1
|
Chen Y, Zhou C, Zhang X, Chen M, Wang M, Zhang L, Chen Y, Huang L, Sun J, Wang D, Chen Y. Construction of a novel radioresistance-related signature for prediction of prognosis, immune microenvironment and anti-tumour drug sensitivity in non-small cell lung cancer. Ann Med 2025; 57:2447930. [PMID: 39797413 PMCID: PMC11727174 DOI: 10.1080/07853890.2024.2447930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/26/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a fatal disease, and radioresistance is an important factor leading to treatment failure and disease progression. The objective of this research was to detect radioresistance-related genes (RRRGs) with prognostic value in NSCLC. METHODS The weighted gene coexpression network analysis (WGCNA) and differentially expressed genes (DEGs) analysis were performed to identify RRRGs using expression profiles from TCGA and GEO databases. The least absolute shrinkage and selection operator (LASSO) regression and random survival forest (RSF) were used to screen for prognostically relevant RRRGs. Multivariate Cox regression was used to construct a risk score model. Then, Immune landscape and drug sensitivity were evaluated. The biological functions exerted by the key gene LBH were verified by in vitro experiments. RESULTS Ninety-nine RRRGs were screened by intersecting the results of DEGs and WGCNA, then 11 hub RRRGs associated with survival were identified using machine learning algorithms (LASSO and RSF). Subsequently, an eight-gene (APOBEC3B, DOCK4, IER5L, LBH, LY6K, RERG, RMDN2 and TSPAN2) risk score model was established and demonstrated to be an independent prognostic factor in NSCLC on the basis of Cox regression analysis. The immune landscape and sensitivity to anti-tumour drugs showed significant disparities between patients categorized into different risk score subgroups. In vitro experiments indicated that overexpression of LBH enhanced the radiosensitivity of A549 cells, and knockdown LBH reversed the cytotoxicity induced by X-rays. CONCLUSION Our study developed an eight-gene risk score model with potential clinical value that can be adopted for choice of drug treatment and prognostic prediction. Its clinical routine use may assist clinicians in selecting more rational practices for individuals, which is important for improving the prognosis of NSCLC patients. These findings also provide references for the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Yanliang Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Chan Zhou
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xiaoqiao Zhang
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Min Chen
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Meifang Wang
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Lisha Zhang
- Department of Obstetrics, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Yanhui Chen
- Department of Neuroscience and Endocrinology, Tangshan Caofeidian District Hospital, Tangshan, Hebei, China
| | - Litao Huang
- Department of Clinical Research Management, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junjun Sun
- Department of Emergency Surgery, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei, , China
| | - Dandan Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | - Yong Chen
- Department of Radio-Chemotherapy, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
2
|
Ma C, Hao Y, Shi B, Wu Z, Jin D, Yu X, Jin B. Unveiling mitochondrial and ribosomal gene deregulation and tumor microenvironment dynamics in acute myeloid leukemia. Cancer Gene Ther 2024; 31:1034-1048. [PMID: 38806621 DOI: 10.1038/s41417-024-00788-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Acute myeloid leukemia (AML) is a malignant clonal hematopoietic disease with a poor prognosis. Understanding the interaction between leukemic cells and the tumor microenvironment (TME) can help predict the prognosis of leukemia and guide its treatment. Re-analyzing the scRNA-seq data from the CSC and G20 cohorts, using a Python-based pipeline including machine-learning-based scVI-tools, recapitulated the distinct hierarchical structure within the samples of AML patients. Weighted correlation network analysis (WGCNA) was conducted to construct a weighted gene co-expression network and to identify gene modules primarily focusing on hematopoietic stem cells (HSCs), multipotent progenitors (MPPs), and natural killer (NK) cells. The analysis revealed significant deregulation in gene modules associated with aerobic respiration and ribosomal/cytoplasmic translation. Cell-cell communications were elucidated by the CellChat package, revealing an imbalance of activating and inhibitory immune signaling pathways. Interception of genes upregulated in leukemic HSCs & MPPs as well as in NKG2A-high NK cells was used to construct prognostic models. Normal Cox and artificial neural network models based on 10 genes were developed. The study reveals the deregulation of mitochondrial and ribosomal genes in AML patients and suggests the co-occurrence of stimulatory and inhibitory factors in the AML TME.
Collapse
Affiliation(s)
- Chao Ma
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Yuchao Hao
- Department of Hematology, The Second Hospital of Dalian Medical University, West Section Lvshun South Road, Dalian, 116027, Liaoning, China
| | - Bo Shi
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Zheng Wu
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Di Jin
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China
| | - Xiao Yu
- NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, South Jiefang Road, Taiyuan, 030001, Shanxi, China.
| | - Bilian Jin
- Institute of Cancer Stem Cell, Dalian Medical University, West Section Lvshun South Road, Dalian, 116044, Liaoning, China.
| |
Collapse
|
3
|
Zhang H, Song Q, Shang K, Li Y, Jiang L, Yang L. Tspan protein family: focusing on the occurrence, progression, and treatment of cancer. Cell Death Discov 2024; 10:187. [PMID: 38649381 PMCID: PMC11035590 DOI: 10.1038/s41420-024-01961-0] [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: 07/15/2023] [Revised: 03/29/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
The Tetraspanins (Tspan) protein family, also known as the tetraspanin family, contains 33 family members that interact with other protein molecules such as integrins, adhesion molecules, and T cell receptors by forming dimers or heterodimers. The Tspan protein family regulates cell proliferation, cell cycle, invasion, migration, apoptosis, autophagy, tissue differentiation, and immune response. More and more studies have shown that Tspan proteins are involved in tumorigenesis, epithelial-mesenchymal transition, thrombosis, tumor stem cell, and exosome signaling. Some drugs and microRNAs can inhibit Tspan proteins, thus providing new strategies for tumor therapy. An in-depth understanding of the functions and regulatory mechanisms of the Tspan protein family, which can promote or inhibit tumor development, will provide new strategies for targeted interventions in the future.
Collapse
Affiliation(s)
- Huhu Zhang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, 266071, China
| | - Qinghang Song
- Health Science Center, Qingdao University, Qingdao, 266071, China
| | - Kaiwen Shang
- Health Science Center, Qingdao University, Qingdao, 266071, China
| | - Ya Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, 266071, China
| | - Liangqian Jiang
- Department of Medical Genetics, Linyi People's Hospital, Linyi, China
| | - Lina Yang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
4
|
Igarashi T, Mazevet M, Yasuhara T, Yano K, Mochizuki A, Nishino M, Yoshida T, Yoshida Y, Takamatsu N, Yoshimi A, Shiraishi K, Horinouchi H, Kohno T, Hamamoto R, Adachi J, Zou L, Shiotani B. An ATR-PrimPol pathway confers tolerance to oncogenic KRAS-induced and heterochromatin-associated replication stress. Nat Commun 2023; 14:4991. [PMID: 37591859 PMCID: PMC10435487 DOI: 10.1038/s41467-023-40578-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023] Open
Abstract
Activation of the KRAS oncogene is a source of replication stress, but how this stress is generated and how it is tolerated by cancer cells remain poorly understood. Here we show that induction of KRASG12V expression in untransformed cells triggers H3K27me3 and HP1-associated chromatin compaction in an RNA transcription dependent manner, resulting in replication fork slowing and cell death. Furthermore, elevated ATR expression is necessary and sufficient for tolerance of KRASG12V-induced replication stress to expand replication stress-tolerant cells (RSTCs). PrimPol is phosphorylated at Ser255, a potential Chk1 substrate site, under KRASG12V-induced replication stress and promotes repriming to maintain fork progression and cell survival in an ATR/Chk1-dependent manner. However, ssDNA gaps are generated at heterochromatin by PrimPol-dependent repriming, leading to genomic instability. These results reveal a role of ATR-PrimPol in enabling precancerous cells to survive KRAS-induced replication stress and expand clonally with accumulation of genomic instability.
