1
|
Guérin C, Tulasne D. Recording and classifying MET receptor mutations in cancers. eLife 2024; 13:e92762. [PMID: 38652103 PMCID: PMC11042802 DOI: 10.7554/elife.92762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Tyrosine kinase inhibitors (TKI) directed against MET have been recently approved to treat advanced non-small cell lung cancer (NSCLC) harbouring activating MET mutations. This success is the consequence of a long characterization of MET mutations in cancers, which we propose to outline in this review. MET, a receptor tyrosine kinase (RTK), displays in a broad panel of cancers many deregulations liable to promote tumour progression. The first MET mutation was discovered in 1997, in hereditary papillary renal cancer (HPRC), providing the first direct link between MET mutations and cancer development. As in other RTKs, these mutations are located in the kinase domain, leading in most cases to ligand-independent MET activation. In 2014, novel MET mutations were identified in several advanced cancers, including lung cancers. These mutations alter splice sites of exon 14, causing in-frame exon 14 skipping and deletion of a regulatory domain. Because these mutations are not located in the kinase domain, they are original and their mode of action has yet to be fully elucidated. Less than five years after the discovery of such mutations, the efficacy of a MET TKI was evidenced in NSCLC patients displaying MET exon 14 skipping. Yet its use led to a resistance mechanism involving acquisition of novel and already characterized MET mutations. Furthermore, novel somatic MET mutations are constantly being discovered. The challenge is no longer to identify them but to characterize them in order to predict their transforming activity and their sensitivity or resistance to MET TKIs, in order to adapt treatment.
Collapse
Affiliation(s)
- Célia Guérin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
| | - David Tulasne
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 – UMR1277 - Canther – Cancer Heterogeneity, Plasticity and Resistance to TherapiesLilleFrance
| |
Collapse
|
2
|
Imamura R, Sato H, Chih-Cheng Voon D, Shirasaki T, Honda M, Kurachi M, Sakai K, Matsumoto K. Met receptor is essential for MAVS-mediated antiviral innate immunity in epithelial cells independent of its kinase activity. Proc Natl Acad Sci U S A 2023; 120:e2307318120. [PMID: 37748074 PMCID: PMC10556573 DOI: 10.1073/pnas.2307318120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023] Open
Abstract
Epithelial tissue is at the forefront of innate immunity, playing a crucial role in the recognition and elimination of pathogens. Met is a receptor tyrosine kinase that is necessary for epithelial cell survival, proliferation, and regeneration. Here, we showed that Met is essential for the induction of cytokine production by cytosolic nonself double-stranded RNA through retinoic acid-inducible gene-I-like receptors (RLRs) in epithelial cells. Surprisingly, the tyrosine kinase activity of Met was dispensable for promoting cytokine production. Rather, the intracellular carboxy terminus of Met interacted with mitochondrial antiviral-signaling protein (MAVS) in RLR-mediated signaling to directly promote MAVS signalosome formation. These studies revealed a kinase activity-independent function of Met in the promotion of antiviral innate immune responses, defining dual roles of Met in both regeneration and immune responses in the epithelium.
Collapse
Affiliation(s)
- Ryu Imamura
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa920-1192, Japan
- The World Premier International Research Center Initiative (WPI)-Nano Life Science Institute, Kanazawa University, Kanazawa920-1192, Japan
| | - Hiroki Sato
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa920-1192, Japan
| | - Dominic Chih-Cheng Voon
- Innovative Cancer Model Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa920-1192, Japan
| | - Takayoshi Shirasaki
- Department of Clinical Laboratory Medicine, Kanazawa University, Graduate School of Medical Science, Kanazawa920-8641, Japan
| | - Masao Honda
- Department of Clinical Laboratory Medicine, Kanazawa University, Graduate School of Medical Science, Kanazawa920-8641, Japan
- Department of Gastroenterology, Kanazawa University, Graduate School of Medical Science, Kanazawa920-8641, Japan
| | - Makoto Kurachi
- Department of Molecular Genetics, Kanazawa University, Graduate School of Medical Science, Kanazawa920-8640, Japan
| | - Katsuya Sakai
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa920-1192, Japan
- The World Premier International Research Center Initiative (WPI)-Nano Life Science Institute, Kanazawa University, Kanazawa920-1192, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa920-1192, Japan
- The World Premier International Research Center Initiative (WPI)-Nano Life Science Institute, Kanazawa University, Kanazawa920-1192, Japan
| |
Collapse
|
3
|
Fernandes M, Paget S, Kherrouche Z, Truong MJ, Vinchent A, Meneboo JP, Sebda S, Werkmeister E, Descarpentries C, Figeac M, Cortot AB, Tulasne D. Transforming properties of MET receptor exon 14 skipping can be recapitulated by loss of the CBL ubiquitin ligase binding site. FEBS Lett 2023; 597:2301-2315. [PMID: 37468447 DOI: 10.1002/1873-3468.14702] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/07/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023]
Abstract
MET is a receptor tyrosine kinase that is activated in many cancers through various mechanisms. MET exon 14 (Ex14) skipping occurs in 3% of nonsmall cell lung tumors. However, the contribution of the regulatory sites lost upon this skipping, which include a phosphorylated serine (S985) and a binding site for the E3 ubiquitin ligase CBL (Y1003), remains elusive. Sequencing of 2808 lung tumors revealed 71 mutations leading to MET exon 14 skipping and three mutations affecting Y1003 or S985. In addition, MET exon 14 skipping and MET Y1003F induced similar transcriptional programs, increased the activation of downstream signaling pathways, and increased cell mobility. Therefore, the MET Y1003F mutation is able to fully recapitulate responses induced by MET exon 14 skipping, suggesting that loss of the CBL binding site is the main contributor of cell transformation induced by MET Ex14 mutations.
