101
|
Sandilands E, Schoenherr C, Frame MC. p70S6K is regulated by focal adhesion kinase and is required for Src-selective autophagy. Cell Signal 2015; 27:1816-23. [PMID: 26071201 PMCID: PMC4508348 DOI: 10.1016/j.cellsig.2015.05.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/25/2015] [Accepted: 05/27/2015] [Indexed: 12/23/2022]
Abstract
Here we report that focal adhesion kinase (FAK) is required for optimal signalling to the Akt-p70S6K-S6 pathway in squamous cell carcinoma (SCC) cells. Specifically, in SCCs that are genetically deficient for FAK, there is reduced phosphorylation of Akt, p70S6K and S6, and signalling to Akt-p70S6K-S6 is more sensitive to inhibition by multiple agents that suppress the pathway. By contrast, mTOR is unaffected. Indeed, pharmacological agents that inhibit the Akt-p70S6K-S6 pathway, and PDK1 that lies upstream of Akt, also impair the autophagic targeting of activated c-Src (p-Src) in FAK deficient cells. This is associated with loss of a complex between p-Src and the autophagy protein LC3, a biochemical surrogate of impaired Src-selective autophagy. In keeping with a vital role for p70S6K, inhibition by a selective inhibitor and specific siRNA also impaired Src-selective autophagy. Finally, components of the PDK1-Akt-p70S6K signalling pathway were co-located with p-Src at autophagosomes, and Src and p70S6K co-exist in the same biochemical complex. We therefore deduce that the FAK-regulated signalling module PDK1-Akt-p70S6K that controls Src's intracellular trafficking operates at Src-containing autophagosomes.
Collapse
Affiliation(s)
- Emma Sandilands
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Christina Schoenherr
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, United Kingdom
| | - Margaret C Frame
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh EH4 2XR, United Kingdom.
| |
Collapse
|
102
|
Xu Z, Yang L, Xu S, Zhang Z, Cao Y. The receptor proteins: pivotal roles in selective autophagy. Acta Biochim Biophys Sin (Shanghai) 2015; 47:571-80. [PMID: 26112016 DOI: 10.1093/abbs/gmv055] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/05/2015] [Accepted: 03/30/2015] [Indexed: 02/05/2023] Open
Abstract
Autophagy is a highly regulated and multistep biological process whereby cells under metabolic, proteotoxic, or other stresses remove dysfunctional organelles and/or misfolded/polyubiquitinated proteins by shuttling them via specialized structures called autophagosomes to the lysosome for degradation. Although autophagy is generally considered to be a non-selective process, accumulating evidence suggests that it can also selectively degrade specific target cargoes. These selective targets include proteins, mitochondria, and even invading bacteria. The discovery and characterization of autophagic adapters, such as p62/Sequestosome 1 (SQSTM1) and Neighbor of BRCA1 gene 1 (NBR1), have provided mechanistic insights into selective autophagy. These receptors are all able to act as cargo receptors for the degradation of ubiquitinated substrates. This review mainly summarizes the most up-to-date findings regarding the key receptor proteins that play important roles in regulating selective autophagy.
Collapse
Affiliation(s)
- Zhijie Xu
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| | - Lifang Yang
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| | - San Xu
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| | - Zhibao Zhang
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| | - Ya Cao
- Cancer Research Institute, Central South University, Changsha 410078, China Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha 410078, China Key Laboratory of Carcinogenesis, Ministry of Health, Changsha 410078, China
| |
Collapse
|
103
|
Abstract
Macro-autophagy is a major catabolic process in the cell used to degrade protein aggregates, dysfunctional organelles and intracellular pathogens that would otherwise become toxic. Autophagy also generates energy and metabolites for the cell through recycling of degraded autophagosomal cargo, which can be particularly important for cell viability under stress. The significance of changes in the rates of autophagic flux for cellular function and disease is being increasingly appreciated, and interest in measuring autophagy in different experimental systems is growing accordingly. Here, we describe key methodologies used in the field to measure autophagic flux, including monitoring LC3 processing by western blot, fluorescent cell staining, and flow cytometry, in addition to changes in the levels or posttranslational modifications of other autophagy markers, such as p62/Sqstm1 and the Atg5-Atg12 conjugate. We also describe what cellular stresses may be used to induce autophagy and how to control for changes in the rates of autophagic flux as opposed to inhibition of flux. Finally, we detail available techniques to monitor autophagy in vivo.
Collapse
Affiliation(s)
- Marina N. Sharifi
- The Ben May Department for Cancer Research, The Gordon
Center for Integrative Sciences, 929 East 57 Street, The University of
Chicago, Chicago, IL 60637
- The Committee on Cancer Biology, The Gordon Center for
Integrative Sciences, 929 East 57 Street, The University of Chicago,
Chicago, IL 60637
- Medical Scientist Training Program, The Gordon Center for
Integrative Sciences, 929 East 57 Street, The University of Chicago,
Chicago, IL 60637
| | - Erin E. Mowers
- The Ben May Department for Cancer Research, The Gordon
Center for Integrative Sciences, 929 East 57 Street, The University of
Chicago, Chicago, IL 60637
- Medical Scientist Training Program, The Gordon Center for
Integrative Sciences, 929 East 57 Street, The University of Chicago,
Chicago, IL 60637
| | - Lauren E Drake
- The Ben May Department for Cancer Research, The Gordon
Center for Integrative Sciences, 929 East 57 Street, The University of
Chicago, Chicago, IL 60637
- The Committee on Molecular Pathogenesis and Molecular
Medicine, The Gordon Center for Integrative Sciences, 929 East 57
Street, The University of Chicago, Chicago, IL 60637
| | - Kay F Macleod
- The Ben May Department for Cancer Research, The Gordon
Center for Integrative Sciences, 929 East 57 Street, The University of
Chicago, Chicago, IL 60637
- The Committee on Cancer Biology, The Gordon Center for
Integrative Sciences, 929 East 57 Street, The University of Chicago,
Chicago, IL 60637
- The Committee on Molecular Pathogenesis and Molecular
Medicine, The Gordon Center for Integrative Sciences, 929 East 57
Street, The University of Chicago, Chicago, IL 60637
- Corresponding author: Tel: 773-834-8309, Fax:
773-702-4476,
| |
Collapse
|
104
|
Schoenherr C, Serrels B, Proby C, Cunningham DL, Findlay JE, Baillie GS, Heath JK, Frame MC. Eps8 controls Src- and FAK-dependent phenotypes in squamous carcinoma cells. J Cell Sci 2014; 127:5303-16. [PMID: 25359883 PMCID: PMC4265741 DOI: 10.1242/jcs.157560] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/03/2014] [Accepted: 10/18/2014] [Indexed: 01/19/2023] Open
Abstract
Eps8 is an actin regulatory scaffold protein whose expression is increased in squamous cell carcinoma (SCC) cells. It forms a complex with both focal adhesion kinase (FAK, also known as PTK2) and Src in SCC cells derived from skin carcinomas induced by administration of the chemical DMBA followed by TPA (the DMBA/TPA model). Here, we describe two new roles for Eps8. Firstly, it controls the spatial distribution of active Src in a FAK-dependent manner. Specifically, Eps8 participates in, and regulates, a biochemical complex with Src and drives trafficking of Src to autophagic structures that SCC cells use to cope with high levels of active Src when FAK is absent. Secondly, when FAK is expressed in SCC cells, thereby meaning active Src becomes tethered at focal adhesion complexes, Eps8 is also recruited to focal adhesions and is required for FAK-dependent polarization and invasion. Therefore, Eps8 is a crucial mediator of Src- and FAK-regulated processes; it participates in specific biochemical complexes and promotes actin re-arrangements that determine the spatial localization of Src, and modulates the functions of Src and FAK during invasive migration.