Collapse
Affiliation(s)
- Taichi Igarashi
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Biosciences, School of Science, Kitasato University, Minami-ku, Sagamihara-city, Kanagawa, 252-0373, Japan
| | - Marianne Mazevet
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takaaki Yasuhara
- Department of Late Effects Studies, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Kimiyoshi Yano
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
| | - Akifumi Mochizuki
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Makoto Nishino
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yukihiro Yoshida
- Department of Thoracic Surgery, National Cancer Center Hospital, Chuo-ku, Tokyo, 104-0045, Japan
| | - Nobuhiko Takamatsu
- Department of Biosciences, School of Science, Kitasato University, Minami-ku, Sagamihara-city, Kanagawa, 252-0373, Japan
| | - Akihide Yoshimi
- Department of Biosciences, School of Science, Kitasato University, Minami-ku, Sagamihara-city, Kanagawa, 252-0373, Japan
- Division of Cancer RNA Research, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Clinical Genomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
| | - Ryuji Hamamoto
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Laboratory of Clinical and Analytical Chemistry, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki-city, Osaka, 567-0085, Japan
| | - Lee Zou
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Bunsyo Shiotani
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
5
|
PRIM2 Promotes Cell Cycle and Tumor Progression in p53-Mutant Lung Cancer. Cancers (Basel) 2022; 14:cancers14143370. [PMID: 35884433 PMCID: PMC9320259 DOI: 10.3390/cancers14143370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary The mutation or inactivation of tumor suppressor genes is a key driving force during tumorigenesis, among which, p53 mutation is a common feature of human cancer. Therefore, exploring the potential role of p53 mutation in the occurrence and development of tumors is a powerful support for tumor diagnosis and treatment. In this study, we found that PRIM2 expression was abnormally elevated in p53-mutated lung cancer patients, and the elevated PRIM2 promoted DNA replication, enhanced mismatch repair, activated cell cycle, and promoted lung cancer progression. Here, we first report that the expression of PRIM2 is regulated by p53, and is identified as a biomarker of lung cancer malignancy and survival prognosis. Abstract p53 is a common tumor suppressor, and its mutation drives tumorigenesis. What is more, p53 mutations have also been reported to be indicative of poor prognosis in lung cancer, but the detailed mechanism has not been elucidated. In this study, we found that DNA primase subunit 2 (PRIM2) had a high expression level and associated with poor prognosis in lung cancer. Furthermore, we found that PRIM2 expression was abnormally increased in lung cancer cells with p53 mutation or altered the p53/RB pathway based on database. We also verified that PRIM2 expression was elevated by mutation or deletion of p53 in lung cancer cell lines. Lastly, silence p53 increased the expression of RPIM2. Thus, these data suggest that PRIM2 is a cancer-promoting factor which is regulated by the p53/RB pathway. The p53 tumor-suppressor gene integrates numerous signals that control cell proliferation, cell cycle, and cell death; and the p53/RB pathway determines the cellular localization of transcription factor E2F, which regulates the expression of downstream targets. Next, we explored the role of PRIM2 in lung cancer and found that knockdown of PRIM2 induced cell cycle arrest, increased DNA damage, and increased cell senescence, leading to decreased lung cancer cell proliferation. Lastly, the positive correlation between PRIM2 and E2F/CDK also indicated that PRIM2 was involved in promoting cell cycle mediated by p53/RB pathway. These results confirmed that the expression of PRIM2 is regulated by the p53/RB pathway in lung cancer cells, promotes DNA replication and mismatch repair, and activates the cell cycle. Overall, we found that frequent p53 mutations increased PRIM2 expression, activated the cell cycle, and promoted lung cancer progression.
Collapse
|
6
|
Cristofoletti C, Bresin A, Fioretti M, Russo G, Narducci MG. Combined High-Throughput Approaches Reveal the Signals Driven by Skin and Blood Environments and Define the Tumor Heterogeneity in Sézary Syndrome. Cancers (Basel) 2022; 14:cancers14122847. [PMID: 35740513 PMCID: PMC9221051 DOI: 10.3390/cancers14122847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Sézary syndrome (SS) is a leukemic and incurable variant of cutaneous T-cell lymphoma characterized by the accumulation of neoplastic CD4+ lymphocytes in the blood, lymph nodes, and skin. With the exception of allogenic transplantation, no curative chance is available to treat SS, and it is a priority to find new therapies that target SS cells within all disease compartments. This review aims to summarize the more recent analyses conducted on skin- and blood-derived SS cells concurrently obtained from the same SS patients. The results highlighted that skin-SS cells were more active/proliferating with respect to matched blood SS cells that instead appeared quiescent. These data shed the light on the possibility to treat blood and skin SS cells with different compounds, respectively. Moreover, this review recaps the more recent findings on the heterogeneity of circulating SS cells that presented a series of novel markers that could improve diagnosis, prognosis and therapy of this lymphoma. Abstract Sézary syndrome (SS) is an aggressive variant of cutaneous t-cell lymphoma characterized by the accumulation of neoplastic CD4+ lymphocytes—the SS cells—mainly in blood, lymph nodes, and skin. The tumor spread pattern of SS makes this lymphoma a unique model of disease that allows a concurrent blood and skin sampling for analysis. This review summarizes the recent studies highlighting the transcriptional programs triggered by the crosstalk between SS cells and blood–skin microenvironments. Emerging data proved that skin-derived SS cells show consistently higher activation/proliferation rates, mainly driven by T-cell receptor signaling with respect to matched blood SS cells that instead appear quiescent. Biochemical analyses also demonstrated an hyperactivation of PI3K/AKT/mTOR, a targetable pathway by multiple inhibitors currently in clinical trials, in skin SS cells compared with a paired blood counterpart. These results indicated that active and quiescent SS cells coexist in this lymphoma, and that they could be respectively treated with different therapeutics. Finally, this review underlines the more recent discoveries into the heterogeneity of circulating SS cells, highlighting a series of novel markers that could improve the diagnosis and that represent novel therapeutic targets (GPR15, PTPN13, KLRB1, and ITGB1) as well as new genetic markers (PD-1 and CD39) able to stratify SS patients for disease aggressiveness.
Collapse
|
7
|
Garcia-Mayea Y, Mir C, Carballo L, Sánchez-García A, Bataller M, LLeonart ME. TSPAN1, a novel tetraspanin member highly involved in carcinogenesis and chemoresistance. Biochim Biophys Acta Rev Cancer 2021; 1877:188674. [PMID: 34979155 DOI: 10.1016/j.bbcan.2021.188674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022]
Abstract
The tetraspanin (TSPAN) family constitutes a poorly explored family of membrane receptors involved in various physiological processes, with relevant roles in anchoring multiple proteins, acting as scaffolding proteins, and cell signaling. Recent studies have increasingly demonstrated the involvement of TSPANs in cancer. In particular, tetraspanin 1 (also known as TSPAN1, NET-1, TM4C, C4.8 or GEF) has been implicated in cell survival, proliferation and invasion. Recently, our laboratory revealed a key role of TSPAN1 in the acquired resistance of tumor cells to conventional chemotherapy (e.g., cisplatin). In this review, we summarize and discuss the latest research on the physiological mechanisms of TSPANs in cancer and, in particular, on TSPAN1 regulating resistance to chemotherapy. A model of TSPAN1 action is proposed, and the potential of targeting TSPAN1 in anticancer therapeutic strategies is discussed.