Collapse
Affiliation(s)
- Marie Fernandes
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, France
| | - Sonia Paget
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, France
| | - Zoulika Kherrouche
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, France
| | - Marie-José Truong
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, France
| | - Audrey Vinchent
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, France
| | - Jean-Pascal Meneboo
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, France
| | - Shéhérazade Sebda
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, France
| | - Elisabeth Werkmeister
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US41 - UMS2014 - PLBS, Univ. Lille, France
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL - Center for Infection and Immunity of Lille, Univ. Lille, France
| | | | - Martin Figeac
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, France
| | - Alexis B Cortot
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, France
- Thoracic Oncology Department, CHU Lille, Univ. Lille, France
| | - David Tulasne
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, France
| |
Collapse
|
4
|
When the MET receptor kicks in to resist targeted therapies. Oncogene 2021; 40:4061-4078. [PMID: 34031544 DOI: 10.1038/s41388-021-01835-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 02/04/2023]
Abstract
Although targeted therapies have increased the life expectancy of patients with druggable molecular alterations directly involved in tumor development, the efficacy of these therapies is limited by acquired resistances leading to treatment failure. Most targeted therapies, including ones exploiting therapeutic antibodies and kinase inhibitors, are directed against receptor tyrosine kinases (RTKs) or major signaling hubs. Resistances to these therapies arise when inhibition of these targets is bypassed through activation of alternative signaling pathways. In recent years, activation of the receptor tyrosine kinase MET has been shown to promote resistance to various targeted therapies. This casts MET as important actor in resistance. In this review, we describe how the MET receptor triggers resistance to targeted therapies against RTKs such as EGFR, VEGFR, and HER2 and against signaling hubs such as BRAF. We also describe how MET can be its own resistance factor, as illustrated by on-target resistance of lung tumors harboring activating mutations causing MET exon 14 skipping. Interestingly, investigation of all these situations reveals functional physiological relationships between MET and the target of the therapy to which the cancer becomes resistant, suggesting that resistance stems from preexisting mechanisms. Identification of MET as a resistance factor opens the way to co-treatment strategies that are being tested in current clinical trials.
Collapse
|
5
|
Huang H. Proteolytic Cleavage of Receptor Tyrosine Kinases. Biomolecules 2021; 11:biom11050660. [PMID: 33947097 PMCID: PMC8145142 DOI: 10.3390/biom11050660] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/15/2021] [Accepted: 04/26/2021] [Indexed: 01/18/2023] Open
Abstract
The receptor tyrosine kinases (RTKs) are a large family of cell-surface receptors, which are essential components of signal transduction pathways. There are more than fifty human RTKs that can be grouped into multiple RTK subfamilies. RTKs mediate cellular signaling transduction, and they play important roles in the regulation of numerous cellular processes. The dysregulation of RTK signaling is related to various human diseases, including cancers. The proteolytic cleavage phenomenon has frequently been found among multiple receptor tyrosine kinases. More and more information about proteolytic cleavage in RTKs has been discovered, providing rich insight. In this review, we summarize research about different aspects of RTK cleavage, including its relation to cancer, to better elucidate this phenomenon. This review also presents proteolytic cleavage in various members of the RTKs.
Collapse
Affiliation(s)
- Hao Huang
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; or
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Duplaquet L, Leroy C, Vinchent A, Paget S, Lefebvre J, Vanden Abeele F, Lancel S, Giffard F, Paumelle R, Bidaux G, Heliot L, Poulain L, Furlan A, Tulasne D. Control of cell death/survival balance by the MET dependence receptor. eLife 2020; 9:50041. [PMID: 32091387 PMCID: PMC7039684 DOI: 10.7554/elife.50041] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 02/12/2020] [Indexed: 12/31/2022] Open
Abstract
Control of cell death/survival balance is an important feature to maintain tissue homeostasis. Dependence receptors are able to induce either survival or cell death in presence or absence of their ligand, respectively. However, their precise mechanism of action and their physiological importance are still elusive for most of them including the MET receptor. We evidence that pro-apoptotic fragment generated by caspase cleavage of MET localizes to the mitochondria-associated membrane region. This fragment triggers a calcium transfer from endoplasmic reticulum to mitochondria, which is instrumental for the apoptotic action of the receptor. Knock-in mice bearing a mutation of MET caspase cleavage site highlighted that p40MET production is important for FAS-driven hepatocyte apoptosis, and demonstrate that MET acts as a dependence receptor in vivo. Our data shed light on new signaling mechanisms for dependence receptors’ control of cell survival/death balance, which may offer new clues for the pathophysiology of epithelial structures.
Collapse
Affiliation(s)
- Leslie Duplaquet
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| | - Catherine Leroy
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| | - Audrey Vinchent
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| | - Sonia Paget
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| | - Jonathan Lefebvre
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| | | | - Steve Lancel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, Lille, France
| | - Florence Giffard
- Normandie Université, UNICAEN, INSERM U1086 ANTICIPE, UNICANCER, Cancer Centre F. Baclesse, Caen, France
| | - Réjane Paumelle
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, Lille, France
| | - Gabriel Bidaux
- Univ. Lille, CNRS, UMR8523 - PhLAM - laboratoire de Physique des Lasers, Atomes et Molécules, Lille, France
| | - Laurent Heliot
- Univ. Lille, CNRS, UMR8523 - PhLAM - laboratoire de Physique des Lasers, Atomes et Molécules, Lille, France
| | - Laurent Poulain
- Normandie Université, UNICAEN, INSERM U1086 ANTICIPE, UNICANCER, Cancer Centre F. Baclesse, Caen, France
| | - Alessandro Furlan
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France.,Univ. Lille, CNRS, UMR8523 - PhLAM - laboratoire de Physique des Lasers, Atomes et Molécules, Lille, France
| | - David Tulasne
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, Lille, France
| |
Collapse
|
7
|
Fernandes M, Duplaquet L, Tulasne D. Proteolytic cleavages of MET: the divide-and-conquer strategy of a receptor tyrosine kinase. BMB Rep 2019. [PMID: 30670153 PMCID: PMC6507848 DOI: 10.5483/bmbrep.2019.52.4.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Membrane-anchored full-length MET stimulated by its ligand HGF/SF induces various biological responses, including survival, growth, and invasion. This panel of responses, referred to invasive growth, is required for embryogenesis and tissue regeneration in adults. On the contrary, MET deregulation is associated with tumorigenesis in many kinds of cancer. In addition to its well-documented ligand-stimulated downstream signaling, the receptor can be cleaved by proteases such as secretases, caspases, and calpains. These cleavages are involved either in MET receptor inactivation or, more interestingly, in generating active fragments that can modify cell fate. For instance, MET fragments can promote cell death or invasion. Given a large number of proteases capable of cleaving MET, this receptor appears as a prototype of proteolytic-cleavage-regulated receptor tyrosine kinase. In this review, we describe and discuss the mechanisms and consequences, both physiological and pathological, of MET proteolytic cleavages.
