting tumor cell proliferation through activation of the spindle assembly checkpoint, which is currently in phase I/II clinical development. Broad anticancer activity has been demonstrated against human cancer models, including tumors refractory to conventional treatments. We have shown that overexpression of microtubule + end-binding 1-protein (EB1) correlates with glioblastoma progression and poor survival. Here, we show that BAL27862 inhibits the growth of two glioblastoma CSLCs. As EB1 is overexpressed in the CSLC line GBM6, which displays a high tumorigenicity and infiltrative pattern of migration in vivo, we investigated drug activity on GBM6 according to EB1 expression. BAL27862 inhibited migration and colony formation at subcytotoxic concentrations in EB1-expressing control cells (GBM6-sh0) but only at cytotoxic concentrations in EB1-downregulated (GBM-shE1) cells. Three administrations of BAL101553 were sufficient to provoke an EB1-dependent survival benefit in tumor-bearing mice. Patterns of invasion and quantification of tumor cells in brain demonstrated that GBM6-sh0 cells were more invasive than GBM6-shEB1 cells, and that the antiproliferative and anti-invasive effects of BAL101553 were more potent in mice bearing control tumors than in EB1-downregulated tumors. This was associated with inhibition of stem cell properties in the GBM6-sh0 model. Finally, BAL27862 triggered astrocytic differentiation of GBM6 in an EB1-dependent manner. These results support the potential of BAL101553 for glioblastoma treatment, with EB1 expression as a predictive biomarker of response. Mol Cancer Ther; 15(11); 2740-9. ©2016 AACR.
Collapse
Affiliation(s)
- Raphaël Bergès
- Aix Marseille Univ, INSERM, CRO2 UMR911, Marseille, France
| | | | - Stéphane Honoré
- Aix Marseille Univ, INSERM, CRO2 UMR911, Marseille, France.,APHM, CHU Timone, Marseille, France
| | - Marie-Anne Estève
- Aix Marseille Univ, INSERM, CRO2 UMR911, Marseille, France.,APHM, CHU Timone, Marseille, France
| | | | - Felix Bachmann
- Basilea Pharmaceutica International Ltd., Basel, Switzerland
| | - Heidi A Lane
- Basilea Pharmaceutica International Ltd., Basel, Switzerland.
| | - Diane Braguer
- Aix Marseille Univ, INSERM, CRO2 UMR911, Marseille, France. .,APHM, CHU Timone, Marseille, France
| |
Collapse
|
18
|
Peng F, Li J, Guo T, Yang H, Li M, Sang S, Li X, Desiderio DM, Zhan X. Nitroproteins in Human Astrocytomas Discovered by Gel Electrophoresis and Tandem Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:2062-76. [PMID: 26450359 DOI: 10.1007/s13361-015-1270-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/20/2015] [Accepted: 09/01/2015] [Indexed: 05/17/2023]
Abstract
Protein tyrosine nitration is involved in the pathogenesis of highly fatal astrocytomas, a type of brain cancer. To understand the molecular mechanisms of astrocytomas and to discover new biomarkers/therapeutic targets, we sought to identify nitroproteins in human astrocytoma tissue. Anti-nitrotyrosine immunoreaction-positive proteins from a high-grade astrocytoma tissue were detected with two-dimensional gel electrophoresis (2DGE)-based nitrotyrosine immunoblots, and identified with liquid chromatography-tandem mass spectrometry (LC-MS/MS). Fifty-seven nitrotyrosine immunopositive protein spots were detected. A total of 870 proteins (nitrated and non-nitrated) in nitrotyrosine-immunopositive 2D gel spots were identified, and 18 nitroproteins and their 20 nitrotyrosine sites were identified with MS/MS analysis. These nitroproteins participate in multiple processes, including drug-resistance, signal transduction, cytoskeleton, transcription and translation, cell proliferation and apoptosis, immune response, phenotypic dedifferentiation, cell migration, and metastasis. Among those nitroproteins that might play a role in astrocytomas was nitro-sorcin, which is involved in drug resistance and metastasis and might play a role in the spread and treatment of an astrocytoma. Semiquantitative immune-based measurements of different sorcin expressions were found among different grades of astrocytomas relative to controls, and a semiquantitative increased nitration level in high-grade astrocytoma relative to control. Nitro-β-tubulin functions in cytoskeleton and cell migration. Semiquantitative immunoreactivity of β-tubulin showed increased expression among different grades of astrocytomas relative to controls and semiquantitatively increased nitration level in high-grade astrocytoma relative to control. Each nitroprotein was rationalized and related to the corresponding functional system to provide new insights into tyrosine nitration and its potential role in the pathogenesis of astrocytoma formation. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Fang Peng
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Jianglin Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, 410018, People's Republic of China
| | - Tianyao Guo
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Haiyan Yang
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Changsha, Hunan, 410013, People's Republic of China
| | - Maoyu Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Shushan Sang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Dominic M Desiderio
- The Charles B. Stout Neuroscience Mass Spectrometry Laboratory, Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China.