Collapse
Affiliation(s)
- Christina Schoenherr
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XR Edinburgh, UK
| | - Bryan Serrels
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XR Edinburgh, UK
| | - Charlotte Proby
- Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Debbie L Cunningham
- Cancer Research UK Growth Factor Signalling Group, School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Jane E Findlay
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - George S Baillie
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - John K Heath
- Cancer Research UK Growth Factor Signalling Group, School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Margaret C Frame
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XR Edinburgh, UK
| |
Collapse
|
105
|
FAK signaling in human cancer as a target for therapeutics. Pharmacol Ther 2014; 146:132-49. [PMID: 25316657 DOI: 10.1016/j.pharmthera.2014.10.001] [Citation(s) in RCA: 307] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/06/2014] [Accepted: 10/07/2014] [Indexed: 02/08/2023]
Abstract
Focal adhesion kinase (FAK) is a key regulator of growth factor receptor- and integrin-mediated signals, governing fundamental processes in normal and cancer cells through its kinase activity and scaffolding function. Increased FAK expression and activity occurs in primary and metastatic cancers of many tissue origins, and is often associated with poor clinical outcome, highlighting FAK as a potential determinant of tumor development and metastasis. Indeed, data from cell culture and animal models of cancer provide strong lines of evidence that FAK promotes malignancy by regulating tumorigenic and metastatic potential through highly-coordinated signaling networks that orchestrate a diverse range of cellular processes, such as cell survival, proliferation, migration, invasion, epithelial-mesenchymal transition, angiogenesis and regulation of cancer stem cell activities. Such an integral role in governing malignant characteristics indicates that FAK represents a potential target for cancer therapeutics. While pharmacologic targeting of FAK scaffold function is still at an early stage of development, a number of small molecule-based FAK tyrosine kinase inhibitors are currently undergoing pre-clinical and clinical testing. In particular, PF-00562271, VS-4718 and VS-6063 show promising clinical activities in patients with selected solid cancers. Clinical testing of rationally designed FAK-targeting agents with implementation of predictive response biomarkers, such as merlin deficiency for VS-4718 in mesothelioma, may help improve clinical outcome for cancer patients. In this article, we have reviewed the current knowledge regarding FAK signaling in human cancer, and recent developments in the generation and clinical application of FAK-targeting pharmacologic agents.
Collapse
|
106
|
The Role of Src Kinase in the Caspase-1 Pathway After Hypoxia in the Brain of Newborn Piglets. Neurochem Res 2014; 39:2118-26. [DOI: 10.1007/s11064-014-1404-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/16/2014] [Revised: 06/28/2014] [Accepted: 07/28/2014] [Indexed: 01/06/2023]
|
107
|
Abstract
Selective autophagy is a quality control pathway through which cellular components are sequestered into double-membrane vesicles and delivered to specific intracellular compartments. This process requires autophagy receptors that link cargo to growing autophagosomal membranes. Selective autophagy is also implicated in various membrane trafficking events. Here we discuss the current view on how cargo selection and transport are achieved during selective autophagy, and point out molecular mechanisms that are congruent between autophagy and vesicle trafficking pathways.
Collapse
|
108
|
Gilad Y, Shiloh R, Ber Y, Bialik S, Kimchi A. Discovering protein-protein interactions within the programmed cell death network using a protein-fragment complementation screen. Cell Rep 2014; 8:909-21. [PMID: 25066129 DOI: 10.1016/j.celrep.2014.06.049] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/28/2014] [Revised: 06/08/2014] [Accepted: 06/25/2014] [Indexed: 12/22/2022] Open
Abstract
Apoptosis and autophagy are distinct biological processes, each driven by a different set of protein-protein interactions, with significant crosstalk via direct interactions among apoptotic and autophagic proteins. To measure the global profile of these interactions, we adapted the Gaussia luciferase protein-fragment complementation assay (GLuc PCA), which monitors binding between proteins fused to complementary fragments of a luciferase reporter. A library encompassing 63 apoptotic and autophagic proteins was constructed for the analysis of ∼3,600 protein-pair combinations. This generated a detailed landscape of the apoptotic and autophagic modules and points of interface between them, identifying 46 previously unknown interactions. One of these interactions, between DAPK2, a Ser/Thr kinase that promotes autophagy, and 14-3-3τ, was further investigated. We mapped the region responsible for 14-3-3τ binding and proved that this interaction inhibits DAPK2 dimerization and activity. This proof of concept underscores the power of the GLuc PCA platform for the discovery of biochemical pathways within the cell death network.
Collapse
Affiliation(s)
- Yuval Gilad
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ruth Shiloh
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yaara Ber
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Shani Bialik
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
109
|
Tyryshkin A, Bhattacharya A, Eissa NT. SRC kinase is a novel therapeutic target in lymphangioleiomyomatosis. Cancer Res 2014; 74:1996-2005. [PMID: 24691995 DOI: 10.1158/0008-5472.can-13-1256] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/12/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a progressive cystic lung disease affecting some women with tuberous sclerosis complex (TSC). Sporadic LAM can develop in women without TSC, owing to somatic mutations in the TSC2 gene. Accumulating evidence supports the view of LAM as a low-grade, destructive, metastasizing neoplasm. The mechanisms underlying the metastatic capability of LAM cells remain poorly understood. The observed behavior of LAM cells with respect to their infiltrative growth pattern, metastatic potential, and altered cell differentiation bears similarity to cells undergoing epithelial-mesenchymal transition. Here, we report increased levels of active Src kinase in LAM lungs and in TSC2(-/-) cells, caused by a reduction of autophagy. Furthermore, increased Src kinase activation promoted migration, invasion, and inhibition of E-cadherin expression in TSC2(-/-) cells by upregulating the transcription factor Snail. Notably, Src kinase inhibitors reduced migration and invasion properties of TSC2(-/-) cells and attenuated lung colonization of intravenously injected TSC2(-/-) cells in vivo to a greater extent than control TSC2(+/+) cells. Our results reveal mechanistic basis for the pathogenicity of LAM cells and they rationalize Src kinase as a novel therapeutic target for treatment of LAM and TSC.
Collapse
Affiliation(s)
- Alexey Tyryshkin
- Authors' Affiliation: Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | | | |
Collapse
|
110
|
Abstract
(Macro)autophagy is a fundamental degradation process for macromolecules and organelles of vital importance for cell and tissue homeostasis. Autophagy research has gained a strong momentum in recent years because of its relevance to cancer, neurodegenerative diseases, muscular dystrophy, lipid storage disorders, development, ageing and innate immunity. Autophagy has traditionally been thought of as a bulk degradation process that is mobilized upon nutritional starvation to replenish the cell with building blocks and keep up with the energy demand. This view has recently changed dramatically following an array of papers describing various forms of selective autophagy. A main driving force has been the discovery of specific autophagy receptors that sequester cargo into forming autophagosomes (phagophores). At the heart of this selectivity lies the LC3-interacting region (LIR) motif, which ensures the targeting of autophagy receptors to LC3 (or other ATG8 family proteins) anchored in the phagophore membrane. LIR-containing proteins include cargo receptors, members of the basal autophagy apparatus, proteins associated with vesicles and of their transport, Rab GTPase-activating proteins (GAPs) and specific signaling proteins that are degraded by selective autophagy. Here, we comment on these new insights and focus on the interactions of LIR-containing proteins with members of the ATG8 protein family.
Collapse
Affiliation(s)
- Åsa Birna Birgisdottir
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø, 9037 Tromsø, Norway
| | | | | |
Collapse
|
111
|
Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature 2014; 509:105-9. [PMID: 24695223 PMCID: PMC4180099 DOI: 10.1038/nature13148] [Citation(s) in RCA: 1312] [Impact Index Per Article: 119.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/09/2013] [Accepted: 02/12/2014] [Indexed: 02/07/2023]
Abstract
Autophagy, the process by which proteins and organelles are sequestered in double-membrane structures called autophagosomes and delivered to lysosomes for degradation, is critical in diseases such as cancer and neurodegeneration1,2. Much of our understanding of this process has emerged from analysis of bulk cytoplasmic autophagy, but our understanding of how specific cargo including organelles, proteins, or intracellular pathogens are targeted for selective autophagy is limited3. We employed quantitative proteomics to identify a cohort of novel and known autophagosome-enriched proteins, including cargo receptors. Like known cargo receptors, NCOA4 was highly enriched in autophagosomes, and associated with ATG8 proteins that recruit cargo-receptor complexes into autophagosomes. Unbiased identification of NCOA4-associated proteins revealed ferritin heavy and light chains, components of an iron-filled cage structure that protects cells from reactive iron species4 but is degraded via autophagy to release iron5,6 through an unknown mechanism. We found that delivery of ferritin to lysosomes required NCOA4, and an inability of NCOA4-deficient cells to degrade ferritin leads to decreased bioavailable intracellular iron. This work identifies NCOA4 as a selective cargo receptor for autophagic turnover of ferritin (ferritinophagy) critical for iron homeostasis and provides a resource for further dissection of autophagosomal cargo-receptor connectivity.