Collapse
Affiliation(s)
- Marie Fernandes
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Target Therapies, F-59000 Lille, France
| | - Leslie Duplaquet
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Target Therapies, F-59000 Lille, France
| | - David Tulasne
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Target Therapies, F-59000 Lille, France
| |
Collapse
|
8
|
M10 peptide attenuates silica-induced pulmonary fibrosis by inhibiting Smad2 phosphorylation. Toxicol Appl Pharmacol 2019; 376:46-57. [DOI: 10.1016/j.taap.2019.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/18/2019] [Accepted: 05/20/2019] [Indexed: 01/06/2023]
|
9
|
Huang X, Gan G, Wang X, Xu T, Xie W. The HGF-MET axis coordinates liver cancer metabolism and autophagy for chemotherapeutic resistance. Autophagy 2019; 15:1258-1279. [PMID: 30786811 PMCID: PMC6613896 DOI: 10.1080/15548627.2019.1580105] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Notwithstanding the numerous drugs available for liver cancer, emerging evidence suggests that chemotherapeutic resistance is a significant issue. HGF and its receptor MET play critical roles in liver carcinogenesis and metastasis, mainly dependent on the activity of receptor tyrosine kinase. However, for unknown reasons, all HGF-MET kinase activity-targeted drugs have failed or have been suspended in clinical trials thus far. Macroautophagy/autophagy is a protective ‘self-eating’ process for resisting metabolic stress by recycling obsolete components, whereas the impact of autophagy-mediated reprogrammed metabolism on therapeutic resistance is largely unclear, especially in liver cancer. In the present study, we first observed that HGF stimulus facilitated the Warburg effect and glutaminolysis to promote biogenesis in multiple liver cancer cells. We then identified the pyruvate dehydrogenase complex (PDHC) and GLS/GLS1 as crucial substrates of HGF-activated MET kinase; MET-mediated phosphorylation inhibits PDHC activity but activates GLS to promote cancer cell metabolism and biogenesis. We further found that the key residues of kinase activity in MET (Y1234/1235) also constitute a conserved LC3-interacting region motif (Y1234-Y1235-x-V1237). Therefore, on inhibiting HGF-mediated MET kinase activation, Y1234/1235-dephosphorylated MET induced autophagy to maintain biogenesis for cancer cell survival. Moreover, we verified that Y1234/1235-dephosphorylated MET correlated with autophagy in clinical liver cancer. Finally, a combination of MET inhibitor and autophagy suppressor significantly improved the therapeutic efficiency of liver cancer in vitro and in mice. Together, our findings reveal an HGF-MET axis-coordinated functional interaction between tyrosine kinase signaling and autophagy, and establish a MET-autophagy double-targeted strategy to overcome chemotherapeutic resistance in liver cancer. Abbreviations: ALDO: aldolase, fructose-bisphosphate; CQ: chloroquine; DLAT/PDCE2: dihydrolipoamide S-acetyltransferase; EMT: epithelial-mesenchymal transition; ENO: enolase; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GLS/GLS1: glutaminase; GLUL/GS: glutamine-ammonia ligase; GPI/PGI: glucose-6-phosphate isomerase; HCC: hepatocellular carcinoma; HGF: hepatocyte growth factor; HK: hexokinase; LDH: lactate dehydrogenase; LIHC: liver hepatocellular carcinoma; LIR: LC3-interacting region; PDH: pyruvate dehydrogenase; PDHA1: pyruvate dehydrogenase E1 alpha 1 subunit; PDHX: pyruvate dehydrogenase complex component X; PFK: phosphofructokinase; PK: pyruvate kinase; RTK: receptor tyrosine kinase; TCGA: The Cancer Genome Atlas
Collapse
Affiliation(s)
- Xing Huang
- a The Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province , First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou , Zhejiang , China.,b The Key Laboratory of Developmental Genes and Human Disease , Institute of Life Sciences, Southeast University , Nanjing , Jiangsu , China.,c The Therapeutic Antibody Research Center of SEU-Alphamab , Southeast University , Nanjing , China
| | - Guangming Gan
- b The Key Laboratory of Developmental Genes and Human Disease , Institute of Life Sciences, Southeast University , Nanjing , Jiangsu , China
| | - Xiaoxiao Wang
- c The Therapeutic Antibody Research Center of SEU-Alphamab , Southeast University , Nanjing , China
| | - Ting Xu
- b The Key Laboratory of Developmental Genes and Human Disease , Institute of Life Sciences, Southeast University , Nanjing , Jiangsu , China.,c The Therapeutic Antibody Research Center of SEU-Alphamab , Southeast University , Nanjing , China
| | - Wei Xie
- b The Key Laboratory of Developmental Genes and Human Disease , Institute of Life Sciences, Southeast University , Nanjing , Jiangsu , China.,c The Therapeutic Antibody Research Center of SEU-Alphamab , Southeast University , Nanjing , China
| |
Collapse
|
10
|
Xie Y, Nurkesh AA, Ibragimova N, Zhanzak Z, Meyerbekova A, Alexeyeva Z, Yesbolatova A, Satayeva M, Mustafa A, Manarbek L, Maipas A, Altaikyzy A, Keneskhanova Z, Akbay B, Chen Z. Systematic analysis of NLMP suggests nuclear localization of RTK/MET kinases resemble cancer cell clearance. J Exp Clin Cancer Res 2019; 38:43. [PMID: 30700325 PMCID: PMC6354337 DOI: 10.1186/s13046-018-1004-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/13/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Some membrane proteins can translocate into the nucleus, defined as nuclear localized membrane proteins (NLMPs), including receptor tyrosine kinases (RTKs). We previously showed that nuclear MET (nMET), a member of RTKs, mediates cancer stem-like cells self-renewal to promote cancer recurrence. However, it is unknown that nMET or mMET, which is the ancestor in the evolution of cancer cell survival and clearance. Here, we aim to study the NLMP functions in cell death, differentiation and survival. METHOD We applied the systematic reanalysis of functional NLMP and clinical investigations of nMET from databases. In addition, we used soft agar assay, immunoblotting, flow cytometry, and immunofluorescence confocal microscopy for examinations of nMET functions including stem-like cell formation, cell signaling, cell cycle regulation, and co-localization with regulators of cell signaling. ShRNA, antibody of recognizing surface membrane MET based treatment were used to downregulate endogenous nMET to uncover its function. RESULTS We predicted and demonstrated that nMET and nEGFR are most likely not ancestors. nMET overexpression induces both cell death and survival with drug resistance and stem cell-like characters. Moreover, the paradoxical function of nMET in both cell death and cell survival is explained by the fact that nMET induces stem cell-like cell growth, DNA damage repair, to evade the drug sensitization for survival of single cells while non-stem cell-like nMET expressing single cells may undergo clearance by cell death through cell cycle arrest induced by p21. CONCLUSION Taken together, our data suggest a link between nuclear RTK and cancer cell evolutionary clearance via cell death, and drug resistance for survival through stemness selection. Targeting evolved nuclear RTKs in cancer stem cells would be a novel avenue for precision cancer therapy.
