1
|
Rosell R, Codony-Servat J, González J, Santarpia M, Jain A, Shivamallu C, Wang Y, Giménez-Capitán A, Molina-Vila MA, Nilsson J, González-Cao M. KRAS G12C-mutant driven non-small cell lung cancer (NSCLC). Crit Rev Oncol Hematol 2024; 195:104228. [PMID: 38072173 DOI: 10.1016/j.critrevonc.2023.104228] [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: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 02/20/2024] Open
Abstract
KRAS G12C mutations in non-small cell lung cancer (NSCLC) partially respond to KRAS G12C covalent inhibitors. However, early adaptive resistance occurs due to rewiring of signaling pathways, activating receptor tyrosine kinases, primarily EGFR, but also MET and ligands. Evidence indicates that treatment with KRAS G12C inhibitors (sotorasib) triggers the MRAS:SHOC2:PP1C trimeric complex. Activation of MRAS occurs from alterations in the Scribble and Hippo-dependent pathways, leading to YAP activation. Other mechanisms that involve STAT3 signaling are intertwined with the activation of MRAS. The high-resolution MRAS:SHOC2:PP1C crystallization structure allows in silico analysis for drug development. Activation of MRAS:SHOC2:PP1C is primarily Scribble-driven and downregulated by HUWE1. The reactivation of the MRAS complex is carried out by valosin containing protein (VCP). Exploring these pathways as therapeutic targets and their impact on different chemotherapeutic agents (carboplatin, paclitaxel) is crucial. Comutations in STK11/LKB1 often co-occur with KRAS G12C, jeopardizing the effect of immune checkpoint (anti-PD1/PDL1) inhibitors.
Collapse
Affiliation(s)
- Rafael Rosell
- Germans Trias i Pujol Research Institute, Badalona (IGTP), Spain; IOR, Hospital Quiron-Dexeus, Barcelona, Spain.
| | | | - Jessica González
- Germans Trias i Pujol Research Institute, Badalona (IGTP), Spain
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Italy
| | - Anisha Jain
- Department of Microbiology, JSS Academy of Higher Education & Research, Mysuru, India
| | - Chandan Shivamallu
- Department of Biotechnology & Bioinformatics, JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| | - Yu Wang
- Genfleet Therapeutics, Shanghai, China
| | | | | | - Jonas Nilsson
- Department Radiation Sciences, Oncology, Umeå University, Sweden
| | | |
Collapse
|
2
|
Abou Hammoud A, Giraud J, Gauthereau X, Blanchard C, Daburon S, Zese M, Molina-Castro S, Dubus P, Varon C, Boeuf H. The "StemDif Sensor Test": A Straightforward, Non-Invasive Assay to Characterize the Secreted Stemness and/or Differentiation Activities of Tumor-Derived Cancer Cell Lines. Biomedicines 2023; 11:3293. [PMID: 38137514 PMCID: PMC10741605 DOI: 10.3390/biomedicines11123293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer stem cells are a subpopulation of tumor cells characterized by their ability to self-renew, induce tumors upon engraftment in animals and exhibit strong resistance to chemotherapy and radiotherapy. These cells exhibit numerous characteristics in common with embryonic stem cells, expressing some of their markers, typically absent in non-pathological adult differentiated cells. The aim of this study was to investigate the potential of conditioned media from cancer stem cells to modulate the fate of Leukemia Inhibitory Factor (LIF)-dependent murine embryonic stem cells (mESCs) as a way to obtain a direct readout of the secretome of cancer cells. A functional assay, "the StemDif sensor test", was developed with two types of cancer stem cells derived from grade IV glioblastoma (adult and pediatric) or from gastric adenocarcinoma. We show that conditioned media from the selection of adult but not pediatric Glioma-Inducing Cells (GICs) maintain mESCs' pluripotency in correlation with LIF secretion and activation of STAT3 protein. In contrast, conditioned media from gastric adenocarcinoma cells display LIF-independent stemness and differentiation activities on mESC. Our test stands out for its user-friendly procedures, affordability and straightforward output, positioning it as a pioneering tool for in-depth exploration of cancer stem cell secretome characteristics.