Collapse
|
112
|
Macagno JP, Diaz Vera J, Yu Y, MacPherson I, Sandilands E, Palmer R, Norman JC, Frame M, Vidal M. FAK acts as a suppressor of RTK-MAP kinase signalling in Drosophila melanogaster epithelia and human cancer cells. PLoS Genet 2014; 10:e1004262. [PMID: 24676055 PMCID: PMC3967952 DOI: 10.1371/journal.pgen.1004262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/12/2013] [Accepted: 02/10/2014] [Indexed: 11/18/2022] Open
Abstract
Receptor Tyrosine Kinases (RTKs) and Focal Adhesion Kinase (FAK) regulate multiple signalling pathways, including mitogen-activated protein (MAP) kinase pathway. FAK interacts with several RTKs but little is known about how FAK regulates their downstream signalling. Here we investigated how FAK regulates signalling resulting from the overexpression of the RTKs RET and EGFR. FAK suppressed RTKs signalling in Drosophila melanogaster epithelia by impairing MAPK pathway. This regulation was also observed in MDA-MB-231 human breast cancer cells, suggesting it is a conserved phenomenon in humans. Mechanistically, FAK reduced receptor recycling into the plasma membrane, which resulted in lower MAPK activation. Conversely, increasing the membrane pool of the receptor increased MAPK pathway signalling. FAK is widely considered as a therapeutic target in cancer biology; however, it also has tumour suppressor properties in some contexts. Therefore, the FAK-mediated negative regulation of RTK/MAPK signalling described here may have potential implications in the designing of therapy strategies for RTK-driven tumours.
Collapse
Affiliation(s)
- Juan Pablo Macagno
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Jesica Diaz Vera
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Yachuan Yu
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Iain MacPherson
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Emma Sandilands
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Ruth Palmer
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Jim C. Norman
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Margaret Frame
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Marcos Vidal
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| |
Collapse
|
113
|
|
114
|
Modulation of apoptosis sensitivity through the interplay with autophagic and proteasomal degradation pathways. Cell Death Dis 2014; 5:e1011. [PMID: 24457955 PMCID: PMC4040655 DOI: 10.1038/cddis.2013.520] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/28/2013] [Revised: 11/16/2013] [Accepted: 11/21/2013] [Indexed: 12/24/2022]
Abstract
Autophagic and proteasomal degradation constitute the major cellular proteolysis pathways. Their physiological and pathophysiological adaptation and perturbation modulates the relative abundance of apoptosis-transducing proteins and thereby can positively or negatively adjust cell death susceptibility. In addition to balancing protein expression amounts, components of the autophagic and proteasomal degradation machineries directly interact with and co-regulate apoptosis signal transduction. The influence of autophagic and proteasomal activity on apoptosis susceptibility is now rapidly gaining more attention as a significant modulator of cell death signalling in the context of human health and disease. Here we present a concise and critical overview of the latest knowledge on the molecular interplay between apoptosis signalling, autophagy and proteasomal protein degradation. We highlight that these three pathways constitute an intricate signalling triangle that can govern and modulate cell fate decisions between death and survival. Owing to rapid research progress in recent years, it is now possible to provide detailed insight into the mechanisms of pathway crosstalk, common signalling nodes and the role of multi-functional proteins in co-regulating both protein degradation and cell death.
Collapse
|
115
|
Gocek E, Moulas AN, Studzinski GP. Non-receptor protein tyrosine kinases signaling pathways in normal and cancer cells. Crit Rev Clin Lab Sci 2014; 51:125-37. [PMID: 24446827 DOI: 10.3109/10408363.2013.874403] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/06/2023]
Abstract
Protein tyrosine kinases (PTKs) are enzymes that transfer phosphate groups to tyrosine residues on protein substrates. Phosphorylation of proteins causes changes in their function and/or enzymatic activity resulting in specific biological responses. There are two classes of PTKs: the transmembrane receptor PTKs and the cytoplasmic non-receptor PTKs (NRTKs). NRTKs are involved in transduction of signals originating from extracellular clues, which often interact with transmembrane receptors. Thus, they are important components of signaling pathways which regulate fundamental cellular functions such as cell differentiation, apoptosis, survival, and proliferation. The activity of NRTKs is tightly regulated, and de-regulation and/or overexpression of NRTKs has been implicated in malignant transformation and carcinogenesis. Research on NRTKs has shed light on the mechanisms of a number of cellular processes including those involved in carcinogenesis. Not surprisingly, several tyrosine kinase inhibitors are in use as treatment for a number of malignancies, and more are under investigation. This review deals with the structure, function, and signaling pathways of nine main families of NRTKs in normal and cancer cells.
Collapse
Affiliation(s)
- Elzbieta Gocek
- Department of Protein Biotechnology, Faculty of Biotechnology, University of Wroclaw , Wroclaw , Poland
| | | | | |
Collapse
|
116
|
Feng W, Chang C, Luo D, Su H, Yu S, Hua W, Chen Z, Hu H, Liu W. Dissection of autophagy in human platelets. Autophagy 2014; 10:642-51. [PMID: 24458007 PMCID: PMC4091151 DOI: 10.4161/auto.27832] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/18/2023] Open
Abstract
Continuous turnover of intracellular components by autophagy is necessary to preserve cellular homeostasis in all tissues. Despite recent advances in identifying autophagy-related genes and understanding the functions of autophagy in developmental and pathological conditions, so far, the role of autophagy in platelet, a specific anucleate cell type, is poorly understood. In this study, we showed that human platelets express the autophagy-related proteins ATG5, ATG7, and LC3. The same as in nucleated mammalian cells, autophagy was stimulated by cell starvation or the MTOR inhibitor rapamycin in a phosphatidylinositol 3-kinase (PtdIns3K)-dependent manner. Disruption of autophagic flux led to impairment of platelet aggregation and adhesion. Furthermore, Becn1 heterozygous knockout mice displayed a prolonged bleeding time and reduced platelet aggregation. These results suggest a potential role of autophagy in the regulation of platelet function, and imply that gene transcription may not be an essential prerequisite for adaptive autophagy.
Collapse
Affiliation(s)
- Wenfeng Feng
- Department of Biochemistry and Molecular Biology; Program in Molecular and Cell Biology; Zhejiang University School of Medicine; Hangzhou, China
| | - Chunmei Chang
- Department of Biochemistry and Molecular Biology; Program in Molecular and Cell Biology; Zhejiang University School of Medicine; Hangzhou, China
| | - Dongjiao Luo
- Department of Pathology and Pathophysiology; Zhejiang University School of Medicine; Hangzhou, China
| | - Hua Su
- Department of Biochemistry and Molecular Biology; Program in Molecular and Cell Biology; Zhejiang University School of Medicine; Hangzhou, China
| | - Shanshan Yu
- Department of Pathology and Pathophysiology; Zhejiang University School of Medicine; Hangzhou, China
| | - Wen Hua
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou, China
| | - Zhihua Chen
- Department of Respiratory Medicine; Second Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou, China
| | - Hu Hu
- Department of Pathology and Pathophysiology; Zhejiang University School of Medicine; Hangzhou, China
| | - Wei Liu
- Department of Biochemistry and Molecular Biology; Program in Molecular and Cell Biology; Zhejiang University School of Medicine; Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease; First Affiliated Hospital; Zhejiang University School of Medicine; Hangzhou, China
| |
Collapse
|
117
|
Golubovskaya VM. Targeting FAK in human cancer: from finding to first clinical trials. Front Biosci (Landmark Ed) 2014; 19:687-706. [PMID: 24389213 DOI: 10.2741/4236] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/19/2022]
Abstract
It is twenty years since Focal Adhesion Kinase (FAK) was found to be overexpressed in many types of human cancer. FAK plays an important role in adhesion, spreading, motility, invasion, metastasis, survival, angiogenesis, and recently has been found to play an important role as well in epithelial to mesenchymal transition (EMT), cancer stem cells and tumor microenvironment. FAK has kinase-dependent and kinase independent scaffolding, cytoplasmic and nuclear functions. Several years ago FAK was proposed as a potential therapeutic target; the first clinical trials were just reported, and they supported further studies of FAK as a promising therapeutic target. This review discusses the main functions of FAK in cancer, and specifically focuses on recent novel findings on the role of FAK in cancer stem cells, microenvironment, epithelial-to-mesenchymal transition, invasion, metastasis, and also highlight new approaches of targeting FAK and critically discuss challenges that lie ahead for its targeted therapeutics. The review provides a summary of translational approaches of FAK-targeted and combination therapies and outline perspectives and future directions of FAK research.
Collapse
|
118
|
Abstract
Genetic ablation of endothelial focal adhesion kinase (FAK) can inhibit pathological angiogenesis, suggesting that loss of endothelial FAK is sufficient to reduce neovascularization. Here we show that reduced stromal FAK expression in FAK-heterozygous mice unexpectedly enhances both B16F0 and CMT19T tumour growth and angiogenesis. We further demonstrate that cell proliferation and microvessel sprouting, but not migration, are increased in serum-stimulated FAK-heterozygous endothelial cells. FAK-heterozygous endothelial cells display an imbalance in FAK phosphorylation at pY397 and pY861 without changes in Pyk2 or Erk1/2 activity. By contrast, serum-stimulated phosphorylation of Akt is enhanced in FAK-heterozygous endothelial cells and these cells are more sensitive to Akt inhibition. Additionally, low doses of a pharmacological FAK inhibitor, although too low to affect FAK autophosphorylation in vitro, can enhance angiogenesis ex vivo and tumour growth in vivo. Our results highlight a potential novel role for FAK as a nonlinear, dose-dependent regulator of angiogenesis where heterozygous levels of FAK enhance angiogenesis.