Collapse
Affiliation(s)
- Yingqiu Xie
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Ayan A. Nurkesh
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Nazgul Ibragimova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhuldyz Zhanzak
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aizhan Meyerbekova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhanna Alexeyeva
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aiya Yesbolatova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Madina Satayeva
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aidana Mustafa
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Limara Manarbek
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Aisulu Maipas
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Akerke Altaikyzy
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhibek Keneskhanova
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Burkitkan Akbay
- Department of Biology, School of Science and Technology, Nazarbayev University, Qabanbay Batyr Ave 53, Astana, 010000 Kazakhstan
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208 USA
| |
Collapse
|
11
|
Greish K, Nehoff H, Bahman F, Pritchard T, Taurin S. Raloxifene nano-micelles effect on triple-negative breast cancer is mediated through estrogen receptor-β and epidermal growth factor receptor. J Drug Target 2019; 27:903-916. [PMID: 30615483 DOI: 10.1080/1061186x.2019.1566341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that differs in progression, recurrence, and prognosis from other forms of breast cancer. The heterogeneity of TNBC has remained a challenge as no targeted therapy is currently available. Previously, we and others have demonstrated that raloxifene, a selective oestrogen receptor modulator, was also acting independently of the oestrogen receptor-α. However, raloxifene is characterised by a low bioavailability in vivo. Thus, we encapsulated raloxifene into a styrene-maleic acid (SMA) micelle to improve its pharmacokinetics. The micellar raloxifene had higher cytotoxicity when compared to the free formulation, promoted a higher cellular uptake and affected critical signalling pathways. Furthermore, SMA-raloxifene reduced TNBC tumour growth more efficiently than free raloxifene. Finally, we showed that this effect was partially mediated through oestrogen receptor-β. In conclusion, we have provided new insight into the role of raloxifene nanoformulation in improving the management of TNBC.
Collapse
Affiliation(s)
- Khaled Greish
- a Department of Molecular Medicine, and Nanomedicine Unit , College of Medicine and Medical Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University , Manama , Kingdom of Bahrain.,b Department of Oncology , Suez Canal University , Ismailia , Egypt
| | - Hayley Nehoff
- c Department of Pharmacology and Toxicology , University of Otago , Dunedin , New Zealand
| | - Fatemah Bahman
- a Department of Molecular Medicine, and Nanomedicine Unit , College of Medicine and Medical Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Tara Pritchard
- d Malaghan Institute of Medical Research , Wellington , New Zealand
| | - Sebastien Taurin
- a Department of Molecular Medicine, and Nanomedicine Unit , College of Medicine and Medical Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University , Manama , Kingdom of Bahrain
| |
Collapse
|
12
|
Huang H. Anaplastic Lymphoma Kinase (ALK) Receptor Tyrosine Kinase: A Catalytic Receptor with Many Faces. Int J Mol Sci 2018; 19:E3448. [PMID: 30400214 PMCID: PMC6274813 DOI: 10.3390/ijms19113448] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 10/24/2018] [Accepted: 10/30/2018] [Indexed: 12/11/2022] Open
Abstract
The anaplastic lymphoma kinase (ALK) receptor is a membrane-bound tyrosine kinase. The pathogenesis of several cancers is closely related to aberrant forms of ALK or aberrant ALK expression, including ALK fusion proteins, ALK-activated point mutations, and ALK amplification. Clinical applications of different ALK inhibitors represent significant progress in targeted therapy. Knowledge of different aspects of ALK biology can provide significant information to further the understanding of this receptor tyrosine kinase. In this mini-review, we briefly summarize different features of ALK. We also summarize some recent research advances on ALK fusion proteins in cancers.
Collapse
Affiliation(s)
- Hao Huang
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Baldacci S, Kherrouche Z, Descarpentries C, Wislez M, Dansin E, Furlan A, Tulasne D, Cortot AB. [MET exon 14 splicing sites mutations: A new therapeutic opportunity in lung cancer]. Rev Mal Respir 2018; 35:796-812. [PMID: 30174236 DOI: 10.1016/j.rmr.2018.01.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 01/08/2018] [Indexed: 01/23/2023]
Abstract
The mutations leading to MET exon 14 skipping represent a new class of molecular alterations described in various cancers. These alterations are observed in 2 to 3 % of cases of non-small cell lung cancer (NSCLC). Several cases of NSCLC carrying such alterations and achieving objective response to MET tyrosine kinase inhibitorshave recently been published. This review summarizes the molecular mechanisms responsible for MET exon 14 skipping as well as the consequences of the loss of this exon on receptor activity. We also describe the clinical characteristics of patients with METΔ14 mutations. Finally, we address the issues related to the detection of these mutations in lung cancer, and the need to anticipate resistance to MET inhibitors.
Collapse
Affiliation(s)
- S Baldacci
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France; Service de pneumologie et oncologie thoracique, OncoLille, université de Lille, CHU de Lille, 59000 Lille, France
| | - Z Kherrouche
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France
| | - C Descarpentries
- Service de biochimie et biologie moléculaire, plateforme de biologie moléculaire des cancers, CHU de Lille, 59000 Lille, France
| | - M Wislez
- Service de pneumologie, hôpital Tenon, AP-HP, 75020 Paris , France
| | - E Dansin
- Département de cancérologie cervico-faciale & thoracique, CLCC Oscar Lambret, 59000 Lille, France
| | - A Furlan
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France; Équipe de biophotonique cellulaire fonctionnelle, université Lille, CNRS UMR 8523 PhLAM, 59000 Lille, France
| | - D Tulasne
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France
| | - A B Cortot
- Université Lille, CNRS, UMR 8161-M3T, mécanismes de tumorigenèse et thérapies ciblées, Institut Pasteur de Lille, 59000 Lille, France; Service de pneumologie et oncologie thoracique, OncoLille, université de Lille, CHU de Lille, 59000 Lille, France.
| |
Collapse
|
14
|
The multiple paths towards MET receptor addiction in cancer. Oncogene 2018; 37:3200-3215. [PMID: 29551767 DOI: 10.1038/s41388-018-0185-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/30/2018] [Accepted: 01/30/2018] [Indexed: 12/14/2022]
Abstract
Targeted therapies against receptor tyrosine kinases (RTKs) are currently used with success on a small proportion of patients displaying clear oncogene activation. Lung cancers with a mutated EGFR provide a good illustration. The efficacy of targeted treatments relies on oncogene addiction, a situation in which the growth or survival of the cancer cells depends on a single deregulated oncogene. MET, a member of the RTK family, is a promising target because it displays many deregulations in a broad panel of cancers. Although clinical trials having evaluated MET inhibitors in large populations have yielded disappointing results, many recent case reports suggest that MET inhibition may be effective in a subset of patients with unambiguous MET activation and thus, most probably, oncogene addiction. Interestingly, preclinical studies have revealed a particularity of MET addiction: it can arise through several mechanisms, and the mechanism involved can differ according to the cancer type. The present review describes the different mechanisms of MET addiction and their consequences for diagnosis and therapeutic strategies. Although in each cancer type MET addiction affects a restricted number of patients, pooling of these patients across all cancer types yields a targetable population liable to benefit from addiction-targeting therapies.