Collapse
Affiliation(s)
- Aya Abou Hammoud
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (A.A.H.); (C.B.); (M.Z.)
- Univ. Bordeaux, INSERM, BRIC-MIRCADE Team, U1312, F-33000 Bordeaux, France
- Univ. Bordeaux, INSERM, BRIC, U1312, F-33000 Bordeaux, France; (J.G.); (S.M.-C.); (P.D.); (C.V.)
| | - Julie Giraud
- Univ. Bordeaux, INSERM, BRIC, U1312, F-33000 Bordeaux, France; (J.G.); (S.M.-C.); (P.D.); (C.V.)
- Univ. Bordeaux, CNRS, ImmunoConcEpT, U5164, F-33000 Bordeaux, France;
| | - Xavier Gauthereau
- Univ. Bordeaux, CNRS, ImmunoConcEpT, U5164, F-33000 Bordeaux, France;
| | - Camille Blanchard
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (A.A.H.); (C.B.); (M.Z.)
| | | | - Marco Zese
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (A.A.H.); (C.B.); (M.Z.)
| | - Silvia Molina-Castro
- Univ. Bordeaux, INSERM, BRIC, U1312, F-33000 Bordeaux, France; (J.G.); (S.M.-C.); (P.D.); (C.V.)
| | - Pierre Dubus
- Univ. Bordeaux, INSERM, BRIC, U1312, F-33000 Bordeaux, France; (J.G.); (S.M.-C.); (P.D.); (C.V.)
| | - Christine Varon
- Univ. Bordeaux, INSERM, BRIC, U1312, F-33000 Bordeaux, France; (J.G.); (S.M.-C.); (P.D.); (C.V.)
| | - Helene Boeuf
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000 Bordeaux, France; (A.A.H.); (C.B.); (M.Z.)
| |
Collapse
|
3
|
Vaziri N, Shariati L, Javanmard SH. Leukemia inhibitory factor: A main controller of breast cancer. J Biosci 2020. [DOI: 10.1007/s12038-020-00115-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
4
|
Leukaemia Inhibitory Factor (LIF) Inhibits Cancer Stem Cells Tumorigenic Properties through Hippo Kinases Activation in Gastric Cancer. Cancers (Basel) 2020; 12:cancers12082011. [PMID: 32707998 PMCID: PMC7464447 DOI: 10.3390/cancers12082011] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/30/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) present chemo-resistance mechanisms contributing to tumour maintenance and recurrence, making their targeting of utmost importance in gastric cancer (GC) therapy. The Hippo pathway has been implicated in gastric CSC properties and was shown to be regulated by leukaemia inhibitory factor receptor (LIFR) and its ligand LIF in breast cancer. This study aimed to determine LIF’s effect on CSC properties in GC cell lines and patient-derived xenograft (PDX) cells, which remains unexplored. LIF’s treatment effect on CSC markers expression and tumoursphere formation was evaluated. The Hippo kinase inhibitor XMU-MP-1 and/or the JAK1 inhibitor Ruxolitinib were used to determine Hippo and canonical JAK/STAT pathway involvement in gastric CSCs’ response to LIF. Results indicate that LIF decreased tumorigenic and chemo-resistant CSCs, in both GC cell lines and PDX cells. In addition, LIF increased activation of LATS1/2 Hippo kinases, thereby decreasing downstream YAP/TAZ nuclear accumulation and TEAD transcriptional activity. LIF’s anti-CSC effect was reversed by XMU-MP-1 but not by Ruxolitinib treatment, highlighting the opposite effects of these two pathways downstream LIFR. In conclusion, LIF displays anti-CSC properties in GC, through Hippo kinases activation, and could in fine constitute a new CSCs-targeting strategy to help decrease relapse cases and bad prognosis in GC.