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Hunan, 410008, People's Republic of China.
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
- The State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| |
Collapse
|
19
|
Berges R, Baeza-Kallee N, Tabouret E, Chinot O, Petit M, Kruczynski A, Figarella-Branger D, Honore S, Braguer D. End-binding 1 protein overexpression correlates with glioblastoma progression and sensitizes to Vinca-alkaloids in vitro and in vivo. Oncotarget 2015; 5:12769-87. [PMID: 25473893 PMCID: PMC4350359 DOI: 10.18632/oncotarget.2646] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 10/26/2014] [Indexed: 01/08/2023] Open
Abstract
End-binding 1 protein (EB1) is a key player in the regulation of microtubule (MT) dynamics. Here, we investigated the role of EB1 in glioblastoma (GBM) tumor progression and its potential predictive role for response to Vinca-alkaloid chemotherapy. Immunohistological analysis of the 109 human GBM cases revealed that EB1 overexpression correlated with poor outcome including progression-free survival and overall survival. Downregulation of EB1 by shRNA inhibited cell migration and proliferation in vitro. Conversely, EB1 overexpression promoted them and accelerated tumor growth in orthotopically-transplanted nude mice. Furthermore, EB1 was largely overexpressed in stem-like GBM6 that display in vivo a higher tumorigenicity with a more infiltrative pattern of migration than stem-like GBM9. GBM6 showed strong and EB1-dependent migratory potential. The predictive role of EB1 in the response of GBM cells to chemotherapy was investigated. Vinflunine and vincristine increased survival of EB1-overexpressing U87 bearing mice and were more effective to inhibit cell migration and proliferation in EB1-overexpressing clones than in controls. Vinca inhibited the increase of MT growth rate and growth length induced by EB1 overexpression. Altogether, our results show that EB1 expression level has a prognostic value in GBM, and that Vinca-alkaloid chemotherapy could improve the treatment of GBM patients with EB1-overexpressing tumor.
Collapse
Affiliation(s)
- Raphael Berges
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Marseille 13385, France
| | | | - Emeline Tabouret
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Marseille 13385, France. APHM, CHU Timone, Marseille 13385, France
| | - Olivier Chinot
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Marseille 13385, France. APHM, CHU Timone, Marseille 13385, France
| | - Marie Petit
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Marseille 13385, France. APHM, CHU Timone, Marseille 13385, France
| | - Anna Kruczynski
- Centre de Recherche d'Oncologie Expérimentale, Institut de Recherche Pierre Fabre, Toulouse, France
| | - Dominique Figarella-Branger
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Marseille 13385, France. APHM, CHU Timone, Marseille 13385, France
| | - Stephane Honore
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Marseille 13385, France. APHM, CHU Timone, Marseille 13385, France
| | - Diane Braguer
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Marseille 13385, France. APHM, CHU Timone, Marseille 13385, France
| |
Collapse
|
20
|
Katsetos CD, Reginato MJ, Baas PW, D'Agostino L, Legido A, Tuszyn Ski JA, Dráberová E, Dráber P. Emerging microtubule targets in glioma therapy. Semin Pediatr Neurol 2015; 22:49-72. [PMID: 25976261 DOI: 10.1016/j.spen.2015.03.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Major advances in the genomics and epigenomics of diffuse gliomas and glioblastoma to date have not been translated into effective therapy, necessitating pursuit of alternative treatment approaches for these therapeutically challenging tumors. Current knowledge of microtubules in cancer and the development of new microtubule-based treatment strategies for high-grade gliomas are the topic in this review article. Discussed are cellular, molecular, and pharmacologic aspects of the microtubule cytoskeleton underlying mitosis and interactions with other cellular partners involved in cell