Collapse
|
119
|
Abstract
Continuous synthesis of all cellular components requires their constant turnover in order for a cell to achieve homeostasis. To this end, eukaryotic cells are endowed with two degradation pathways - the ubiquitin-proteasome system and the lysosomal pathway. The latter pathway is partly fed by autophagy, which targets intracellular material in distinct vesicles, termed autophagosomes, to the lysosome. Central to this pathway is a set of key autophagy proteins, including the ubiquitin-like modifier Atg8, that orchestrate autophagosome initiation and biogenesis. In higher eukaryotes, the Atg8 family comprises six members known as the light chain 3 (LC3) or γ-aminobutyric acid (GABA)-receptor-associated protein (GABARAP) proteins. Considerable effort during the last 15 years to decipher the molecular mechanisms that govern autophagy has significantly advanced our understanding of the functioning of this protein family. In this Cell Science at a Glance article and the accompanying poster, we present the current LC3 protein interaction network, which has been and continues to be vital for gaining insight into the regulation of autophagy.
Collapse
Affiliation(s)
- Philipp Wild
- Institute of Biochemistry II, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | | | | |
Collapse
|
120
|
Belaid A, Ndiaye PD, Cerezo M, Cailleteau L, Brest P, Klionsky DJ, Carle GF, Hofman P, Mograbi B. Autophagy and SQSTM1 on the RHOA(d) again: emerging roles of autophagy in the degradation of signaling proteins. Autophagy 2013; 10:201-8. [PMID: 24300375 DOI: 10.4161/auto.27198] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/28/2022] Open
Abstract
Degradation of signaling proteins is one of the most powerful tumor-suppressive mechanisms by which a cell can control its own growth, its survival, and its motility. Emerging evidence suggests that autophagy limits several signaling pathways by degrading kinases, downstream components, and transcription factors; however, this often occurs under stressful conditions. Our recent studies revealed that constitutive autophagy temporally and spatially controls the RHOA pathway. Specifically, inhibition of autophagosome degradation induces the accumulation of the GTP-bound form of RHOA. The active RHOA is sequestered via SQSTM1/p62 within autolysosomes, and accordingly fails to localize to the spindle midbody or to the cell surface, as we demonstrate herein. As a result, all RHOA-downstream responses are deregulated, thus driving cytokinesis failure, aneuploidy and motility, three processes that directly have an impact upon cancer progression. We therefore propose that autophagy acts as a degradative brake for RHOA signaling and thereby controls cell proliferation, migration, and genome stability.
Collapse
Affiliation(s)
- Amine Belaid
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France
| | - Papa Diogop Ndiaye
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France
| | - Michaël Cerezo
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; INSERM U895/C3M: Centre Méditerranéen de Médecine Moléculaire; Nice, France
| | - Laurence Cailleteau
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France
| | - Patrick Brest
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France
| | | | - Georges F Carle
- Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Centre Antoine Lacassagne; Nice, France; Laboratoire TIRO-MATOs UMR E4320; Commissariat à l'Energie Atomique; Nice, France
| | - Paul Hofman
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France; Centre Hospitalier Universitaire de Nice; Pasteur Hospital; Laboratory of Clinical and Experimental Pathology; Nice, France
| | - Baharia Mograbi
- Institute of Research on Cancer and Ageing of Nice (IRCAN); INSERM U1081; CNRS UMR7284; Nice, France; Université de Nice-Sophia Antipolis; Faculté de Médecine; Nice, France; Equipe Labellisée par l'ARC; Villejuif, France; Centre Antoine Lacassagne; Nice, France
| |
Collapse
|
121
|
Trichothecene mycotoxins activate NLRP3 inflammasome through a P2X7 receptor and Src tyrosine kinase dependent pathway. Hum Immunol 2013; 75:134-40. [PMID: 24269698 DOI: 10.1016/j.humimm.2013.11.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/18/2013] [Revised: 11/05/2013] [Accepted: 11/11/2013] [Indexed: 12/23/2022]
Abstract
Inflammasome is an intracellular molecular platform of the innate immunity that is a key mediator of inflammation. The inflammasome complex detects pathogens and different danger signals, and triggers cysteine protease caspase-1-dependent processing of pro-inflammatory cytokines IL-1β, and IL-18 in dendritic cells and macrophages. Previously, we have shown that water-damaged building associated trichothecene mycotoxins, including roridin A, trigger IL-1β and IL-18 secretion in human macrophages. However, the molecular basis as well as mechanism behind this trichothecene-induced cytokine secretion has remained uncharacterized. Here, we show that the trichothecene-induced IL-1β secretion is dependent on NLRP3 inflammasome in human primary macrophages. Pharmacological inhibition and small interfering RNA approach showed that the trichothecene-induced NLRP3 inflammasome activation is mediated through ATP-gated P2X7 receptor. Moreover, we show that trichothecene-triggered NLRP3 inflammasome activation is dependent on Src tyrosine kinase activity. In addition, gene silencing of c-Cbl, a negative autophagy-related regulator of c-Src, resulted in enhanced secretion of IL-1β and IL-18 in response to trichothecene mycotoxin stimulation in human macrophages. In conclusion, our results suggest that roridin A, a fungal trichothecene mycotoxin, acts as microbial danger signals that trigger activation of NLRP3 inflammasome through P2X7R and Src tyrosine kinase signaling dependent pathway in human primary macrophages.
Collapse
|
122
|
Lee BY, Hochgräfe F, Lin HM, Castillo L, Wu J, Raftery MJ, Martin Shreeve S, Horvath LG, Daly RJ. Phosphoproteomic profiling identifies focal adhesion kinase as a mediator of docetaxel resistance in castrate-resistant prostate cancer. Mol Cancer Ther 2013; 13:190-201. [PMID: 24194567 DOI: 10.1158/1535-7163.mct-13-0225-t] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
Docetaxel remains the standard-of-care for men diagnosed with metastatic castrate-resistant prostate cancer (CRPC). However, only approximately 50% of patients benefit from treatment and all develop docetaxel-resistant disease. Here, we characterize global perturbations in tyrosine kinase signaling associated with docetaxel resistance and thereby develop a potential therapeutic strategy to reverse this phenotype. Using quantitative mass spectrometry-based phosphoproteomics, we identified that metastatic docetaxel-resistant prostate cancer cell lines (DU145-Rx and PC3-Rx) exhibit increased phosphorylation of focal adhesion kinase (FAK) on Y397 and Y576, in comparison with parental controls (DU145 and PC3, respectively). Bioinformatic analyses identified perturbations in pathways regulating focal adhesions and the actin cytoskeleton and in protein-protein interaction networks related to these pathways in docetaxel-resistant cells. Treatment with the FAK tyrosine kinase inhibitor (TKI) PF-00562271 reduced FAK phosphorylation in the resistant cells, but did not affect cell viability or Akt phosphorylation. Docetaxel administration reduced FAK and Akt phosphorylation, whereas cotreatment with PF-00562271 and docetaxel resulted in an additive attenuation of FAK and Akt phosphorylation and overcame the chemoresistant phenotype. The enhanced efficacy of cotreatment was due to increased autophagic cell death, rather than apoptosis. These data strongly support that enhanced FAK activation mediates chemoresistance in CRPC, and identify a potential clinical niche for FAK TKIs, where coadministration with docetaxel may be used in patients with CRPC to overcome chemoresistance.
Collapse
Affiliation(s)
- Brian Y Lee
- Corresponding Author: Roger J. Daly, Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Level 1, Building 77, Monash University, VIC 3800, Australia. Telephone: 61-3-990-29301;
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Salotti J, Dias MH, Koga MM, Armelin HA. Fibroblast growth factor 2 causes G2/M cell cycle arrest in ras-driven tumor cells through a Src-dependent pathway. PLoS One 2013; 8:e72582. [PMID: 23991123 PMCID: PMC3753234 DOI: 10.1371/journal.pone.0072582] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/13/2011] [Accepted: 07/17/2013] [Indexed: 11/26/2022] Open
Abstract
We recently reported that paracrine Fibroblast Growth Factor 2 (FGF2) triggers senescence in Ras-driven Y1 and 3T3Ras mouse malignant cell lines. Here, we show that although FGF2 activates mitogenic pathways in these Ras-dependent malignant cells, it can block cell proliferation and cause a G2/M arrest. These cytostatic effects of FGF2 are inhibited by PD173074, an FGF receptor (FGFR) inhibitor. To determine which downstream pathways are induced by FGF2, we tested specific inhibitors targeting mitogen-activated protein kinase (MEK), phosphatidylinositol 3 kinase (PI3K) and protein kinase C (PKC). We show that these classical mitogenic pathways do not mediate the cytostatic activity of FGF2. On the other hand, the inhibition of Src family kinases rescued Ras-dependent malignant cells from the G2/M irreversible arrest induced by FGF2. Taken together, these data indicate a growth factor-sensitive point in G2/M that likely involves FGFR/Ras/Src pathway activation in a MEK, PI3K and PKC independent manner.