Collapse
Affiliation(s)
- Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Laia Carballo
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Almudena Sánchez-García
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Marina Bataller
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Matilde E LLeonart
- Biomedical Research in Cancer Stem Cells Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain; Spanish Biomedical Research Network Center in Oncology, CIBERONC, Spain.
| |
Collapse
|
8
|
Kripnerová M, Parmar HS, Šána J, Kopková A, Radová L, Sopper S, Biernacki K, Jedlička J, Kohoutová M, Kuncová J, Peychl J, Rudolf E, Červinka M, Houdek Z, Dvořák P, Houfková K, Pešta M, Tůma Z, Dolejšová M, Tichánek F, Babuška V, Leba M, Slabý O, Hatina J. Complex Interplay of Genes Underlies Invasiveness in Fibrosarcoma Progression Model. J Clin Med 2021; 10:jcm10112297. [PMID: 34070472 PMCID: PMC8197499 DOI: 10.3390/jcm10112297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/03/2022] Open
Abstract
Sarcomas are a heterogeneous group of mesenchymal tumours, with a great variability in their clinical behaviour. While our knowledge of sarcoma initiation has advanced rapidly in recent years, relatively little is known about mechanisms of sarcoma progression. JUN-murine fibrosarcoma progression series consists of four sarcoma cell lines, JUN-1, JUN-2, JUN-2fos-3, and JUN-3. JUN-1 and -2 were established from a single tumour initiated in a H2K/v-jun transgenic mouse, JUN-3 originates from a different tumour in the same animal, and JUN-2fos-3 results from a targeted in vitro transformation of the JUN-2 cell line. The JUN-1, -2, and -3 cell lines represent a linear progression from the least transformed JUN-2 to the most transformed JUN-3, with regard to all the transformation characteristics studied, while the JUN-2fos-3 cell line exhibits a unique transformation mode, with little deregulation of cell growth and proliferation, but pronounced motility and invasiveness. The invasive sarcoma sublines JUN-2fos-3 and JUN-3 show complex metabolic profiles, with activation of both mitochondrial oxidative phosphorylation and glycolysis and a significant increase in spared respiratory capacity. The specific transcriptomic profile of invasive sublines features very complex biological relationships across the identified genes and proteins, with accentuated autocrine control of motility and angiogenesis. Pharmacologic inhibition of one of the autocrine motility factors identified, Ccl8, significantly diminished both motility and invasiveness of the highly transformed fibrosarcoma cell. This progression series could be greatly valuable for deciphering crucial aspects of sarcoma progression and defining new prognostic markers and potential therapeutic targets.
Collapse
Affiliation(s)
- Michaela Kripnerová
- Institute of Biology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Hamendra Singh Parmar
- Institute of Biology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Jiří Šána
- Central European Institute of Technology (CEITEC), Masaryk University, 625 00 Brno, Czech Republic
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 602 00 Brno, Czech Republic
| | - Alena Kopková
- Central European Institute of Technology (CEITEC), Masaryk University, 625 00 Brno, Czech Republic
- Department of Pathology, University Hospital Brno, 625 00 Brno, Czech Republic
| | - Lenka Radová
- Central European Institute of Technology (CEITEC), Masaryk University, 625 00 Brno, Czech Republic
| | - Sieghart Sopper
- Internal Medicine V, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Tyrolean Cancer Research Institute, 6020 Innsbruck, Austria
| | - Krzysztof Biernacki
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-808 Zabrze, Poland
| | - Jan Jedlička
- Institute of Physiology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Michaela Kohoutová
- Institute of Physiology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Jitka Kuncová
- Institute of Physiology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Jan Peychl
- Department of Medical Biology and Genetics, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic
| | - Emil Rudolf
- Department of Medical Biology and Genetics, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic
| | - Miroslav Červinka
- Department of Medical Biology and Genetics, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic
| | - Zbyněk Houdek
- Institute of Biology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Pavel Dvořák
- Institute of Biology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Kateřina Houfková
- Institute of Biology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Martin Pešta
- Institute of Biology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Zdeněk Tůma
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Martina Dolejšová
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Filip Tichánek
- Institute of Pathological Physiology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| | - Václav Babuška
- Institute of Medical Chemistry and Biochemistry, Faculty of Medicine in Pilsen, Charles University, 301 66 Plzen, Czech Republic
| | - Martin Leba
- Department of Cybernetics, Faculty of Applied Sciences, University of West Bohemia in Pilsen, 301 00 Plzen, Czech Republic
| | - Ondřej Slabý
- Central European Institute of Technology (CEITEC), Masaryk University, 625 00 Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Jiří Hatina
- Institute of Biology, Faculty of Medicine in Pilsen, Charles University, 323 00 Plzen, Czech Republic
| |
Collapse
|
9
|
Gao J, Tang T, Zhang B, Li G. A Prognostic Signature Based on Immunogenomic Profiling Offers Guidance for Esophageal Squamous Cell Cancer Treatment. Front Oncol 2021; 11:603634. [PMID: 33718151 PMCID: PMC7943886 DOI: 10.3389/fonc.2021.603634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Our study aimed to develop an immune prognostic signature that could provide accurate guidance for the treatment of esophageal squamous cell cancer (ESCC). By implementing Single-Sample Gene Set Enrichment Analysis (ssGSEA), we established two ESCC subtypes (Immunity High and Immunity Low) in GSE53625 based on immune-genomic profiling of twenty-nine immune signature. We verified the reliability and reproducibility of this classification in the TCGA database. Immunity High could respond optimally to immunotherapy due to higher expression of immune checkpoints, including PD1, PDL1, CTLA4, and CD80. We used WGCNA analysis to explore the underlying regulatory mechanism of the Immunity High group. We further identified differentially expressed immune-related genes (CCR5, TSPAN2) in GSE53625 and constructed an independent two-gene prognostic signature we internally validated through calibration plots. We established that high-risk ESCC patients had worse overall survival (P=0.002, HR=2.03). Besides, high-risk ESCC patients had elevated levels of infiltrating follicle-helper T cells, naïve B cells, and macrophages as well as had overexpressed levels of some immune checkpoints, including B3H7, CTLA4, CD83, OX40L, and GEM. Moreover, through analyzing the Genomics of Drug Sensitivity in Cancer (GDSC) database, the high-risk group demonstrated drug resistance to some chemotherapy and targeted drugs such as paclitaxel, gefitinib, erlotinib, and lapatinib. Furthermore, we established a robust nomogram model to predict the clinical outcome in ESCC patients. Altogether, our proposed immune prognostic signature constitutes a clinically potential biomarker that will aid in evaluating ESCC outcomes and promote personalized treatment.