Collapse
|
5
|
Ai LQY, Yuan RD, Chen X, Liu YJ, Liu WY, Zhu JY, Zhang Z, Yan J, Chen CL, Lin S, Ye J. Retinal blood vessel-origin yes-associated protein (YAP) governs astrocytic maturation via leukaemia inhibitory factor (LIF). Cell Prolif 2020; 53:e12757. [PMID: 31916327 PMCID: PMC7046482 DOI: 10.1111/cpr.12757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/26/2019] [Accepted: 12/16/2019] [Indexed: 12/31/2022] Open
Abstract
Objectives To testify that endothelial cells (ECs) induce astrocyte maturation by leukaemia inhibitory factor (LIF) secretion. Materials and Methods In vivo experiments, mice bearing floxed alleles of YAP were crossed with mice expressing a Cre recombinase driven by the endothelial Tek promoter (Tek‐Cre) to finally obtain the following three genotypes: YAPf/f, Tek‐Cre; YAPf/w, Tek‐Cre; and YAPf/f. Retinal vascularization and astrocyte network were evaluated by whole‐mount fluorescence and Western blotting. In vitro, experiments were performed in an astrocyte and human microvascular endothelial cell (HMEC‐1) coculture model to analyse the mechanisms underlying the effect of endothelial YAP on astrocytes. Results In vivo, YAPf/f;Tek‐Cre mice showed delayed angiogenesis, sparse vessels and decreased glial fibrillary acidic protein (GFAP)+ astrocytes but aberrant growth of endothelial networks and immature astrocytes (platelet‐derived growth factor A, PDGFRA+ astrocytes) overgrowth. In vitro, Yap deletion attenuated the LIF release that delayed the maturation of retinal astrocyte which was consistent with the results of HMEC‐1—astrocyte coculture. The effect of YAP overexpression on LIF‐LIFR axis in HMEC‐1 interferes the GFAP expression of astrocyte. In contrast, LIF protein rescues the astrocytic GFAP expression when EC YAP was inhibited by siRNAs. Conclusions We show that EC yes‐associated protein (YAP) is not only a critical coactivator of Hippo signalling in retinal vessel development but also plays an essential role in retinal astrocyte maturation by regulating LIF production.
Collapse
Affiliation(s)
- Li-Qian-Yu Ai
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Rong-Di Yuan
- Department of Ophthalmology, XinQiao Hospital, Army Medical University, Chongqing, China
| | - Xi Chen
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Yun-Jia Liu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Wen-Yi Liu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jing-Yi Zhu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Zhou Zhang
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jun Yan
- Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Chun-Lin Chen
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Sen Lin
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jian Ye
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| |
Collapse
|
6
|
Liu SC, Hsu T, Chang YS, Chung AK, Jiang SS, OuYang CN, Yuh CH, Hsueh C, Liu YP, Tsang NM. Cytoplasmic LIF reprograms invasive mode to enhance NPC dissemination through modulating YAP1-FAK/PXN signaling. Nat Commun 2018; 9:5105. [PMID: 30504771 PMCID: PMC6269507 DOI: 10.1038/s41467-018-07660-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 11/13/2018] [Indexed: 01/07/2023] Open
Abstract
Metastasis remains a clinically unsolved issue in nasopharyngeal carcinoma. Here, we report that higher levels of cytoplasmic leukemia inhibitory factor (LIF) and LIF receptor are correlated with poorer metastasis/recurrence-free survival. Further, single nucleotide variations and signal peptide mutation of LIF are identified in NPC. Cytoplasmic LIF reprograms the invasive mode from collective to mesenchymal migration via acquisition of EMT and invadopodia-associated characteristics. Higher cytoplasmic LIF enhances cancer vascular dissemination and local invasion mechanistically through modulation of YAP1-FAK/PXN signaling. Immunohistochemical analyses of NPC biopsies reveal a positive correlation of cytoplasmic LIF expression with focal adhesion kinases. Pharmaceutical intervention with AZD0530 markedly reverses LIF-mediated cancer dissemination and local invasion through promotion of cytoplasmic accumulation of YAP1 and suppression of focal adhesion kinases. Given the significant role of LIF/YAP1-focal adhesion signaling in cancer dissemination, targeting of this pathway presents a promising opportunity to block metastasis. Molecular pathways regulating nasopharyngeal carcinoma (NPC) metastasis are unclear. Here they report higher levels of cytoplasmic leukemia inhibitory factor (cLIF) and LIF receptor (LIFR) to correlate with higher metastasis in NPC patients, and show cLIF to promote NPC metastasis and vascular dissemination via the YAP1-FAK/PXN axis.