Collapse
Affiliation(s)
- Jacqueline Salotti
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Matheus H. Dias
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
- Instituto Butantan, CATcepid, São Paulo, Brazil
| | - Marianna M. Koga
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Hugo A. Armelin
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
- Instituto Butantan, CATcepid, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
124
|
Abstract
Ubiquitination is an important mechanism for the regulation of diverse cellular functions, including proteolysis and DNA repair. The human MutS family protein hMSH4 functions in meiotic recombinational DNA double-strand break (DSB) repair. It was previously observed that hMSH4 interacts with the von Hippel-Lindau binding protein 1 (VBP1), a partner of the VHL ubiquitin E3 ligase as well as a subunit of the prefoldin complex. In this study we address how ubiquitination regulates the homeostasis of hMSH4 in the human embryonic kidney cell line HEK293T. We demonstrate that VBP1 targets hMSH4 for degradation and identify a new VBP1 binding partner, p97, an AAA(+) ATPase involved in protein degradation and DNA damage response. VBP1, VHL, and p97 coexist in the hMSH4 immunocomplex and regulate the polyubiquitination of hMSH4. Furthermore, the results of this study demonstrate that VBP1 acts together with p97 to regulate hMSH4 degradation. Overall, this study has revealed a molecular mechanism by which VBP1 controls the levels of hMSH4 by ubiquitination in mitotic cells. Such a mechanism may be important for controlling the role of hMSH4 in regulating homologous recombination and nonhomologous DNA end joining-mediated DSB repair in human cells.
Collapse
Affiliation(s)
- Yang Xu
- 1School of Molecular Biosciences, Mail Drop 64-7520, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-7520, USA.
| | | |
Collapse
|
125
|
Liu WM, Huang P, Kar N, Burgett M, Muller-Greven G, Nowacki AS, Distelhorst CW, Lathia JD, Rich JN, Kappes JC, Gladson CL. Lyn facilitates glioblastoma cell survival under conditions of nutrient deprivation by promoting autophagy. PLoS One 2013; 8:e70804. [PMID: 23936469 PMCID: PMC3732228 DOI: 10.1371/journal.pone.0070804] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/22/2013] [Accepted: 05/23/2013] [Indexed: 11/19/2022] Open
Abstract
Members of the Src family kinases (SFK) can modulate diverse cellular processes, including division, death and survival, but their role in autophagy has been minimally explored. Here, we investigated the roles of Lyn, a SFK, in promoting the survival of human glioblastoma tumor (GBM) cells in vitro and in vivo using lentiviral vector-mediated expression of constitutively-active Lyn (CA-Lyn) or dominant-negative Lyn (DN-Lyn). Expression of either CA-Lyn or DN-Lyn had no effect on the survival of U87 GBM cells grown under nutrient-rich conditions. In contrast, under nutrient-deprived conditions (absence of supplementation with L-glutamine, which is essential for growth of GBM cells, and FBS) CA-Lyn expression enhanced survival and promoted autophagy as well as inhibiting cell death and promoting proliferation. Expression of DN-Lyn promoted cell death. In the nutrient-deprived GBM cells, CA-Lyn expression enhanced AMPK activity and reduced the levels of pS6 kinase whereas DN-Lyn enhanced the levels of pS6 kinase. Similar results were obtained in vitro using another cultured GBM cell line and primary glioma stem cells. On propagation of the transduced GBM cells in the brains of nude mice, the CA-Lyn xenografts formed larger tumors than control cells and autophagosomes were detectable in the tumor cells. The DN-Lyn xenografts formed smaller tumors and contained more apoptotic cells. Our findings suggest that on nutrient deprivation in vitro Lyn acts to enhance the survival of GBM cells by promoting autophagy and proliferation as well as inhibiting cell death, and Lyn promotes the same effects in vivo in xenograft tumors. As the levels of Lyn protein or its activity are elevated in several cancers these findings may be of broad relevance to cancer biology.
Collapse
Affiliation(s)
- Wei Michael Liu
- Department of Cancer Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ping Huang
- Department of Cancer Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Niladri Kar
- Department of Cancer Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Monica Burgett
- Department of Cancer Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Gaelle Muller-Greven
- Department of Cancer Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- School of Biomedical Sciences, Kent State University, Kent, Ohio, United States of America
| | - Amy S. Nowacki
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Clark W. Distelhorst
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Justin D. Lathia
- Department of Stem Cell Biology and Regenerative Medicine, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Jeremy N. Rich
- Department of Stem Cell Biology and Regenerative Medicine, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - John C. Kappes
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Candece L. Gladson
- Department of Cancer Biology, The Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
126
|
Abstract
Tumorigenesis is the process by which normal cells evolve the capacity to evade and overcome the constraints usually placed upon their growth and survival. To ensure the integrity of organs and tissues, the balance of cell proliferation and cell death is tightly maintained. The proteins controlling this balance are either considered oncogenes, which promote tumorigenesis, or tumor suppressors, which prevent tumorigenesis. Phosphoinositide 3-kinase enhancer (PIKE) is a family of GTP-binding proteins that possess anti-apoptotic functions and play an important role in the central nervous system. Notably, accumulating evidence suggests that PIKE is a proto-oncogene involved in tumor progression. The PIKE gene (CENTG1) is amplified in a variety of human cancers, leading to the resistance against apoptosis and the enhancement of invasion. In this review, we will summarize the functions of PIKE proteins in tumorigenesis and discuss their potential implications in cancer therapy.
Collapse
|
127
|
Emerging roles of E3 ubiquitin ligases in autophagy. Trends Biochem Sci 2013; 38:453-60. [PMID: 23870665 DOI: 10.1016/j.tibs.2013.06.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/08/2013] [Revised: 06/10/2013] [Accepted: 06/18/2013] [Indexed: 11/23/2022]
Abstract
Autophagy is an evolutionarily conserved intracellular catabolic process that delivers cytoplasmic components to lysosomes for degradation and recycling. Although originally considered to be a non-selective pathway, it is now recognized that autophagy is involved in selective processes, including the turnover of organelles, removal of protein aggregates, and elimination of intracellular pathogens. This specificity implies that cargo recognition and processing by the autophagy machinery are tightly regulated processes. In support of this, various forms of post-translational modification have been implicated in the regulation of autophagy, one of which is the ubiquitin-proteasome system. Here we review current understanding of the role of ubiquitylation in the control of autophagy.
Collapse
|
128
|
Deegan S, Saveljeva S, Gorman AM, Samali A. Stress-induced self-cannibalism: on the regulation of autophagy by endoplasmic reticulum stress. Cell Mol Life Sci 2013; 70:2425-41. [PMID: 23052213 PMCID: PMC11113399 DOI: 10.1007/s00018-012-1173-4] [Citation(s) in RCA: 217] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/12/2012] [Revised: 09/06/2012] [Accepted: 09/17/2012] [Indexed: 12/26/2022]
Abstract
Macroautophagy (autophagy) is a cellular catabolic process which can be described as a self-cannibalism. It serves as an essential protective response during conditions of endoplasmic reticulum (ER) stress through the bulk removal and degradation of unfolded proteins and damaged organelles; in particular, mitochondria (mitophagy) and ER (reticulophagy). Autophagy is genetically regulated and the autophagic machinery facilitates removal of damaged cell components and proteins; however, if the cell stress is acute or irreversible, cell death ensues. Despite these advances in the field, very little is known about how autophagy is initiated and how the autophagy machinery is transcriptionally regulated in response to ER stress. Some three dozen autophagy genes have been shown to be required for the correct assembly and function of the autophagic machinery; however; very little is known about how these genes are regulated by cellular stress. Here, we will review current knowledge regarding how ER stress and the unfolded protein response (UPR) induce autophagy, including description of the different autophagy-related genes which are regulated by the UPR.