Collapse
|
15
|
MET receptor variant R970C favors calpain-dependent generation of a fragment promoting epithelial cell scattering. Oncotarget 2017; 8:11268-11283. [PMID: 28061464 PMCID: PMC5355264 DOI: 10.18632/oncotarget.14499] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/26/2016] [Indexed: 12/20/2022] Open
Abstract
The receptor tyrosine kinase MET and its ligand, the hepatocyte growth factor, are essential to embryonic development, whereas deregulation of MET signaling is associated with tumorigenesis leading to various cancers, including lung carcinoma. Mutations in the MET kinase domain lead to constitutive kinase activity and are associated with tumorigenesis. In lung cancer, however, some mutations are found in the juxtamembrane domain, and their functional consequences are unknown. Because the juxtamembrane domain of MET is targeted by several proteolytic cleavages, involved in its degradation during cell death or under steady-state conditions, we evaluated the influence of these mutations on the MET proteolytic cleavages. In stably transfected epithelial cells expressing MET, the juxtamembrane mutations R970C, P991S, and T992I were found not to modify the known caspase or presenilin-dependent regulated intramembrane proteolysis. Yet when overexpressed, the R970C variant caused generation of an as yet undescribed 45-kDa fragment (p45 MET). This fragment was found in the confluent lung cancer cell line NCI-H1437 carrying the R970C mutation and at a lesser extent in cell lines expressing WT MET, suggesting that R970C mutation favors this cleavage. Generation of p45 MET required the activity of the calpain proteases, confirming the involvement of proteolysis. Ectopic expression of reconstituted p45 MET in epithelial cell lines favored cell scattering and invasion indicating active role of this fragment in HGF/SF induced responses. Hence, although the juxtamembrane mutations of MET do not affect its known proteolytic cleavages, the R970C MET variant favors calpain dependent proteolytic cleavage in lung cancer cells.
Collapse
|
16
|
Hass R, Jennek S, Yang Y, Friedrich K. c-Met expression and activity in urogenital cancers - novel aspects of signal transduction and medical implications. Cell Commun Signal 2017; 15:10. [PMID: 28212658 PMCID: PMC5316205 DOI: 10.1186/s12964-017-0165-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 02/13/2017] [Indexed: 11/10/2022] Open
Abstract
C-Met is a receptor tyrosine kinase with multiple functions throughout embryonic development, organogenesis and wound healing and is expressed in various epithelia. The ligand of c-Met is Hepatocyte Growth Factor (HGF) which is secreted among others by mesenchymal stroma/stem (MSC) cells. Physiological c-Met functions are centred around processes that underly cellular motility and invasive growth. Aberrant c-Met expression and activity is observed in numerous cancers and makes major contributions to cell malignancy. Importantly, HGF/c-Met signaling is crucial in the context of communication between cancer cells and the the tumor stroma. Here, we review recent findings on roles of dysregulated c-Met in urogenital tumors such as cancers of the urinary bladder, prostate, and ovary. We put emphasis on novel aspects of cancer-associated c-Met expression regulation on both, HGF-dependent and HGF-independent non-canonical mechanisms. Moreover, this review focusses on c-Met-triggered signalling with potential relevance for urogenital oncogenesis, and on strategies to specifically inhibit c-Met activity.
Collapse
Affiliation(s)
- Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Gynecology, Hannover Medical School, Hannover, Germany
| | - Susanne Jennek
- Institute of Biochemistry II, University Hospital Jena, Nonnenplan 2-4, D-07743, Jena, Germany
| | - Yuanyuan Yang
- Biochemistry and Tumor Biology Lab, Department of Gynecology, Hannover Medical School, Hannover, Germany
| | - Karlheinz Friedrich
- Institute of Biochemistry II, University Hospital Jena, Nonnenplan 2-4, D-07743, Jena, Germany.
| |
Collapse
|
17
|
Cortot AB, Kherrouche Z, Descarpentries C, Wislez M, Baldacci S, Furlan A, Tulasne D. Exon 14 Deleted MET Receptor as a New Biomarker and Target in Cancers. J Natl Cancer Inst 2017; 109:2982828. [DOI: 10.1093/jnci/djw262] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 10/04/2016] [Indexed: 01/07/2023] Open
|
18
|
Caspase-mediated proteolysis of the sorting nexin 2 disrupts retromer assembly and potentiates Met/hepatocyte growth factor receptor signaling. Cell Death Discov 2017; 3:16100. [PMID: 28179995 PMCID: PMC5253419 DOI: 10.1038/cddiscovery.2016.100] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 11/23/2016] [Indexed: 12/14/2022] Open
Abstract
The unfolding of apoptosis involves the cleavage of hundreds of proteins by the caspase family of cysteinyl peptidases. Among those substrates are proteins involved in intracellular vesicle trafficking with a net outcome of shutting down the crucial processes governing protein transport to organelles and to the plasma membrane. However, because of the intertwining of receptor trafficking and signaling, cleavage of specific proteins may lead to unintended consequences. Here we show that in apoptosis, sorting nexin 1 and 2 (SNX1 and SNX2), two proteins involved in endosomal sorting, are cleaved by initiator caspases and also by executioner caspase-6 in the case of SNX2. Moreover, SNX1 is cleaved at multiple sites, including following glutamate residues. Cleavage of SNX2 results in a loss of association with the endosome-to-trans-Golgi network transport protein Vps35 and in a delocalization from endosomes of its associated partner Vps26. We also demonstrate that SNX2 depletion causes an increase in hepatocyte growth factor receptor tyrosine phosphorylation and Erk1/2 signaling in cells. Finally, we show that SNX2 mRNA and protein levels are decreased in colorectal carcinoma and that lower SNX2 gene expression correlates with an increase in cancer patient mortality. Our study reveals the importance to characterize the cleavage fragments produced by caspases of specific death substrates given their potential implication in the mechanism of regulation of physiological (signaling/trafficking) pathways or in the dysfunction leading to pathogenesis.