Collapse
Affiliation(s)
- Shu-Chen Liu
- Department of Biomedical Sciences and Engineering, National Central University, 300, Zhongda Rd., Jhongli Dist., 32001, Taoyuan City, Taiwan.
| | - Tien Hsu
- Department of Biomedical Sciences and Engineering, National Central University, 300, Zhongda Rd., Jhongli Dist., 32001, Taoyuan City, Taiwan
| | - Yu-Sun Chang
- Molecular Medicine Research Center, Chang Gung University, 259, Wenhua 1st Rd., Guishan Dist., 33302, Taoyuan City, Taiwan
| | - An-Ko Chung
- Graduate Institute of Biomedical Sciences, Chang Gung University, 259, Wenhua 1st Rd., Guishan Dist., 33302, Taoyuan City, Taiwan
| | - Shih Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Rd., Zhunan, 35053, Miaoli County, Taiwan
| | - Chun-Nan OuYang
- Molecular Medicine Research Center, Chang Gung University, 259, Wenhua 1st Rd., Guishan Dist., 33302, Taoyuan City, Taiwan
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, 35 Keyan Rd., Zhunan, 35053, Miaoli County, Taiwan
| | - Chuen Hsueh
- Department of Pathology, Chang Gung Memorial Hospital at Lin-Kou, 5 Fuxing St., Guishan Dist., 333, Taoyuan City, Taiwan
| | - Ya-Ping Liu
- Pathology Core of the Molecular Medicine Research Center, Chang Gung University, 5, Fuxing St., Guishan Dist., 333, Taoyuan City, Taiwan
| | - Ngan-Ming Tsang
- Department of Radiation Oncology, Chang Gung Memorial Hospital and University at Lin-Kou, 5, Fuxing St., Guishan Dist., 333, Taoyuan City, Taiwan.
| |
Collapse
|
7
|
Lei C, Lv S, Wang H, Liu C, Zhai Q, Wang S, Cai G, Lu D, Sun Z, Wei Q. Leukemia Inhibitory Factor Receptor Suppresses the Metastasis of Clear Cell Renal Cell Carcinoma Through Negative Regulation of the Yes-Associated Protein. DNA Cell Biol 2018; 37:659-669. [PMID: 29902078 DOI: 10.1089/dna.2017.4102] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
- Chengyong Lei
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shidong Lv
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongyi Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuan Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiliang Zhai
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Shanci Wang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Guixing Cai
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Dingheng Lu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhen Sun
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiang Wei
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
The emerging role of ECM crosslinking in T cell mobility as a hallmark of immunosenescence in humans. Ageing Res Rev 2017; 35:322-335. [PMID: 27876574 DOI: 10.1016/j.arr.2016.11.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/26/2016] [Accepted: 11/07/2016] [Indexed: 02/07/2023]
Abstract
Immunosenescence is thought to result from cellular aging and to reflect exposure to environmental stressors and antigens, including cytomegalovirus (CMV). However, not all of the features of immunosenescence are consistent with this view, and this has led to the emergence of the sister theory of "inflammaging". The recently discovered diffuse tissue distribution of resident memory T cells (TRM) which don't recirculate, calls these theories into question. These cells account for most T cells residing in barrier epithelia which sit in and travel through the extracellular matrix (ECM). With almost all studies to date carried out on peripheral blood, the age-related changes of the ECM and their consequences for T cell mobility, which is crucial for the function of these cells, have been largely ignored. We propose an update of the theoretical framework of immunosenescence, based on a novel hypothesis: the increasing stiffness and cross-linking of the senescent ECM lead to a progressive immunodeficiency due to an age-related decrease in T cell mobility and eventually the death of these cells. A key element of this mechanism is the mechanical stress to which the cell cytoplasm and nucleus are subjected during passage through the ECM. This hypothesis is based on an "evo-devo" perspective bringing together some major characteristics of aging, to create a single interpretive framework for immunosenescence.