Collapse
Affiliation(s)
- Shane Deegan
- Apoptosis Research Centre, NUI Galway, Galway, Ireland
- School of Natural Sciences, NUI Galway, Galway, Ireland
| | - Svetlana Saveljeva
- Apoptosis Research Centre, NUI Galway, Galway, Ireland
- School of Natural Sciences, NUI Galway, Galway, Ireland
| | - Adrienne M. Gorman
- Apoptosis Research Centre, NUI Galway, Galway, Ireland
- School of Natural Sciences, NUI Galway, Galway, Ireland
| | - Afshin Samali
- Apoptosis Research Centre, NUI Galway, Galway, Ireland
- School of Natural Sciences, NUI Galway, Galway, Ireland
| |
Collapse
|
129
|
Ward KK, Tancioni I, Lawson C, Miller NL, Jean C, Chen XL, Uryu S, Kim J, Tarin D, Stupack DG, Plaxe SC, Schlaepfer DD. Inhibition of focal adhesion kinase (FAK) activity prevents anchorage-independent ovarian carcinoma cell growth and tumor progression. Clin Exp Metastasis 2013; 30:579-94. [PMID: 23275034 PMCID: PMC3622195 DOI: 10.1007/s10585-012-9562-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/24/2012] [Accepted: 12/05/2012] [Indexed: 10/27/2022]
Abstract
Recurrence and spread of ovarian cancer is the 5th leading cause of death for women in the United States. Focal adhesion kinase (FAK) is a cytoplasmic protein-tyrosine kinase located on chromosome 8q24.3 (gene is Ptk2), a site commonly amplified in serous ovarian cancer. Elevated FAK mRNA levels in serous ovarian carcinoma are associated with decreased (logrank P = 0.0007, hazard ratio 1.43) patient overall survival, but how FAK functions in tumor progression remains undefined. We have isolated aggressive ovarian carcinoma cells termed ID8-IP after intraperitoneal (IP) growth of murine ID8 cells in C57Bl6 mice. Upon orthotopic implantation within the peri-ovarian bursa space, ID8-IP cells exhibit greater tumor growth, local and distant metastasis, and elevated numbers of ascites-associated cells compared to parental ID8 cells. ID8-IP cells exhibit enhanced growth under non-adherent conditions with elevated FAK and c-Src tyrosine kinase activation compared to parental ID8 cells. In vitro, the small molecule FAK inhibitor (Pfizer, PF562,271, PF-271) at 0.1 uM selectively prevented anchorage-independent ID8-IP cell growth with the inhibition of FAK tyrosine (Y)397 but not c-Src Y416 phosphorylation. Oral PF-271 administration (30 mg/kg, twice daily) blocked FAK but not c-Src tyrosine phosphorylation in ID8-IP tumors. This was associated with decreased tumor size, prevention of peritoneal metastasis, reduced tumor-associated endothelial cell number, and increased tumor cell-associated apoptosis. FAK knockdown and re-expression assays showed that FAK activity selectively promoted anchorage-independent ID8-IP cell survival. These results support the continued evaluation of FAK inhibitors as a promising clinical treatment for ovarian cancer.
Collapse
Affiliation(s)
- Kristy K. Ward
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Isabelle Tancioni
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Christine Lawson
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Nichol L.G. Miller
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Christine Jean
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Xiao Lei Chen
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Sean Uryu
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Josephine Kim
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - David Tarin
- Department of Pathology, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Dwayne G. Stupack
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - Steven C. Plaxe
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| | - David D. Schlaepfer
- Department of Reproductive Medicine, Moores UCSD Cancer Center, La Jolla, CA 92093
| |
Collapse
|
130
|
Elzinga BM, Nyhan MJ, Crowley LC, O'Donovan TR, Cahill MR, McKenna SL. Induction of autophagy by Imatinib sequesters Bcr-Abl in autophagosomes and down-regulates Bcr-Abl protein. Am J Hematol 2013; 88:455-62. [PMID: 23440701 DOI: 10.1002/ajh.23428] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/14/2013] [Accepted: 02/22/2013] [Indexed: 01/07/2023]
Abstract
Chronic Myeloid Leukemia (CML) is a disease of hematopoietic stem cells which harbor the chimeric gene Bcr-Abl. Expression levels of this constitutively active tyrosine kinase are critical for response to tyrosine kinase inhibitor treatment and also disease progression, yet the regulation of protein stability is poorly understood. We have previously demonstrated that imatinib can induce autophagy in Bcr-Abl expressing cells. Autophagy has been associated with the clearance of large macromolecular signaling complexes and abnormal proteins, however, the contribution of autophagy to the turnover of Bcr-Abl protein in imatinib treated cells is unknown. In this study, we show that following imatinib treatment, Bcr-Abl is sequestered into vesicular structures that co-localize with the autophagy marker LC3 or GABARAP. This association is inhibited by siRNA mediated knockdown of autophagy regulators (Beclin 1/ATG7). Pharmacological inhibition of autophagy also reduced Bcr-Abl/LC3 co-localization in both K562 and CML patient cells. Bcr-Abl protein expression was reduced with imatinib treatment. Inhibition of both autophagy and proteasome activity in imatinib treated cells was required to restore Bcr-Abl protein levels to those of untreated cells. This ability to down-regulate Bcr-Abl protein levels through the induction of autophagy may be an additional and important feature of the activity of imatinib.
Collapse
Affiliation(s)
- Baukje M. Elzinga
- Leslie C. Quick Laboratory; Cork Cancer Research Centre, BioSciences Institute, University College Cork; Cork; Ireland
| | - Michelle J. Nyhan
- Leslie C. Quick Laboratory; Cork Cancer Research Centre, BioSciences Institute, University College Cork; Cork; Ireland
| | - Lisa C. Crowley
- Leslie C. Quick Laboratory; Cork Cancer Research Centre, BioSciences Institute, University College Cork; Cork; Ireland
| | - Tracey R. O'Donovan
- Leslie C. Quick Laboratory; Cork Cancer Research Centre, BioSciences Institute, University College Cork; Cork; Ireland
| | - Mary R. Cahill
- Department of Haematology; Cork University Hospital; Cork; Ireland
| | - Sharon L. McKenna
- Leslie C. Quick Laboratory; Cork Cancer Research Centre, BioSciences Institute, University College Cork; Cork; Ireland
| |
Collapse
|
131
|
Substrate recognition in selective autophagy and the ubiquitin-proteasome system. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:163-81. [PMID: 23545414 DOI: 10.1016/j.bbamcr.2013.03.019] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 12/16/2012] [Revised: 03/15/2013] [Accepted: 03/20/2013] [Indexed: 12/21/2022]
Abstract
Dynamic protein turnover through regulated protein synthesis and degradation ensures cellular growth, proliferation, differentiation and adaptation. Eukaryotic cells utilize two mechanistically distinct but largely complementary systems - the 26S proteasome and the lysosome (or vacuole in yeast and plants) - to effectively target a wide range of proteins for degradation. The concerted action of the ubiquitination machinery and the 26S proteasome ensures the targeted and tightly regulated degradation of a subset of commonly short-lived cellular proteins. Autophagy is a distinct degradation pathway, which transports a highly heterogeneous set of cargos in dedicated vesicles, called autophagosomes, to the lysosome. There the cargo becomes degraded and its molecular building blocks are recycled. While general autophagy randomly engulfs portions of the cytosol, selective autophagy employs dedicated cargo adaptors to specifically enrich the forming autophagosomes for a certain type of cargo as a response to various intra- or extracellular signals. Selective autophagy targets a wide range of cargos including long-lived proteins and protein complexes, organelles, protein aggregates and even intracellular microbes. In this review we summarize available data on cargo recognition mechanisms operating in selective autophagy and the ubiquitin-proteasome system (UPS), and emphasize their differences and common themes. Moreover, we derive general regulatory principles underlying cargo recognition in selective autophagy, and describe the system-wide crosstalk between these two cellular protein degradation systems. This article is part of a Special Issue entitled: Ubiquitin-Proteasome System. Guest Editors: Thomas Sommer and Dieter H. Wolf.