Collapse
Affiliation(s)
- Jianyao Gao
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ting Tang
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Baohui Zhang
- Department of Physiology, School of Life Science, China Medical University, Shenyang, China
| | - Guang Li
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Zhang C, Luo Y, Zhang Z, Zhang Z, Zhang G, Wang F, Che Y, Fang L, Zhang Y, Sun N, He J. Identification of a Prognostic Immune Signature for Esophageal Squamous Cell Carcinoma to Predict Survival and Inflammatory Landscapes. Front Cell Dev Biol 2020; 8:580005. [PMID: 33392181 PMCID: PMC7773787 DOI: 10.3389/fcell.2020.580005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/03/2020] [Indexed: 01/06/2023] Open
Abstract
Immunotherapy has achieved success in the treatment of esophageal squamous cell carcinoma (ESCC). However, studies concerning immune phenotypes within the ESCC microenvironment and their relationship with prognostic outcomes are limited. We constructed and validated an individual immune-related risk signature for patients with ESCC. We collected 196 ESCC cases, including 119 samples from our previous public data (GSE53624) to use as a training set and an independent cohort with 77 quantitative real-time polymerase chain reaction (qRT-PCR) data, which we used for validation. Head and neck squamous cell carcinoma (HNSCC) and lung squamous cell carcinoma (LUSC) cohorts were also collected for validation. A least absolute shrinkage and selection operator (LASSO) model and a stepwise Cox proportional hazards regression model were used to construct the immune-specific signature. The potential mechanism and inflammatory landscapes of the signature were explored using bioinformatics and immunofluorescence assay methods. This signature predicted different prognoses in clinical subgroups and the independent cohort, as well as in patients with HNSCC and LUSC. Further exploration revealed that the signature was associated with specific inflammatory activities (activation of macrophages and T-cell signaling transduction). Additionally, high-risk patients exhibited distinctive immune checkpoints panel and higher regulatory T cell and fibroblast infiltration. This signature served as an independent prognostic factor in ESCC. This was the first applicable immune-related risk signature for ESCC. Our results furnished new hints of immune profiling of ESCC, which may provide some clues to further optimize associated cancer immunotherapies.
Collapse
Affiliation(s)
- Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuejun Luo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihui Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Feng Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yun Che
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lingling Fang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Orinska Z, Hagemann PM, Halova I, Draber P. Tetraspanins in the regulation of mast cell function. Med Microbiol Immunol 2020; 209:531-543. [PMID: 32507938 PMCID: PMC7395004 DOI: 10.1007/s00430-020-00679-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022]
Abstract
Mast cells (MCs) are long-living immune cells highly specialized in the storage and release of different biologically active compounds and are involved in the regulation of innate and adaptive immunity. MC degranulation and replacement of MC granules are accompanied by active membrane remodelling. Tetraspanins represent an evolutionary conserved family of transmembrane proteins. By interacting with lipids and other membrane and intracellular proteins, they are involved in organisation of membrane protein complexes and act as "molecular facilitators" connecting extracellular and cytoplasmic signaling elements. MCs express different tetraspanins and MC degranulation is accompanied by changes in membrane organisation. Therefore, tetraspanins are very likely involved in the regulation of MC exocytosis and membrane reorganisation after degranulation. Antiviral response and production of exosomes are further aspects of MC function characterized by dynamic changes of membrane organization. In this review, we pay a particular attention to tetraspanin gene expression in different human and murine MC populations, discuss tetraspanin involvement in regulation of key MC signaling complexes, and analyze the potential contribution of tetraspanins to MC antiviral response and exosome production. In-depth knowledge of tetraspanin-mediated molecular mechanisms involved in different aspects of the regulation of MC response will be beneficial for patients with allergies, characterized by overwhelming MC reactions.
Collapse
Affiliation(s)
- Zane Orinska
- Division of Experimental Pneumology, Research Center Borstel, Leibniz Lungenzentrum, Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.
| | - Philipp M Hagemann
- Division of Experimental Pneumology, Research Center Borstel, Leibniz Lungenzentrum, Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany
| | - Ivana Halova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
12
|
TSPAN8 as a Novel Emerging Therapeutic Target in Cancer for Monoclonal Antibody Therapy. Biomolecules 2020; 10:biom10030388. [PMID: 32138170 PMCID: PMC7175299 DOI: 10.3390/biom10030388] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 12/13/2022] Open
Abstract
Tetraspanin 8 (TSPAN8) is a member of the tetraspanin superfamily that forms TSPAN8-mediated protein complexes by interacting with themselves and other various cellular signaling molecules. These protein complexes help build tetraspanin-enriched microdomains (TEMs) that efficiently mediate intracellular signal transduction. In physiological conditions, TSPAN8 plays a vital role in the regulation of biological functions, including leukocyte trafficking, angiogenesis and wound repair. Recently, reports have increasingly shown the functional role and clinical relevance of TSPAN8 overexpression in the progression and metastasis of several cancers. In this review, we will highlight the physiological and pathophysiological roles of TSPAN8 in normal and cancer cells. Additionally, we will cover the current status of monoclonal antibodies specifically targeting TSPAN8 and the importance of TSPAN8 as an emerging therapeutic target in cancers for monoclonal antibody therapy.
Collapse
|
13
|
Lin SY, Miao YR, Hu FF, Hu H, Zhang Q, Li Q, Chen Z, Guo AY. A 6-Membrane Protein Gene score for prognostic prediction of cytogenetically normal acute myeloid leukemia in multiple cohorts. J Cancer 2020; 11:251-259. [PMID: 31892991 PMCID: PMC6930412 DOI: 10.7150/jca.35382] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Cytogenetically normal acute myeloid leukemia (CN-AML) is a large proportion of AMLs with diverse prognostic outcomes. Identifying membrane protein genes as prognostic factors to stratify CN-AML patients will be critical to improve their outcomes. Purpose: This study aims to identify prognostic factors to stratify CN-AML patients to choose better treatments and improve their outcomes. Methods: CN-AML data were from TCGA cohort (n = 79) and four GEO datasets. We identified independent prognostic genes by Cox regression and Kaplan-Meier methods, and constructed linear regression model using LASSO algorithm. The prediction error curve was calculated using R package “pec”. Results: Based on independent prognostic membrane genes, we constructed a regression model for CN-AML prognosis prediction: score = (0.0492 * CD52) - (0.0018 * CD96) + (0.0131 * EMP1) + (0.2058 * TSPAN2) + (0.0234 * STAB1) - (0.3658 * MBTPS1), which was named as MPG6 (6-Membrane Protein Gene) score. Tested in multiple CN-AML datasets, consistent results showed that CN-AML patients with high MPG6 score had poor survival, higher WBC count and shorter EFS. Comparing with other reported scoring models, the benchmark result of MPG6 achieved better association with survival in multiple cohorts. Moreover, by combining with other clinical indicators in CN-AML, MPG6 could improve the performance of survival prediction and serve as a robust prognostic factor. Conclusions: We identified the MPG6 score as a stable indicator with great potential for clinical application in risk stratification and outcome prediction in CN-AML.