cycle progression, directional cell migration, and tumor invasion. Special focus is placed on (1) the aberrant overexpression of βIII-tubulin, a survival factor associated with hypoxic tumor microenvironment and dynamic instability of microtubules; (2) the ectopic overexpression of γ-tubulin, which in addition to its conventional role as a microtubule-nucleating protein has recently emerged as a transcription factor interacting with oncogenes and kinases; (3) the microtubule-severing ATPase spastin and its emerging role in cell motility of glioblastoma cells; and (4) the modulating role of posttranslational modifications of tubulin in the context of interaction of microtubules with motor proteins. Specific antineoplastic strategies discussed include downregulation of targeted molecules aimed at achieving a sensitization effect on currently used mainstay therapies. The potential role of new classes of tubulin-binding agents and ATPase inhibitors is also examined. Understanding the cellular and molecular mechanisms underpinning the distinct behaviors of microtubules in glioma tumorigenesis and drug resistance is key to the discovery of novel molecular targets that will fundamentally change the prognostic outlook of patients with diffuse high-grade gliomas.
Collapse
Affiliation(s)
- Christos D Katsetos
- Department of Pediatrics, Drexel University College of Medicine, Section of Neurology and Pediatric Neuro-oncology Program, St Christopher's Hospital for Children, Philadelphia, PA; Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, Philadelphia, PA.
| | - Mauricio J Reginato
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA
| | - Peter W Baas
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA
| | - Luca D'Agostino
- Department of Pediatrics, Drexel University College of Medicine, Section of Neurology and Pediatric Neuro-oncology Program, St Christopher's Hospital for Children, Philadelphia, PA
| | - Agustin Legido
- Department of Pediatrics, Drexel University College of Medicine, Section of Neurology and Pediatric Neuro-oncology Program, St Christopher's Hospital for Children, Philadelphia, PA
| | - Jack A Tuszyn Ski
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada; Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Eduarda Dráberová
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Pavel Dráber
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
21
|
ROS-mediated EB1 phosphorylation through Akt/GSK3β pathway: implication in cancer cell response to microtubule-targeting agents. Oncotarget 2015; 5:3408-23. [PMID: 24930764 PMCID: PMC4102819 DOI: 10.18632/oncotarget.1982] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Microtubule-targeting agents (MTAs) are largely administered in adults and children cancers. Better deciphering their mechanism of action is of prime importance to develop more convenient therapy strategies. Here, we addressed the question of how reactive oxygen species (ROS) generation by mitochondria can be necessary for MTA efficacy. We showed for the first time that EB1 associates with microtubules in a phosphorylation-dependent manner, under control of ROS. By using phospho-defective mutants, we further characterized the Serine 155 residue as critical for EB1 accumulation at microtubule plus-ends, and both cancer cell migration and proliferation. Phosphorylation of EB1 on the Threonine 166 residue triggered opposite effects, and was identified as a requisite molecular switch in MTA activities. We then showed that GSK3β activation was responsible for MTA-triggered EB1 phosphorylation, resulting from ROS-mediated inhibition of upstream Akt. We thus disclosed here a novel pathway by which generation of mitochondrial ROS modulates microtubule dynamics through phosphorylation of EB1, improving our fundamental knowledge about this oncogenic protein, and pointing out the need to re-examine the current dogma of microtubule targeting by MTAs. The present work also provides a strong mechanistic rational to the promising therapeutic strategies that currently combine MTAs with anti-Akt targeted therapies.