Collapse
|
9
|
Taccioli C, Sorrentino G, Zannini A, Caroli J, Beneventano D, Anderlucci L, Lolli M, Bicciato S, Del Sal G. MDP, a database linking drug response data to genomic information, identifies dasatinib and statins as a combinatorial strategy to inhibit YAP/TAZ in cancer cells. Oncotarget 2016; 6:38854-65. [PMID: 26513174 PMCID: PMC4770742 DOI: 10.18632/oncotarget.5749] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/05/2015] [Indexed: 02/07/2023] Open
Abstract
Targeted anticancer therapies represent the most effective pharmacological strategies in terms of clinical responses. In this context, genetic alteration of several oncogenes represents an optimal predictor of response to targeted therapy. Integration of large-scale molecular and pharmacological data from cancer cell lines promises to be effective in the discovery of new genetic markers of drug sensitivity and of clinically relevant anticancer compounds. To define novel pharmacogenomic dependencies in cancer, we created the Mutations and Drugs Portal (MDP, http://mdp.unimore.it), a web accessible database that combines the cell-based NCI60 screening of more than 50,000 compounds with genomic data extracted from the Cancer Cell Line Encyclopedia and the NCI60 DTP projects. MDP can be queried for drugs active in cancer cell lines carrying mutations in specific cancer genes or for genetic markers associated to sensitivity or resistance to a given compound. As proof of performance, we interrogated MDP to identify both known and novel pharmacogenomics associations and unveiled an unpredicted combination of two FDA-approved compounds, namely statins and Dasatinib, as an effective strategy to potently inhibit YAP/TAZ in cancer cells.
Collapse
Affiliation(s)
- Cristian Taccioli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Giovanni Sorrentino
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste 34149, Italy.,Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste 34149, Italy
| | - Alessandro Zannini
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste 34149, Italy.,Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste 34149, Italy
| | - Jimmy Caroli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy
| | | | | | - Marco Lolli
- Department of Science and Drug Technology, University of Torino, Torino 10125, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Giannino Del Sal
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, Trieste 34149, Italy.,Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Trieste 34149, Italy
| |
Collapse
|
10
|
Guo H, Cheng Y, Martinka M, McElwee K. High LIFr expression stimulates melanoma cell migration and is associated with unfavorable prognosis in melanoma. Oncotarget 2016; 6:25484-98. [PMID: 26329521 PMCID: PMC4694846 DOI: 10.18632/oncotarget.4688] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 07/13/2015] [Indexed: 11/25/2022] Open
Abstract
Increased or decreased expression of LIF receptor (LIFr) has been reported in several human cancers, including skin cancer, but its role in melanoma is unknown. In this study, we investigated the expression pattern of LIFr in melanoma and assessed its prognostic value. Using tissue microarrays consisting of 441 melanomas and 96 nevi, we found that no normal nevi showed high LIFr expression. LIFr staining was significantly increased in primary melanoma compared to dysplastic nevi (P = 0.0003) and further increased in metastatic melanoma (P = 0.0000). Kaplan–Meier survival curve and univariate Cox regression analyses showed that increased expression of LIFr was correlated with poorer 5-year patient survival (overall survival, P = 0.0000; disease-specific survival, P = 0.0000). Multivariate Cox regression analyses indicated that increased LIFr expression was an independent prognostic marker for primary melanoma (P = 0.036). LIFr knockdown inhibited melanoma cell migration in wound healing assays and reduced stress fiber formation. LIFr knockdown correlated with STAT3 suppression, but not YAP, suggesting that LIFr activation might stimulate melanoma cell migration through the STAT3 pathway. Our data indicate that strong LIFr expression identifies potentially highly malignant melanocytic lesions at an early stage and LIFr may be a potential target for the development of early intervention therapeutics.
Collapse
Affiliation(s)
- Hongwei Guo
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, Canada.,Department of Dermatology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Yabin Cheng
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, Canada
| | - Magdalena Martinka
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kevin McElwee
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, Canada
| |
Collapse
|
11
|
Sun Y, Zhang J, Ma L. α-catenin. A tumor suppressor beyond adherens junctions. Cell Cycle 2015; 13:2334-9. [PMID: 25483184 DOI: 10.4161/cc.29765] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Yutong Sun
- a Department of Molecular and Cellular Oncology; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| | | | | |
Collapse
|