Collapse
Affiliation(s)
- Sheng-Yan Lin
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Ya-Ru Miao
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Fei-Fei Hu
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Hui Hu
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qiong Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qiubai Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhichao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - An-Yuan Guo
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
14
|
Li P, Dong M, Wang Z. Downregulation of TSPAN13 by miR-369-3p inhibits cell proliferation in papillary thyroid cancer (PTC). Bosn J Basic Med Sci 2019; 19:146-154. [PMID: 30114378 DOI: 10.17305/bjbms.2018.2865] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 01/30/2018] [Indexed: 01/07/2023] Open
Abstract
Previous studies demonstrated dysregulation of different microRNAs in thyroid cancer. Tetraspanins (TSPANs) are cell surface proteins with critical roles in many cellular processes, and implications in tumor development. Here we investigated the role of miR-369-3p in papillary thyroid cancer (PTC) and its association with TSPAN13. miR-369-3p and the TSPAN13 gene expression profiles of 513 thyroid cancer and 59 normal thyroid tissues were downloaded from the Cancer Genome Atlas database. Thyroid cancer tissues were classified according to the histological type, grouped based on low and high median miR-369-3p and TSPAN13 expression, and analyzed in relation to overall survival (OS) of patients. Human PTC cell lines (TPC-1 and GLAG-66) and human embryonic kidney 293T (HEK293T) cells were used for in vitro analysis. Transfection experiments were performed with synthetic miRNA mimics for miR-369-3p and small interfering RNAs for TSPAN13. Relative expression of miR-369-3p and TSPAN13 mRNA was determined by RT-qPCR. Protein levels of TSPAN13 were determined by western blotting. Cell proliferation (CCK-8 assay), colony formation, and apoptosis (flow cytometry) were analyzed in transfected cells. Binding sites of miR-369-3p in TSPAN13 mRNA were determined by bioinformatics analysis and dual luciferase reporter assay. miR-369-3p was downregulated and TSPAN13 upregulated in PTC, follicular thyroid cancer, and tall cell variant tissues. Both low expression of miR-369-3p and high expression of TSPAN13 were associated with shorter OS in thyroid cancer patients. Overexpression of miR-369-3p significantly suppressed proliferation and promoted apoptosis in PTC cells. TSPAN13 was a direct target of miR-369-3p, and silencing of TSPAN13 phenocopied the effect of miR-369-3p mimics in PTC cells. Overall, the downregulation of miR-369-3p and consequent upregulation of its target TSPAN13 appear to be involved in pathophysiology of PTC.
Collapse
Affiliation(s)
- Peng Li
- Department of Emergency, Jingmen NO.1 People's Hospital, Jingmen, Hubei, China.
| | | | | |
Collapse
|
15
|
Borcherding N, Voigt AP, Liu V, Link BK, Zhang W, Jabbari A. Single-Cell Profiling of Cutaneous T-Cell Lymphoma Reveals Underlying Heterogeneity Associated with Disease Progression. Clin Cancer Res 2019; 25:2996-3005. [PMID: 30718356 PMCID: PMC6659117 DOI: 10.1158/1078-0432.ccr-18-3309] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/07/2018] [Accepted: 01/25/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Cutaneous T-cell lymphomas (CTCL), encompassing a spectrum of T-cell lymphoproliferative disorders involving the skin, have collectively increased in incidence over the last 40 years. Sézary syndrome is an aggressive form of CTCL characterized by significant presence of malignant cells in both the blood and skin. The guarded prognosis for Sézary syndrome reflects a lack of reliably effective therapy, due, in part, to an incomplete understanding of disease pathogenesis. EXPERIMENTAL DESIGN Using single-cell sequencing of RNA and the machine-learning reverse graph embedding approach in the Monocle package, we defined a model featuring distinct transcriptomic states within Sézary syndrome. Gene expression used to differentiate the unique transcriptional states were further used to develop a boosted tree classification for early versus late CTCL disease. RESULTS Our analysis showed the involvement of FOXP3 + malignant T cells during clonal evolution, transitioning from FOXP3 + T cells to GATA3 + or IKZF2 + (HELIOS) tumor cells. Transcriptomic diversities in a clonal tumor can be used to predict disease stage, and we were able to characterize a gene signature that predicts disease stage with close to 80% accuracy. FOXP3 was found to be the most important factor to predict early disease in CTCL, along with another 19 genes used to predict CTCL stage. CONCLUSIONS This work offers insight into the heterogeneity of Sézary syndrome, providing better understanding of the transcriptomic diversities within a clonal tumor. This transcriptional heterogeneity can predict tumor stage and thereby offer guidance for therapy.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Andrew P Voigt
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Vincent Liu
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
- Department of Dermatology, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Brian K Link
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
- Department of Internal Medicine, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Weizhou Zhang
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, Iowa
- Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
- Interdisciplinary Program in Immunology, University of Iowa, College of Medicine, Iowa City, Iowa
| | - Ali Jabbari
- Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, Iowa.
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, Iowa
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, Iowa
- Department of Dermatology, University of Iowa, College of Medicine, Iowa City, Iowa
- Interdisciplinary Program in Immunology, University of Iowa, College of Medicine, Iowa City, Iowa
| |
Collapse
|
16
|
Goulart MR, Hlavaty SI, Chang YM, Polton G, Stell A, Perry J, Wu Y, Sharma E, Broxholme J, Lee AC, Szladovits B, Turmaine M, Gribben J, Xia D, Garden OA. Phenotypic and transcriptomic characterization of canine myeloid-derived suppressor cells. Sci Rep 2019; 9:3574. [PMID: 30837603 PMCID: PMC6400936 DOI: 10.1038/s41598-019-40285-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 02/13/2019] [Indexed: 01/19/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are key players in immune evasion, tumor progression and metastasis. MDSCs accumulate under various pathological states and fall into two functionally and phenotypically distinct subsets that have been identified in humans and mice: polymorphonuclear (PMN)-MDSCs and monocytic (M)-MDSCs. As dogs are an excellent model for human tumor development and progression, we set out to identify PMN-MDSCs and M-MDSCs in clinical canine oncology patients. Canine hypodense MHC class II-CD5-CD21-CD11b+ cells can be subdivided into polymorphonuclear (CADO48A+CD14-) and monocytic (CADO48A-CD14+) MDSC subsets. The transcriptomic signatures of PMN-MDSCs and M-MDSCs are distinct, and moreover reveal a statistically significant similarity between canine and previously published human PMN-MDSC gene expression patterns. As in humans, peripheral blood frequencies of canine PMN-MDSCs and M-MDSCs are significantly higher in dogs with cancer compared to healthy control dogs (PMN-MDSCs: p < 0.001; M-MDSCs: p < 0.01). By leveraging the power of evolution, we also identified additional conserved genes in PMN-MDSCs of multiple species that may play a role in MDSC function. Our findings therefore validate the dog as a model for studying MDSCs in the context of cancer.
Collapse
Affiliation(s)
- Michelle R Goulart
- Royal Veterinary College, London, UK
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Sabina I Hlavaty
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - James Perry
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Wu
- Royal Veterinary College, London, UK
| | - Eshita Sharma
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - John Broxholme
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Avery C Lee
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Mark Turmaine
- Division of Bioscience, University College London, London, UK
| | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Dong Xia
- Royal Veterinary College, London, UK
| | - Oliver A Garden
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Voic H, Li X, Jang JH, Zou C, Sundd P, Alder J, Rojas M, Chandra D, Randell S, Mallampalli RK, Tesfaigzi Y, Ryba T, Nyunoya T. RNA sequencing identifies common pathways between cigarette smoke exposure and replicative senescence in human airway epithelia. BMC Genomics 2019; 20:22. [PMID: 30626320 PMCID: PMC6325884 DOI: 10.1186/s12864-018-5409-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 12/26/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Aging is affected by genetic and environmental factors, and cigarette smoking is strongly associated with accumulation of senescent cells. In this study, we wanted to identify genes that may potentially be beneficial for cell survival in response to cigarette smoke and thereby may contribute to development of cellular senescence. RESULTS Primary human bronchial epithelial cells from five healthy donors were cultured, treated with or without 1.5% cigarette smoke extract (CSE) for 24 h or were passaged into replicative senescence. Transcriptome changes were monitored using RNA-seq in CSE and non-CSE exposed cells and those passaged into replicative senescence. We found that, among 1534 genes differentially regulated during senescence and 599 after CSE exposure, 243 were altered in both conditions, representing strong enrichment. Pathways and gene sets overrepresented in both conditions belonged to cellular processes that regulate reactive oxygen species, proteasome degradation, and NF-κB signaling. CONCLUSIONS Our results offer insights into gene expression responses during cellular aging and cigarette smoke exposure, and identify potential molecular pathways that are altered by cigarette smoke and may also promote airway epithelial cell senescence.