Collapse
|
19
|
Copin MC, Lesaffre M, Berbon M, Doublet L, Leroy C, Tresch E, Porte H, Vicogne J, B Cortot A, Dansin E, Tulasne D. High-MET status in non-small cell lung tumors correlates with receptor phosphorylation but not with the serum level of soluble form. Lung Cancer 2016; 101:59-67. [PMID: 27794409 DOI: 10.1016/j.lungcan.2016.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/01/2016] [Accepted: 09/13/2016] [Indexed: 01/29/2023]
Abstract
OBJECTIVES The receptor tyrosine kinase MET is essential to embryonic development and organ regeneration. Its deregulation is associated with tumorigenesis. While MET gene amplification and mutations leading to MET self-activation concern only a few patients, a high MET level has been found in about half of the non-small cell lung cancers (NSCLCs) tested. How this affects MET activation in tumors is unclear. Also uncertain is the prognostic value, in cancer, of a phenomenon well described in cell models: MET shedding, i.e. its cleavage by membrane proteases leading to release of a soluble fragment into the medium. MATERIALS AND METHODS A prospective cohort of 39 NSCLC patients was constituted at diagnosis or soon after. Normal tissues, tumor tissues, and blood samples were obtained. This allowed, for the same patient, synchronous determination of (i) the MET level in the tumor, (ii) receptor phosphorylation, and (iii) the concentration of soluble MET fragment (sMET) in the serum. RESULTS After confirming the adequacy of an ELISA for measuring the serum level of sMET, we found no correlation between this level and the concentration of MET in tumors, as evaluated by immunohistochemistry and western blotting. Nevertheless, all but one tumor displaying a high MET level also displayed receptor phosphorylation, restricted to a small number of tumor cells. CONCLUSION Our results thus demonstrate that the serum level of sMET is not indicative of the amount of MET present in the tumor cells and cannot be used as a biomarker for therapeutic purposes. However, MET scoring of tumor biopsies could be a first step prior to determination of MET receptor activation in high-MET tumors.
Collapse
Affiliation(s)
- Marie-Christine Copin
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France; Univ. Lille, Institut de Pathologie, CHU Lille, Avenue Oscar Lambret, F-59000 Lille, France
| | - Marie Lesaffre
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Mélanie Berbon
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Louis Doublet
- Département de Cancérologie Générale, CLCC Oscar Lambret, 3 rue Fréderic Combemale, Lille 59020, France
| | - Catherine Leroy
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Emmanuelle Tresch
- Département de Cancérologie Générale, CLCC Oscar Lambret, 3 rue Fréderic Combemale, Lille 59020, France
| | - Henri Porte
- Service de Chirurgie Thoracique, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Jérôme Vicogne
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France
| | - Alexis B Cortot
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France; Univ. Lille, CHU Lille, Thoracic Oncology Department, F-59000 Lille, France
| | - Eric Dansin
- Département de Cancérologie Générale, CLCC Oscar Lambret, 3 rue Fréderic Combemale, Lille 59020, France
| | - David Tulasne
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France.
| |
Collapse
|
20
|
Atanelishvili I, Shirai Y, Akter T, Noguchi A, Ash KT, Misra S, Ghatak S, Silver RM, Bogatkevich GS. D1398G Variant of MET Is Associated with Impaired Signaling of Hepatocyte Growth Factor in Alveolar Epithelial Cells and Lung Fibroblasts. PLoS One 2016; 11:e0162357. [PMID: 27584154 PMCID: PMC5008815 DOI: 10.1371/journal.pone.0162357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/22/2016] [Indexed: 02/07/2023] Open
Abstract
Pulmonary fibrosis represents the terminal stage of a diverse group of lung diseases including scleroderma associated interstitial lung disease. The molecular mechanisms underlying the pathogenesis of lung fibrosis are not well understood and there is a great need for more effective treatment for this lethal disease. We recently discovered a small fragment of hepatocyte growth factor (HGF) receptor MET as a peptide designated “M10,” with strong antifibrotic properties. Furthermore, we showed that aspartic acid at position 1398 of MET is essential for M10 generation. The current study was undertaken to investigate the D1398G variant of MET in which aspartic acid at position 1398 was mutated to glycine resulting in loss of M10. We demonstrate that lung fibroblasts, A549, and primary alveolar epithelial cells (AEC) expressing D1398G MET exhibit reduced auto-phosphorylation on tyrosine residues and reduced activation of Ras and MAPK. HGF treatment of scleroderma lung fibroblasts as well as HGF treatment of TGFβ-treated normal lung fibroblasts transfected with wild type MET is associated with decreased collagen, connective tissue growth factor (CTGF, CCN2) and smooth muscle α-actin (SMA). However, HGF has no such effects in cells transfected with MET D1398G. Cisplatin- and FasL-induced apoptosis is significantly reduced in AEC transfected with MET wild type, but not in AEC transfected with MET D1398G. We conclude that the D1398G variant of MET is associated with compromised phosphorylation and impaired HGF signaling in lung fibroblasts and AEC, two cell types implicated in the pathogenesis of pulmonary fibrosis associated with scleroderma. Ongoing studies will explore the frequency of this variant and its relationship to pulmonary outcomes in scleroderma patients.
Collapse
Affiliation(s)
- Ilia Atanelishvili
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Yuichiro Shirai
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- Department of Allergy and Rheumatology, Nippon Medical School, Tokyo, Japan
| | - Tanjina Akter
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Atsushi Noguchi
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Kurt T. Ash
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Suniti Misra
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Sibnath Ghatak
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Richard M. Silver
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Galina S. Bogatkevich
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
21
|
Atanelishvili I, Shirai Y, Akter T, Buckner T, Noguchi A, Silver RM, Bogatkevich GS. M10, a caspase cleavage product of the hepatocyte growth factor receptor, interacts with Smad2 and demonstrates antifibrotic properties in vitro and in vivo. Transl Res 2016; 170:99-111. [PMID: 26772959 PMCID: PMC4789156 DOI: 10.1016/j.trsl.2015.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/13/2015] [Accepted: 12/16/2015] [Indexed: 01/07/2023]
Abstract
Hepatocyte growth factor receptor, also known as cellular mesenchymal-epithelial transition factor (c-MET, MET), is an important antifibrotic molecule that protects various tissues, including lung, from injury and fibrosis. The intracellular cytoplasmic tail of MET contains a caspase-3 recognition motif "DEVD-T" that on cleavage by caspase-3 generates a 10-amino acid peptide, TRPASFWETS, designated as "M10". M10 contains at its N-terminus the uncharged amino acid proline (P) directly after a cationic amino acid arginine (R) which favors the transport of the peptide through membranes. M10, when added to cell culture medium, remains in the cytoplasm and nuclei of cells for up to 24 hours. M10 effectively decreases collagen in both scleroderma and TGFβ-stimulated normal lung and skin fibroblasts. M10 interacts with the Mad Homology 2 domain of Smad2 and inhibits TGFβ-induced Smad2 phosphorylation, suggesting that the antifibrotic effects of M10 are mediated in part by counteracting Smad-dependent fibrogenic pathways. In the bleomycin murine model of pulmonary fibrosis, M10 noticeably reduced lung inflammation and fibrosis. Ashcroft fibrosis scores and lung collagen content were significantly lower in bleomycin-treated mice receiving M10 as compared with bleomycin-treated mice receiving scrambled peptide. We conclude that M10 peptide interacts with Smad2 and demonstrates strong antifibrotic effects in vitro and in vivo in an animal model of lung fibrosis and should be considered as a potential therapeutic agent for systemic sclerosis and other fibrosing diseases.