Collapse
|
22
|
Morjen M, Honoré S, Bazaa A, Abdelkafi-Koubaa Z, Ellafi A, Mabrouk K, Kovacic H, El Ayeb M, Marrakchi N, Luis J. PIVL, a snake venom Kunitz-type serine protease inhibitor, inhibits in vitro and in vivo angiogenesis. Microvasc Res 2014; 95:149-56. [PMID: 25173589 DOI: 10.1016/j.mvr.2014.08.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 08/17/2014] [Accepted: 08/21/2014] [Indexed: 12/22/2022]
Abstract
Development and homeostasis of the vascular system requires integrin-promoting endothelial cell adhesion, migration and survival. Nowadays, integrins represent potential targets for pharmacological agents and open new avenues for the control of metastatic spread in the treatment of tumor malignancies. We have already reported that PIVL, a serine protease inhibitor isolated from Macrovipera lebetina venom, displays an anti-tumor effect through interference with integrin receptor function. Here, we report that PIVL inhibits human vascular endothelial cell adhesion and migration onto fibrinogen and fibronectin in a dose-dependent manner without any cytotoxicity. Furthermore, we show that PIVL increases microtubule dynamic instability in HMEC-1 transfected with EGFP-tagged α-tubulin. Using Matrigel™ and chick chorioallantoic membrane assays, we demonstrate that PIVL exhibits a strong anti-angiogenic effect both in vitro and in vivo. Interestingly, results herein reveal that the potent anti-angiogenic properties of PIVL are mediated by its RGD-like motif ((41)RGN(43)).
Collapse
Affiliation(s)
- Maram Morjen
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia.
| | - Stéphane Honoré
- Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale, UMR_S 911, Marseille, France; APHM, Hôpital Timone, Service Pharmacie, Marseille, France
| | - Amine Bazaa
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia
| | | | - Ameneallah Ellafi
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia
| | - Kamel Mabrouk
- Equipe CROPS, Institut de Chimie Radicalaire - UMR 7273, Université d'Aix-Marseille, Site de Saint Jérôme, Av. Escadrille Normandie Niemen, 13397 Marseille, France
| | - Hervé Kovacic
- APHM, Hôpital Timone, Service Pharmacie, Marseille, France
| | - Mohamed El Ayeb
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia
| | - Naziha Marrakchi
- Laboratoire des Venins et Biomolécules Thérapeutiques, Institut Pasteur de Tunis, Tunisia; Faculté de Médecine de Tunis, Tunisia
| | - José Luis
- APHM, Hôpital Timone, Service Pharmacie, Marseille, France
| |
Collapse
|
23
|
Andre J, Kerry L, Qi X, Hawkins E, Drizyte K, Ginger ML, McKean PG. An alternative model for the role of RP2 protein in flagellum assembly in the African trypanosome. J Biol Chem 2013; 289:464-75. [PMID: 24257747 PMCID: PMC3879569 DOI: 10.1074/jbc.m113.509521] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The tubulin cofactor C domain-containing protein TbRP2 is a basal body (centriolar) protein essential for axoneme formation in the flagellate protist Trypanosoma brucei, the causal agent of African sleeping sickness. Here, we show how TbRP2 is targeted and tethered at mature basal bodies and provide novel insight into TbRP2 function. Regarding targeting, understanding how several hundred proteins combine to build a microtubule axoneme is a fundamental challenge in eukaryotic cell biology. We show that basal body localization of TbRP2 is mediated by twinned, N-terminal TOF (TON1, OFD1, and FOP) and LisH motifs, motifs that otherwise facilitate localization of only a few conserved proteins at microtubule-organizing centers in animals, plants, and flagellate protists. Regarding TbRP2 function, there is a debate as to whether the flagellar assembly function of specialized, centriolar tubulin cofactor C domain-containing proteins is processing tubulin, the major component of axonemes, or general vesicular trafficking in a flagellum assembly context. Here we report that TbRP2 is required for the recruitment of T. brucei orthologs of MKS1 and MKS6, proteins that, in animal cells, are part of a complex that assembles at the base of the flagellum to regulate protein composition and cilium function. We also identify that TbRP2 is detected by YL1/2, an antibody classically used to detect α-tubulin. Together, these data suggest a general processing role for TbRP2 in trypanosome flagellum assembly and challenge the notion that TbRP2 functions solely in assessing tubulin “quality” prior to tubulin incorporation into the elongating axoneme.
Collapse
Affiliation(s)
- Jane Andre
- From the Faculty of Health and Medicine, Biomedical and Life Sciences, Lancaster University, Lancaster LA1 4YQ, United Kingdom
| | | | | | | | | | | | | |
Collapse
|