Collapse
|
132
|
Lv J, Tian Y. [Effect of Src tyrosine kinase inhibition on the drug-resistance as well as MDR1 and LRP expression of the human cis-platinum-resistant lung cancer cell line A549/DDP]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2013; 15:501-6. [PMID: 22989452 PMCID: PMC5999856 DOI: 10.3779/j.issn.1009-3419.2012.09.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The aim of this study is to investigate the effect of Src tyrosine kinase inhibition on the drug-resistance as well as the expression of multidrug resistance 1 (MDR1) and lung resistance-related protein (LRP) of the human cis-platinum-resistant lung cancer cell line A549/DDP. METHODS 4-Anilinoquirazoline was used to inhibit Src tyrosine kinase activity in A549/DDP. Western blot analysis was used to detect the Src tyrosine kinase activity. CellTiter-Glo assay was used to detect the drug sensitivity of tumor cells. Flow cytometry was used to detect the intracellular Rh-123 content. Western blot and real-time PCR assay were used to detect the expression of tumor MDR1 and LRP. RESULTS 4-Anilinoquirazoline can down-regulate the cellular Src tyrosine kinase activity in A549/DDP. After treatment with 2.5 μM and 10 μM of 4-anilinoquirazoline, the cells became more sensitive to the drug and the reversal folds (RFs) of tumor cell sensitivity to the drug were 1.59- and 2.10-fold, respectively. The intracellular content of Rh-123 improved by 1.21- and 1.59-fold, respectively. The mRNA levels of MDR1 were 53.8% and 27.5% of the control, respectively. The mRNA level of LRP was 59.3% and 21.4% of the control, respectively. The expression of MDR1 and LRP protein significantly decreased. CONCLUSIONS The inhibition of Src tyrosine kinase activity in A549/DDP cells can reverse multi-drug resistance and increase the sensitivity of the cells to the drug. The mechanism may be related to the down-regulation of cellular MDR1 and LRP.
Collapse
Affiliation(s)
- Jie Lv
- Department of Laboratory Medicine, Rizhao People's Hospital, Rizhao 276826, China.
| | | |
Collapse
|
133
|
Wang BS, Yang Y, Lu HZ, Shang L, Zhang Y, Hao JJ, Shi ZZ, Wang XM, Liu YZ, Zhan QM, Jia XM, Wang MR. Inhibition of atypical protein kinase Cι induces apoptosis through autophagic degradation of β-catenin in esophageal cancer cells. Mol Carcinog 2013; 53:514-25. [PMID: 23359356 DOI: 10.1002/mc.22003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/23/2012] [Accepted: 12/19/2012] [Indexed: 12/21/2022]
Abstract
Atypical protein kinase Cι (PKCι) has been identified as an oncoprotein in esophageal squamous cell carcinomas. However, the mechanisms underlying the role of PKCι in this disease remain unclear. In the present work, we found that inhibition of PKCι expression by RNAi induced apoptosis via the down-regulation of β-catenin in esophageal cancer cells. Furthermore, we found that PKCι regulated β-catenin in an autophagy dependent way. Since down-regulation of β-catenin induced by knockdown of PKCι could be rescued by autophagy inhibition; knockdown of PKCι activated autophagy and promoted the recruitment of β-catenin into autophagosome. These results suggested that PKCι positively regulated β-catenin through negatively regulated autophagy and depletion of PKCι promoted apoptosis via autophagic degradation of β-catenin in esophageal cancer cells. These data provide new insights into PKCι signaling in human cancer.
Collapse
Affiliation(s)
- Bo-Shi Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Barth JMI, Hafen E, Köhler K. The lack of autophagy triggers precocious activation of Notch signaling during Drosophila oogenesis. BMC DEVELOPMENTAL BIOLOGY 2012; 12:35. [PMID: 23217079 PMCID: PMC3564699 DOI: 10.1186/1471-213x-12-35] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 07/26/2012] [Accepted: 11/30/2012] [Indexed: 11/10/2022]
Abstract
BACKGROUND The proper balance of autophagy, a lysosome-mediated degradation process, is indispensable for oogenesis in Drosophila. We recently demonstrated that egg development depends on autophagy in the somatic follicle cells (FC), but not in the germline cells (GCs). However, the lack of autophagy only affects oogenesis when FCs are autophagy-deficient but GCs are wild type, indicating that a dysfunctional signaling between soma and germline may be responsible for the oogenesis defects. Thus, autophagy could play an essential role in modulating signal transduction pathways during egg development. RESULTS Here, we provide further evidence for the necessity of autophagy during oogenesis and demonstrate that autophagy is especially required in subsets of FCs. Generation of autophagy-deficient FCs leads to a wide range of phenotypes that are similar to mutants with defects in the classical cell-cell signaling pathways in the ovary. Interestingly, we observe that loss of autophagy leads to a precocious activation of the Notch pathway in the FCs as monitored by the expression of Cut and Hindsight, two downstream effectors of Notch signaling. CONCLUSION Our findings point to an unexpected function for autophagy in the modulation of the Notch signaling pathway during Drosophila oogenesis and suggest a function for autophagy in proper receptor activation. Egg development is affected by an imbalance of autophagy between signal sending (germline) and signal receiving cell (FC), thus the lack of autophagy in the germline is likely to decrease the amount of active ligand and accordingly compensates for increased signaling in autophagy-defective follicle cells.
Collapse
Affiliation(s)
- Julia M I Barth
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
135
|
Abstract
There have been multiple lines of evidence suggesting that autophagy selectively targets signalling proteins and regulates cancer cell signalling in addition to bulk clearance of long-lived proteins and organelles. Protein degradation through autophagy requires receptor protein LC3B to sequester the substrates into the autophagosome. In the present study, we screened LC3B (light-chain 3B)-binding partners and identified autophagic substrates in cancer cells. With lung cancer NCI-H1975 and oesophageal cancer KYSE30 cell lines as models, we found that VPRBP (viral protein R-binding protein) was a novel LC3B-binding protein through GST (glutathione transferase)–LC3B pull-down combined with LC–MS/MS (liquid chromatography–tandem MS) methods. Co-immunoprecipitation assay showed that VPRBP–LC3/p62 were in the same protein complex as the two cell lines. Induction of autophagy led to a down-regulation of VPRPB, which could be rescued by the inhibition of autophagy degradation by BFA1 (bafilomycin A1) and by the disruption of autophagy through ATG5-knockdown. We also found that induction of autophagy promotes VPRBP–LC3/p62 interaction. Immunohistochemical examination of human NSCLC (non-small cell lung cancer) tissues showed that VPRBP was positively correlated with p62 and negatively correlated with LC3B. Moreover, p62 and VPRBP were associated with poor prognosis in lung ADC (adenocarcinoma) (p62, P=0.019; VPRBP, P=0.005). Patients with low expression of both p62 and VPRBP showed the best prognosis.
Collapse
|
136
|
Affiliation(s)
- Ravi K Amaravadi
- Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsvylania, Philadelphia, PA, USA.
| |
Collapse
|
137
|
Colecchia D, Strambi A, Sanzone S, Iavarone C, Rossi M, Dall'Armi C, Piccioni F, Verrotti di Pianella A, Chiariello M. MAPK15/ERK8 stimulates autophagy by interacting with LC3 and GABARAP proteins. Autophagy 2012; 8:1724-40. [PMID: 22948227 PMCID: PMC3541284 DOI: 10.4161/auto.21857] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022] Open
Abstract
Macroautophagy (hereafter referred to as autophagy) is an evolutionarily conserved catabolic process necessary for normal recycling of cellular constituents and for appropriate response to cellular stress. Although several genes belonging to the core molecular machinery involved in autophagosome formation have been discovered, relatively little is known about the nature of signaling networks controlling autophagy upon intracellular or extracellular stimuli. We discovered ATG8-like proteins (MAP1LC3B, GABARAP and GABARAPL1) as novel interactors of MAPK15/ERK8, a MAP kinase involved in cell proliferation and transformation. Based on the role of these proteins in the autophagic process, we demonstrated that MAPK15 is indeed localized to autophagic compartments and increased, in a kinase-dependent fashion, ATG8-like proteins lipidation, autophagosome formation and SQSTM1 degradation, while decreasing LC3B inhibitory phosphorylation. Interestingly, we also identified a conserved LC3-interacting region (LIR) in MAPK15 responsible for its interaction with ATG8-like proteins, for its localization to autophagic structures and, consequently, for stimulation of the formation of these compartments. Furthermore, we reveal that MAPK15 activity was induced in response to serum and amino-acid starvation and that this stimulus, in turn, required endogenous MAPK15 expression to induce the autophagic process. Altogether, these results suggested a new function for MAPK15 as a regulator of autophagy, acting through interaction with ATG8 family proteins. Also, based on the key role of this process in several human diseases, these results supported the use of this MAP kinase as a potential novel therapeutic target.
Collapse
Affiliation(s)
- David Colecchia
- Istituto Toscano Tumori-Core Research Laboratory, Signal Transduction Unit, AOU Senese, Siena, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Li F, Vierstra RD. Autophagy: a multifaceted intracellular system for bulk and selective recycling. TRENDS IN PLANT SCIENCE 2012; 17:526-37. [PMID: 22694835 DOI: 10.1016/j.tplants.2012.05.006] [Citation(s) in RCA: 263] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/20/2012] [Revised: 04/30/2012] [Accepted: 05/02/2012] [Indexed: 05/18/2023]
Abstract
Plants have evolved sophisticated mechanisms to recycle intracellular constituents. One gaining in appreciation is autophagy, which involves specialized vesicles engulfing and delivering unwanted cytoplasmic material to the vacuole for breakdown. Central to this process is the ubiquitin-fold protein autophagy (ATG)-8, which becomes tethered to the developing autophagic membranes by lipidation. Here, we review data showing that the ATG8 moiety provides a docking site not only for proteins that help shape the enclosing vesicles and promote their fusion with the tonoplast, but also for a host of receptors that recruit appropriate autophagic cargo. The identity of these receptors has dramatically altered the view of autophagy as being a relatively nonspecific mechanism to one that may selectively sequester aggregated proteins, protein complexes, organelles, and even invading pathogens.