Collapse
Affiliation(s)
- Ilia Atanelishvili
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Yuichiro Shirai
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; Department of Allergy and Rheumatology, Nippon Medical School, Tokyo, Japan
| | - Tanjina Akter
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Taylor Buckner
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; South Carolina Governor's School for Science & Mathematics; Honors College at the College of Charleston, USA
| | - Atsushi Noguchi
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Richard M Silver
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Galina S Bogatkevich
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
22
|
Leroy C, Belkina NV, Long T, Deruy E, Dissous C, Shaw S, Tulasne D. Caspase Cleavages of the Lymphocyte-oriented Kinase Prevent Ezrin, Radixin, and Moesin Phosphorylation during Apoptosis. J Biol Chem 2016; 291:10148-61. [PMID: 26945071 DOI: 10.1074/jbc.m116.721365] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Indexed: 11/06/2022] Open
Abstract
The lymphocyte-oriented kinase (LOK), also called serine threonine kinase 10 (STK10), is synthesized mainly in lymphocytes. It is involved in lymphocyte migration and polarization and can phosphorylate ezrin, radixin, and moesin (the ERM proteins). In a T lymphocyte cell line and in purified human lymphocytes, we found LOK to be cleaved by caspases during apoptosis. The first cleavage occurs at aspartic residue 332, located between the kinase domain and the coiled-coil regulation domain. This cleavage generates an N-terminal fragment, p50 N-LOK, containing the kinase domain and a C-terminal fragment, which is further cleaved during apoptosis. Although these cleavages preserve the entire kinase domain, p50 N-LOK displays no kinase activity. In apoptotic lymphocytes, caspase cleavages of LOK are concomitant with a decrease in ERM phosphorylation. When non-apoptotic lymphocytes from mice with homozygous and heterozygous LOK knockout were compared, the latter showed a higher level of ERM phosphorylation, but when apoptosis was induced, LOK(-/-) and LOK(+/-) lymphocytes showed the same low level, confirming in vivo that LOK-induced ERM phosphorylation is prevented during lymphocyte apoptosis. Our results demonstrate that cleavage of LOK during apoptosis abolishes its kinase activity, causing a decrease in ERM phosphorylation, crucial to the role of the ERM proteins in linking the plasma membrane to actin filaments.
Collapse
Affiliation(s)
- Catherine Leroy
- From the University of Lille, CNRS, Institut Pasteur de Lille, Unité Mixte de Recherche (UMR) 8161, Mechanisms of Tumorigenesis and Target Therapies, 59000 Lille, France
| | | | - Thavy Long
- the University of Lille, CNRS, INSERM, Centre Hospitalier Régional Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR 8204, Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Emeric Deruy
- the University of Lille, CNRS, INSERM, Centre Hospitalier Régional Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR 8204, Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Colette Dissous
- the University of Lille, CNRS, INSERM, Centre Hospitalier Régional Universitaire de Lille, Institut Pasteur de Lille, U1019-UMR 8204, Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Stephen Shaw
- Experimental Immunology Branch Branches, NCI/National Institutes of Health, Bethesda, Maryland 20892, and
| | - David Tulasne
- From the University of Lille, CNRS, Institut Pasteur de Lille, Unité Mixte de Recherche (UMR) 8161, Mechanisms of Tumorigenesis and Target Therapies, 59000 Lille, France
| |
Collapse
|
23
|
Lefebvre J, Clarkson M, Massa F, Bradford ST, Charlet A, Buske F, Lacas-Gervais S, Schulz H, Gimpel C, Hata Y, Schaefer F, Schedl A. Alternatively spliced isoforms of WT1 control podocyte-specific gene expression. Kidney Int 2015; 88:321-31. [DOI: 10.1038/ki.2015.140] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 03/26/2015] [Accepted: 03/26/2015] [Indexed: 01/26/2023]
|
24
|
Necrosis- and apoptosis-related Met cleavages have divergent functional consequences. Cell Death Dis 2015; 6:e1769. [PMID: 25996296 PMCID: PMC4669710 DOI: 10.1038/cddis.2015.132] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/09/2015] [Accepted: 04/14/2015] [Indexed: 01/06/2023]
Abstract
Upon activation by its ligand hepatocyte growth factor/scatter factor, the receptor tyrosine kinase Met promotes survival, proliferation, and migration of epithelial cells during embryogenesis. Deregulated Met signaling can also promote cancer progression and metastasis. Met belongs to the functional family of dependence receptors whose activity switches from pro-survival to pro-apoptotic during apoptosis upon caspase cleavage. Although apoptosis resistance is a hallmark of cancer cells, some remain sensitive to other cell death processes, including necrosis induced by calcium stress. The role and fate of Met during necrotic cell death are unknown. Following treatment with calcium ionophores, cell lines and primary cells undergo necrosis, and the full-length Met receptor is efficiently degraded. This degradation is achieved by double cleavage of Met in its extracellular domain by a metalloprotease of the A disintegrin and metalloproteinase (ADAM) family and in its intracellular domain by calpains (calcium-dependent proteases). These cleavages separate the Met extracellular region from its kinase domain, thus preventing Met activity and its potential pro-survival activity. Although the intracellular fragment is very similar to the fragment generated by caspases, it displays no pro-apoptotic property, likely because of the presence of the last few amino acids of Met, known to inhibit this pro-apoptotic function. The fragments identified here are observed in lung tumors overexpressing the Met receptor, along with fragments previously identified, suggesting that proteolytic cleavages of Met are involved in its degradation in tumor tissues. Thus, Met is a modulator of necrosis, able to protect cells when activated by its ligand but efficiently degraded by proteolysis when this process is engaged.