Collapse
Affiliation(s)
- Hannah Voic
- 0000 0004 0504 9575grid.422569.eDivision of Natural Sciences, New College of Florida, Sarasota, FL USA
| | - Xiuying Li
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA ,0000 0004 0420 3665grid.413935.9VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| | - Jun-Ho Jang
- 0000 0004 0454 5075grid.417046.0Cardiovascular Institute, Department of Medicine, Allegheny Health Network, Pittsburgh, PA USA
| | - Chunbin Zou
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA ,0000 0004 0420 3665grid.413935.9VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| | - Prithu Sundd
- 0000 0004 1936 9000grid.21925.3dVascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Jonathan Alder
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA
| | - Mauricio Rojas
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA
| | - Divay Chandra
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA
| | - Scott Randell
- 0000 0001 1034 1720grid.410711.2Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC USA
| | - Rama K. Mallampalli
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA ,0000 0004 0420 3665grid.413935.9VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| | - Yohannes Tesfaigzi
- Lovelace Respiratory Research Institute, COPD program, Albuquerque, NM USA
| | - Tyrone Ryba
- 0000 0004 0504 9575grid.422569.eDivision of Natural Sciences, New College of Florida, Sarasota, FL USA
| | - Toru Nyunoya
- 0000 0004 1936 9000grid.21925.3dDepartment of Medicine, University of Pittsburgh, NW628 UPMC Montefiore, 3459 Fifth Avenue, Pittsburgh, PA 15213 USA ,0000 0004 0420 3665grid.413935.9VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| |
Collapse
|
18
|
Fang J, Yuan X, An X, Qu H, Wang C, Hong G, Zheng L, Yi K, Chen S, Wang X, Ma Q. Involvement of the Tetraspanin 2 ( TSPAN2) Gene in Migraine: A Case-Control Study in Han Chinese. Front Neurol 2018; 9:714. [PMID: 30258394 PMCID: PMC6143784 DOI: 10.3389/fneur.2018.00714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/08/2018] [Indexed: 01/03/2023] Open
Abstract
Tetraspanin 2 (TSPAN2) belongs to the tetraspanin superfamily. Previous studies have identified significant associations of the TSPAN2 single nucleotide polymorphisms (SNPs) rs12134493 and rs2078371 with migraine in Western populations; however, these associations need to be confirmed in the Chinese Han population. In addition, we carried out further studies to see whether TSPAN2 is associated with susceptibility to migraine to provide new clinical evidence. A case-control study (425 patients with migraine and 425 healthy controls) in a Chinese Han population was performed to evaluate the associations between migraine and TSPAN2 via a genotype-phenotype analysis between TSPAN2 and clinical symptoms. The SNP rs2078371 was found to be significantly associated with migraines especially in migraines without aura (MO) and in female patients. Meta-analysis revealed that the A allele of rs12134493 was significantly associated with migraines (OR = 1.14, P = 0.0001). Our findings suggested that TSPAN2 is a potential susceptibility factor for migraines. To confirm our results, a large-scale Chinese Han population study should be conducted. Considering that these two SNPs have not been definitively shown to affect TSPAN2 or to regulate nearby genes in this genomic region, the biological function and molecular mechanism of TSPAN2 in migraine should be further explored.
Collapse
Affiliation(s)
- Jie Fang
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xiaodong Yuan
- Department of Gynaecology and Obstetrics, Xiamen Maternal and Child Health Care Hospital, Xiamen, China
| | - Xingkai An
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Hongli Qu
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Chen Wang
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ganji Hong
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | | | - Kehui Yi
- Department of Neurology, Zhongshan Xiamen Hospital, Fudan University, Xiamen, China
| | - Shuai Chen
- Fujian Medical University, Fuzhou, China
| | - Xinrui Wang
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital Affiliated to School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qilin Ma
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Graduate School of Fujian Medical University, Fuzhou, China
| |
Collapse
|
19
|
Miyazaki M, Otomo R, Matsushima-Hibiya Y, Suzuki H, Nakajima A, Abe N, Tomiyama A, Ichimura K, Matsuda K, Watanabe T, Ochiya T, Nakagama H, Sakai R, Enari M. The p53 activator overcomes resistance to ALK inhibitors by regulating p53-target selectivity in ALK-driven neuroblastomas. Cell Death Discov 2018; 4:56. [PMID: 29760954 PMCID: PMC5945735 DOI: 10.1038/s41420-018-0059-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 01/23/2023] Open
Abstract
Anaplastic lymphoma kinase (ALK) is an oncogenic receptor tyrosine kinase that is activated by gene amplification and mutation in neuroblastomas. ALK inhibitors can delay the progression of ALK-driven cancers, but are of limited use owing to ALK inhibitor resistance. Here, we show that resistance to ALK inhibitor in ALK-driven neuroblastomas can be attenuated by combination treatment with a p53 activator. Either ALK inhibition or p53 activator treatment induced cell cycle arrest, whereas combination treatment induced apoptosis, and prevented tumour relapse both in vitro and in vivo. This shift toward apoptosis, and away from cell-cycle arrest, in the presence of an ALK inhibitor and a p53 activator, is mediated by inhibition of the ALK-AKT-FOXO3a axis leading to a specific upregulation of SOX4. SOX4 cooperates with p53 to upregulate the pro-apoptotic protein PUMA. These data therefore suggest a novel combination therapy strategy for treating ALK-driven neuroblastomas.