Collapse
Affiliation(s)
- Faqiang Li
- Department of Genetics, 425-G Henry Mall, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | |
Collapse
|
139
|
Autophagy and transporter-based multi-drug resistance. Cells 2012; 1:558-75. [PMID: 24710490 PMCID: PMC3901113 DOI: 10.3390/cells1030558] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/17/2012] [Revised: 08/11/2012] [Accepted: 08/16/2012] [Indexed: 12/19/2022] Open
Abstract
All the therapeutic strategies for treating cancers aim at killing the cancer cells via apoptosis (programmed cell death type I). Defective apoptosis endow tumor cells with survival. The cell can respond to such defects with autophagy. Autophagy is a cellular process by which cytoplasmic material is either degraded to maintain homeostasis or recycled for energy and nutrients in starvation. A plethora of evidence has shown that the role of autophagy in tumors is complex. A lot of effort is needed to underline the functional status of autophagy in tumor progression and treatment, and elucidate how to tweak autophagy to treat cancer. Furthermore, during the treatment of cancer, the limitation for the cure rate and survival is the phenomenon of multi drug resistance (MDR). The development of MDR is an intricate process that could be regulated by drug transporters, enzymes, anti-apoptotic genes or DNA repair mechanisms. Reports have shown that autophagy has a dual role in MDR. Furthermore, it has been reported that activation of a death pathway may overcome MDR, thus pointing the importance of other death pathways to regulate tumor cell progression and growth. Therefore, in this review we will discuss the role of autophagy in MDR tumors and a possible link amongst these phenomena.
Collapse
|
140
|
Sandilands E, Serrels B, Wilkinson S, Frame MC. Src-dependent autophagic degradation of Ret in FAK-signalling-defective cancer cells. EMBO Rep 2012; 13:733-40. [PMID: 22732841 PMCID: PMC3410392 DOI: 10.1038/embor.2012.92] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/17/2011] [Revised: 05/31/2012] [Accepted: 05/31/2012] [Indexed: 11/09/2022] Open
Abstract
We have recently described that autophagic targeting of Src maintains cancer cell viability when FAK signalling is defective. Here, we show that the Ret tyrosine kinase is also degraded by autophagy in cancer cells with altered/reduced FAK signalling, preventing its binding to FAK at integrin adhesions. Inhibition of autophagy restores Ret localization to focal adhesions. Importantly, Src kinase activity is required to target Ret to autophagosomes and enhance Ret degradation. Src is thus a general mediator of selective autophagic targeting of adhesion-linked kinases, and Ret a second FAK-binding tyrosine kinase degraded through autophagy in cancer cells under adhesion stress. Src--by controlling not only its own degradation but also that of other FAK-binding partners--allows cancer cell survival, suggesting a new therapeutic strategy.
Collapse
Affiliation(s)
- Emma Sandilands
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XR, UK
| | - Bryan Serrels
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XR, UK
| | - Simon Wilkinson
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XR, UK
| | - Margaret C Frame
- Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XR, UK
| |
Collapse
|
141
|
Zhang G, Liu J, Zhang Y, Qu J, Xu L, Zheng H, Liu Y, Qu X. Cbl-b-dependent degradation of FLIP(L) is involved in ATO-induced autophagy in leukemic K562 and gastric cancer cells. FEBS Lett 2012; 586:3104-10. [PMID: 22884570 DOI: 10.1016/j.febslet.2012.07.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/07/2012] [Revised: 07/21/2012] [Accepted: 07/25/2012] [Indexed: 01/07/2023]
Abstract
Various molecular mechanisms are involved in the efficacy of arsenic trioxide (ATO) against malignant hematologic and some solid tumors. FLICE-like inhibitory protein (FLIP) is an inhibitor of apoptosis mediated by death receptors. In this study, we identified a new link between the down-regulation of cellular FLIP(L) and ATO-induced autophagy. ATO induced the degradation of FLIP(L) in K562 and MGC803 cells, which was mediated by the ubiquitin-proteasome pathway. Moreover, the casitas B-lineage lymphoma-b (Cbl-b) was involved in this process, which interacted with FLIP(L) and promoted proteasomal degradation of FLIP(L). Our findings lead to a better understanding of the mechanism of action of ATO, and suggest that a novel signaling pathway is required for ATO-induced autophagy in K562 and MGC803 cells.
Collapse
Affiliation(s)
- Guodong Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Abstract
Ubiquitination has long been recognised as a key determinator of protein fate by tagging proteins for proteasomal degradation. Most recently, the ability of conjugated ubiquitin chains to confer selectivity to autophagy was demonstrated. Although autophagy was first believed to be a bulk, non-selective 'self-eating' degradative process, the molecular mechanisms of selectivity are now starting to emerge. With the discovery of autophagy receptors - which bind both ubiquitinated substrates and autophagy specific light chain 3 (LC3) modifier on the inner sheath of autophagosomes - a new pathway of selective autophagy is being unravelled. In this review, we focus on the special role of ubiquitin signals and selective autophagy receptors in sorting a variety of autophagic cargos.
Collapse
|
143
|
Denton D, Chang TK, Nicolson S, Shravage B, Simin R, Baehrecke EH, Kumar S. Relationship between growth arrest and autophagy in midgut programmed cell death in Drosophila. Cell Death Differ 2012; 19:1299-307. [PMID: 22555456 DOI: 10.1038/cdd.2012.43] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/18/2023] Open
Abstract
Autophagy has been implicated in both cell survival and programmed cell death (PCD), and this may explain the apparently complex role of this catabolic process in tumourigenesis. Our previous studies have shown that caspases have little influence on Drosophila larval midgut PCD, whereas inhibition of autophagy severely delays midgut removal. To assess upstream signals that regulate autophagy and larval midgut degradation, we have examined the requirement of growth signalling pathways. Inhibition of the class I phosphoinositide-3-kinase (PI3K) pathway prevents midgut growth, whereas ectopic PI3K and Ras signalling results in larger cells with decreased autophagy and delayed midgut degradation. Furthermore, premature induction of autophagy is sufficient to induce early midgut degradation. These data indicate that autophagy and the growth regulatory pathways have an important relationship during midgut PCD. Despite the roles of autophagy in both survival and death, our findings suggest that autophagy induction occurs in response to similar signals in both scenarios.
Collapse
Affiliation(s)
- D Denton
- Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, Australia
| | | | | | | | | | | | | |
Collapse
|
144
|
Abstract
Anchorage-independent growth is the most significant hallmark of cell transformation, which has an intimate relevance to cancer. Anchorage or adhesion physically links cells to the extracellular matrix and allows the transmission of external mechanical cues to intracellular signaling machineries. Transformation involves acquiring the ability to proliferate without requiring mechanically initiated signal transduction, known as mechanotransduction. A number of signaling and cytoskeletal molecules are located at focal adhesions. Src and its related proteins, including p130Cas, localize to adhesion sites, where their functions can be mechanically regulated. In addition, the aberrant activation and expression of Src and p130Cas are linked to transformation and malignancy both in vitro and in vivo. These findings shed light on the importance of mechanotransduction in tumorigenesis and the regulation of cancer progression and also provide insights into the mechanical aspects of cancer signaling.
Collapse
Affiliation(s)
- Hiroyuki Matsui
- Laboratory for Mechanical Medicine, Locomotive Syndrome Research Institute, Nadogaya Hospital, Kashiwa, Japan
| | - Ichiro Harada
- Laboratory for Mechanical Medicine, Locomotive Syndrome Research Institute, Nadogaya Hospital, Kashiwa, Japan
| | - Yasuhiro Sawada
- Laboratory for Mechanical Medicine, Locomotive Syndrome Research Institute, Nadogaya Hospital, Kashiwa, Japan
- Mechanobiology Institute of Singapore, National University of Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
| |
Collapse
|
145
|
|
146
|
Abstract
Autophagy can promote both cancer cell survival and death, and the mechanisms by which it mediates these disparate processes are under intense investigation. Autophagosomes are now shown to entrap and promote degradation of the active tyrosine kinase Src, enabling tumour cell survival. The E3 ubiquitin ligase c-Cbl acts as an autophagosome cargo receptor for Src.
Collapse
|