Collapse
|
25
|
Grancara S, Zonta F, Ohkubo S, Brunati AM, Agostinelli E, Toninello A. Pathophysiological implications of mitochondrial oxidative stress mediated by mitochondriotropic agents and polyamines: the role of tyrosine phosphorylation. Amino Acids 2015; 47:869-83. [PMID: 25792113 DOI: 10.1007/s00726-015-1964-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/11/2015] [Indexed: 12/23/2022]
Abstract
Mitochondria, once merely considered as the "powerhouse" of cells, as they generate more than 90 % of cellular ATP, are now known to play a central role in many metabolic processes, including oxidative stress and apoptosis. More than 40 known human diseases are the result of excessive production of reactive oxygen species (ROS), bioenergetic collapse and dysregulated apoptosis. Mitochondria are the main source of ROS in cells, due to the activity of the respiratory chain. In normal physiological conditions, ROS generation is limited by the anti-oxidant enzymatic systems in mitochondria. However, disregulation of the activity of these enzymes or interaction of respiratory complexes with mitochondriotropic agents may lead to a rise in ROS concentrations, resulting in oxidative stress, mitochondrial permeability transition (MPT) induction and triggering of the apoptotic pathway. ROS concentration is also increased by the activity of amine oxidases located inside and outside mitochondria, with oxidation of biogenic amines and polyamines. However, it should also be recalled that, depending on its concentration, the polyamine spermine can also protect against stress caused by ROS scavenging. In higher organisms, cell signaling pathways are the main regulators in energy production, since they act at the level of mitochondrial oxidative phosphorylation and participate in the induction of the MPT. Thus, respiratory complexes, ATP synthase and transition pore components are the targets of tyrosine kinases and phosphatases. Increased ROS may also regulate the tyrosine phosphorylation of target proteins by activating Src kinases or phosphatases, preventing or inducing a number of pathological states.
Collapse
Affiliation(s)
- Silvia Grancara
- Department of Biomedical Sciences, University of Padova, Viale U. Bassi 58B, 35131, Padua, Italy
| | | | | | | | | | | |
Collapse
|
26
|
Jia J, Furlan A, Gonzalez-Hilarion S, Leroy C, Gruenert DC, Tulasne D, Lejeune F. Caspases shutdown nonsense-mediated mRNA decay during apoptosis. Cell Death Differ 2015; 22:1754-63. [PMID: 25744026 DOI: 10.1038/cdd.2015.18] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 01/30/2015] [Accepted: 02/02/2015] [Indexed: 01/18/2023] Open
Abstract
Nonsense-mediated mRNA decay (NMD) is an mRNA surveillance mechanism that plays integral roles in eliminating mRNAs with premature termination codons to prevent the synthesis of truncated proteins that could be pathogenic. One response to the accumulation of detrimental proteins is apoptosis, which involves the activation of enzymatic pathways leading to protein and nucleic acid cleavage and culminating in cell death. It is not clear whether NMD is required to ensure the accurate expression of apoptosis genes or is no longer necessary since cytotoxic proteins are not an issue during cell death. The present study shows that caspases cleave the two NMD factors UPF1 and UPF2 during apoptosis impairing NMD. Our results demonstrate a new regulatory pathway for NMD that occurs during apoptosis and provide evidence for role of the UPF cleaved fragments in apoptosis and NMD inhibition.
Collapse
Affiliation(s)
- J Jia
- Université de Lille, FRE 3642, Lille, France.,CNRS UMR 8161, Institut de Biologie de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - A Furlan
- Université de Lille, FRE 3642, Lille, France.,CNRS UMR 8161, Institut de Biologie de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - S Gonzalez-Hilarion
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, 25 rue du Dr Roux, Paris, France
| | - C Leroy
- Université de Lille, FRE 3642, Lille, France.,CNRS UMR 8161, Institut de Biologie de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - D C Gruenert
- Department of Otolaryngology-Head and Neck Surgery, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Helen Diller Family Comprehensive Cancer Center, Institute for Human Genetics, Cardiovascular Research Institute, University of California, San Francisco, CA, USA.,Department of Pediatrics, University of Vermont College of Medicine, Burlington, VT, USA
| | - D Tulasne
- Université de Lille, FRE 3642, Lille, France.,CNRS UMR 8161, Institut de Biologie de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - F Lejeune
- Université de Lille, FRE 3642, Lille, France.,CNRS UMR 8161, Institut de Biologie de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| |
Collapse
|
27
|
Abstract
The research on colorectal cancer (CRC) biology has been leading the oncology field since the early 1990s. The search for genetic alterations has allowed the identification of the main tumour suppressors or oncogenes. Recent work obtained in CRC has unexpectedly proposed the existence of novel category of tumour suppressors, the so-called 'dependence receptors'. These transmembrane receptors behave as Dr Jekyll and Mr Hyde with two opposite sides: they induce a positive signalling (survival, proliferation, differentiation) in presence of their ligand, but are not inactive in the absence of their ligand and rather trigger apoptosis when unbound. This trait confers them a conditional tumour suppressor activity: they eliminate cells that grow abnormally in an environment offering a limited quantity of ligand. This review will describe how receptors such as deleted in colorectal carcinoma (DCC), uncoordinated 5 (UNC5), rearranged during transfection (RET) or TrkC constrain CRC progression and how this dependence receptor paradigm may open up therapeutical perspectives.
Collapse
Affiliation(s)
- Patrick Mehlen
- Apoptosis, Cancer and Development Laboratory- Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Servane Tauszig-Delamasure
- Apoptosis, Cancer and Development Laboratory- Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| |
Collapse
|
28
|
Furlan A, Tulasne D. How does met regulate the survival/apoptosis balance? Hepatology 2014; 60:1108-9. [PMID: 24347525 DOI: 10.1002/hep.26969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 11/20/2013] [Accepted: 11/27/2013] [Indexed: 01/01/2023]
Affiliation(s)
- Alessandro Furlan
- CNRS UMR 8161, Institut de Biologie de Lille, Institut Pasteur de Lille, Université de Lille 1, Université de Lille 2, IFR142, SIRIC OncoLille, Lille, France
| | | |
Collapse
|
29
|
Pro-survival role of MITF in melanoma. J Invest Dermatol 2014; 135:352-358. [PMID: 25142731 DOI: 10.1038/jid.2014.319] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/16/2014] [Accepted: 07/18/2014] [Indexed: 01/09/2023]
Abstract
Melanoma is a therapy-resistant skin cancer due to numerous mechanisms supporting cell survival. Although components of melanoma cytoprotective mechanisms are overexpressed in many types of tumors, some of their regulators are characteristic for melanoma. Several genes mediating pro-survival functions have been identified as direct targets of microphthalmia-associated transcription factor (MITF), a melanocyte-specific modulator also recognized as a lineage addiction oncogene in melanoma. BRAF(V600E) and other proteins deregulated in melanoma influence MITF expression and activity, or they are the partners of MITF in melanoma response to radiotherapy and chemotherapeutics. In this review, the pro-survival activity of MITF is discussed.
Collapse
|