Collapse
Affiliation(s)
- Makoto Miyazaki
- 1Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,2Department of Computational Biology and Medical Sciences, Laboratory of Clinical Genome Sequencing, Graduate School of Frontier Sciences, The University of Tokyo, Minato-ku, Tokyo 108-8639 Japan.,3Department of Computational Biology and Medical Sciences, Tumour Cell Biology, Graduate School of Frontier Sciences, The University of Tokyo, Minato-ku, Tokyo 108-8639 Japan.,4Division of Brain Tumour Translational Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan
| | - Ryo Otomo
- 1Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,11Present Address: Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Disaster Reconstruction Center, Iwate Medical University, Yahaba-cho, Shiwa-gun, Iwate 028-3694 Japan
| | - Yuko Matsushima-Hibiya
- 1Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,4Division of Brain Tumour Translational Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan
| | - Hidenobu Suzuki
- 1Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,4Division of Brain Tumour Translational Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,5Department of NCC Cancer Science, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510 Japan
| | - Ayana Nakajima
- 1Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,4Division of Brain Tumour Translational Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,6Molecular and Cellular Biology Laboratory, Graduate school of Medical Life Science, Yokohama City University, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Naomi Abe
- 1Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,4Division of Brain Tumour Translational Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan
| | - Arata Tomiyama
- 1Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,4Division of Brain Tumour Translational Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,7Department of Neurosurgery, National Defense Medical College, 3-2, Namiki, Tokorozawa, Saitama, 359-8513 Japan
| | - Koichi Ichimura
- 4Division of Brain Tumour Translational Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan
| | - Koichi Matsuda
- 2Department of Computational Biology and Medical Sciences, Laboratory of Clinical Genome Sequencing, Graduate School of Frontier Sciences, The University of Tokyo, Minato-ku, Tokyo 108-8639 Japan
| | - Toshiki Watanabe
- 3Department of Computational Biology and Medical Sciences, Tumour Cell Biology, Graduate School of Frontier Sciences, The University of Tokyo, Minato-ku, Tokyo 108-8639 Japan
| | - Takahiro Ochiya
- 8Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan
| | - Hitoshi Nakagama
- 9Division of Cancer Development System, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan
| | - Ryuichi Sakai
- 1Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan.,10Division of Biochemistry, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374 Japan
| | - Masato Enari
- 1Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045 Japan
| |
Collapse
|
20
|
Wang J, Zhou Y, Li D, Sun X, Deng Y, Zhao Q. TSPAN
31 is a critical regulator on transduction of survival and apoptotic signals in hepatocellular carcinoma cells. FEBS Lett 2017; 591:2905-2918. [DOI: 10.1002/1873-3468.12737] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Jianglin Wang
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
| | - Yuting Zhou
- Department of Molecular & Cellular Biochemistry College of Medicine University of Kentucky Lexington KY USA
| | - Dan Li
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
| | - Xuemeng Sun
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
| | - Yuanfei Deng
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
| | - Qing Zhao
- Department of Biochemistry and Molecular Biology GMU‐GIBH Joint School of Life Sciences Guangzhou Medical University China
- Sino‐French Hoffmann Institute Guangzhou Medical University China
| |
Collapse
|
21
|
Tspan2: a tetraspanin protein involved in oligodendrogenesis and cancer metastasis. Biochem Soc Trans 2017; 45:465-475. [PMID: 28408487 PMCID: PMC5390497 DOI: 10.1042/bst20160022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 12/14/2022]
Abstract
Tetraspanin 2 (Tspan2) is one of the less well-characterised members of the tetraspanin superfamily, and its precise function in different human tissue types remains to be explored. Initial studies have highlighted its possible association in neuroinflammation and carcinogenesis. In the central nervous system, Tspan2 may contribute to the early stages of the oligodendrocyte differentiation into myelin-forming glia. Furthermore, in human lung cancer, Tspan2 could be involved in the progression of the tumour metastasis by modulating cancer cell motility and invasion functions. In this review, we discuss the available evidence for the potential role of Tspan2 and introduce possible strategies for disease targeting.
Collapse
|
22
|
Zhao J, Wu W, Zhang W, Lu YW, Tou E, Ye J, Gao P, Jourd'heuil D, Singer HA, Wu M, Long X. Selective expression of TSPAN2 in vascular smooth muscle is independently regulated by TGF-β1/SMAD and myocardin/serum response factor. FASEB J 2017; 31:2576-2591. [PMID: 28258189 DOI: 10.1096/fj.201601021r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 02/13/2017] [Indexed: 01/07/2023]
Abstract
Tetraspanins (TSPANs) comprise a large family of 4-transmembrane domain proteins. The importance of TSPANs in vascular smooth muscle cells (VSMCs) is unexplored. Given that TGF-β1 and myocardin (MYOCD) are potent activators for VSMC differentiation, we screened for TGF-β1 and MYOCD/serum response factor (SRF)-regulated TSPANs in VSMC by using RNA-seq analyses and RNA-arrays. TSPAN2 was found to be the only TSPAN family gene induced by TGF-β1 and MYOCD, and reduced by SRF deficiency in VSMCs. We also found that TSPAN2 is highly expressed in smooth muscle-enriched tissues and down-regulated in in vitro models of VSMC phenotypic modulation. TSPAN2 expression is attenuated in mouse carotid arteries after ligation injury and in failed human arteriovenous fistula samples after occlusion by dedifferentiated neointimal VSMC. In vitro functional studies showed that TSPAN2 suppresses VSMC proliferation and migration. Luciferase reporter and chromatin immunoprecipitation assays demonstrated that TSPAN2 is regulated by 2 parallel pathways, MYOCD/SRF and TGF-β1/SMAD, via distinct binding elements within the proximal promoter. Thus, we identified the first VSMC-enriched and MYOCD/SRF and TGF-β1/SMAD-dependent TSPAN family member, whose expression is intimately associated with VSMC differentiation and negatively correlated with vascular disease. Our results suggest that TSPAN2 may play important roles in vascular disease.-Zhao, J., Wu, W., Zhang, W., Lu, Y. W., Tou, E., Ye, J., Gao, P., Jourd'heuil, D., Singer, H. A., Wu, M., Long, X. Selective expression of TSPAN2 in vascular smooth muscle is independently regulated by TGF-β1/SMAD and myocardin/serum response factor.
Collapse
Affiliation(s)
- Jinjing Zhao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Wen Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Wei Zhang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Yao Wei Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Emiley Tou
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Jiemei Ye
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Ping Gao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Mingfu Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| | - Xiaochun Long
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, USA
| |
Collapse
|
23
|
Kayama K, Watanabe S, Takafuji T, Tsuji T, Hironaka K, Matsumoto M, Nakayama KI, Enari M, Kohno T, Shiraishi K, Kiyono T, Yoshida K, Sugimoto N, Fujita M. GRWD1 negatively regulates p53 via the RPL11-MDM2 pathway and promotes tumorigenesis. EMBO Rep 2016; 18:123-137. [PMID: 27856536 DOI: 10.15252/embr.201642444] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 01/11/2023] Open
Abstract
The ribosomal protein L11 (RPL11) binds and inhibits the MDM2 ubiquitin ligase, thereby promoting p53 stability. Thus, RPL11 acts as a tumor suppressor. Here, we show that GRWD1 (glutamate-rich WD40 repeat containing 1) physically and functionally interacts with RPL11. GRWD1 is localized to nucleoli and is released into the nucleoplasm upon nucleolar stress. Silencing of GRWD1 increases p53 induction by nucleolar stress, whereas overexpression of GRWD1 reduces p53 induction. Furthermore, GRWD1 overexpression competitively inhibits the RPL11-MDM2 interaction and alleviates RPL11-mediated suppression of MDM2 ubiquitin ligase activity toward p53. These effects are mediated by the N-terminal region of GRWD1, including the acidic domain. Finally, we show that GRWD1 overexpression in combination with HPV16 E7 and activated KRAS confers anchorage-independent growth and tumorigenic capacity on normal human fibroblasts. Consistent with this, GRWD1 overexpression is associated with poor prognosis in cancer patients. Taken together, our results suggest that GRWD1 is a novel negative regulator of p53 and a potential oncogene.
Collapse
Affiliation(s)
- Kota Kayama
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinya Watanabe
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Takuya Takafuji
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Tsuji
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kensuke Hironaka
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Matsumoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Masato Enari
- Division of Refractory and Advancer Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Tohru Kiyono
- Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, Tokyo, Japan
| | - Kazumasa Yoshida
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Nozomi Sugimoto
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Masatoshi Fujita
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
24
|
Lipchick BC, Fink EE, Nikiforov MA. Oxidative stress and proteasome inhibitors in multiple myeloma. Pharmacol Res 2016; 105:210-5. [PMID: 26827824 PMCID: PMC5044866 DOI: 10.1016/j.phrs.2016.01.029] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 01/20/2016] [Accepted: 01/22/2016] [Indexed: 11/23/2022]
Abstract
Multiple myeloma is a form of plasma cell neoplasm that accounts for approximately 10% of all hematological malignancies. Recently, several novel drugs have been discovered that almost doubled the overall survival of multiple myeloma patients. One of these drugs, the first-in-class proteasome inhibitor bortezomib (Velcade) has demonstrated remarkable response rates in multiple myeloma patients, and yet, currently this disease remains incurable. The major factor undermining the success of multiple myeloma treatment is a rapidly emerging resistance to the available therapy. Thus, the development of stand-alone or adjuvant anti-myeloma agents becomes of paramount importance. Overproduction of intracellular reactive oxygen species (ROS) often accompanies malignant transformation due to oncogene activation and/or enhanced metabolism in tumor cells. As a result, these cells possess higher levels of ROS and lower levels of antioxidant molecules compared to their normal counterparts. Unbalanced production of ROS leads to oxidative stress which, if left unchecked, could be toxic for the cell. In multiple myeloma cells where high rates of immunoglobulin synthesis is an additional factor contributing to overproduction of ROS, further induction of oxidative stress can be an effective strategy to cope with this disease. Here we will review the available data on the role of oxidative stress in the cytotoxicity of proteasome inhibitors and the use of ROS-inducing compounds as anti-myeloma agents.
Collapse
Affiliation(s)
- Brittany C Lipchick
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| | - Emily E Fink
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Mikhail A Nikiforov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
25
|
Murayama T, Nakaoku T, Enari M, Nishimura T, Tominaga K, Nakata A, Tojo A, Sugano S, Kohno T, Gotoh N. Oncogenic Fusion Gene CD74-NRG1 Confers Cancer Stem Cell-like Properties in Lung Cancer through a IGF2 Autocrine/Paracrine Circuit. Cancer Res 2016; 76:974-83. [PMID: 26837769 DOI: 10.1158/0008-5472.can-15-2135] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/26/2015] [Indexed: 11/16/2022]
Abstract
The CD74-Neuregulin1 (NRG1) fusion gene was recently identified as novel driver of invasive mucinous adenocarcinoma, a malignant form of lung cancer. However, the function of the CD74-NRG1 fusion gene in adenocarcinoma pathogenesis and the mechanisms by which it may impart protumorigenic characteristics to cancer stem cells (CSC) is still unclear. In this study, we found that the expression of the CD74-NRG1 fusion gene increased the population of lung cancer cells with CSC-like properties. CD74-NRG1 expression facilitated sphere formation not only of cancer cells, but also of nonmalignant lung epithelial cells. Using a limiting dilution assay in a xenograft model, we further show that the CD74-NRG1 fusion gene enhanced tumor initiation. Mechanistically, we found that CD74-NRG1 expression promoted the phosphorylation of ErbB2/3 and activated the PI3K/Akt/NF-κB signaling pathway. Furthermore, the expression of the secreted insulin-like growth factor 2 (IGF2) and phosphorylation of its receptor, IGF1R, were enhanced in an NF-κB-dependent manner in cells expressing CD74-NRG1. These findings suggest that CD74-NRG1-induced NF-κB activity promotes the IGF2 autocrine/paracrine circuit. Moreover, inhibition of ErbB2, PI3K, NF-κB, or IGF2 suppressed CD74-NRG1-induced tumor sphere formation. Therefore, our study provides a preclinical rationale for developing treatment approaches based on these identified pathways to suppress CSC properties that promote tumor progression and recurrence.
Collapse
Affiliation(s)
- Takahiko Murayama
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan. Laboratory of Functional Genomics, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan. Division of Molecular Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Takashi Nakaoku
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Masato Enari
- Division of Refractory Cancer Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Tatsunori Nishimura
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kana Tominaga
- Division of Molecular Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Asuka Nakata
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Arinobu Tojo
- Division of Molecular Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Sumio Sugano
- Laboratory of Functional Genomics, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan.
| | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan. Division of Molecular Therapy, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
26
|
Yeh CF, Tai W, Lin CH, Juang DS, Wu CC, Chen YW, Hsu CH. Towards an Endpoint Cell Motility Assay by a Microfluidic Platform. IEEE Trans Nanobioscience 2015; 14:835-40. [DOI: 10.1109/tnb.2015.2455537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
27
|
Whitney DH, Elashoff MR, Porta-Smith K, Gower AC, Vachani A, Ferguson JS, Silvestri GA, Brody JS, Lenburg ME, Spira A. Derivation of a bronchial genomic classifier for lung cancer in a prospective study of patients undergoing diagnostic bronchoscopy. BMC Med Genomics 2015; 8:18. [PMID: 25944280 PMCID: PMC4434538 DOI: 10.1186/s12920-015-0091-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/23/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The gene expression profile of cytologically-normal bronchial airway epithelial cells has previously been shown to be altered in patients with lung cancer. Although bronchoscopy is often used for the diagnosis of lung cancer, its sensitivity is imperfect, especially for small and peripheral suspicious lesions. In this study, we derived a gene expression classifier from airway epithelial cells that detects the presence of cancer in current and former smokers undergoing bronchoscopy for suspect lung cancer and evaluated its sensitivity to detect lung cancer among patients from an independent cohort. METHODS We collected bronchial epithelial cells (BECs) from the mainstem bronchus of 299 current or former smokers (223 cancer-positive and 76 cancer-free subjects) undergoing bronchoscopy for suspected lung cancer in a prospective, multi-center study. RNA from these samples was run on gene expression microarrays for training a gene-expression classifier. A logistic regression model was built to predict cancer status, and the finalized classifier was validated in an independent cohort from a previous study. RESULTS We found 232 genes whose expression levels in the bronchial airway are associated with lung cancer. We then built a classifier based on the combination of 17 cancer genes, gene expression predictors of smoking status, smoking history, and gender, plus patient age. This classifier had a ROC curve AUC of 0.78 (95% CI, 0.70-0.86) in patients whose bronchoscopy did not lead to a diagnosis of lung cancer (n = 134). In the validation cohort, the classifier had a similar AUC of 0.81 (95% CI, 0.73-0.88) in this same subgroup (n = 118). The classifier performed similarly across a range of mass sizes, cancer histologies and stages. The negative predictive value was 94% (95% CI, 83-99%) in subjects with a non-diagnostic bronchoscopy. CONCLUSION We developed a gene expression classifier measured in bronchial airway epithelial cells that is able to detect lung cancer in current and former smokers who have undergone bronchoscopy for suspicion of lung cancer. Due to the high NPV of the classifier, it could potentially inform clinical decisions regarding the need for further invasive testing in patients whose bronchoscopy is non diagnostic.
Collapse
Affiliation(s)
- Duncan H Whitney
- Allegro Diagnostics, Corp, Maynard, MA, USA. .,Present affiliation: Veracyte, Inc, South San Francisco, CA, USA.
| | | | - Kate Porta-Smith
- Allegro Diagnostics, Corp, Maynard, MA, USA. .,Present affiliation: Veracyte, Inc, South San Francisco, CA, USA.
| | - Adam C Gower
- Boston University School of Medicine, Boston, MA, USA.
| | - Anil Vachani
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| | - J Scott Ferguson
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | | | | | | | - Avrum Spira
- Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
28
|
Grossmann A, Benlasfer N, Birth P, Hegele A, Wachsmuth F, Apelt L, Stelzl U. Phospho-tyrosine dependent protein-protein interaction network. Mol Syst Biol 2015; 11:794. [PMID: 25814554 PMCID: PMC4380928 DOI: 10.15252/msb.20145968] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Post-translational protein modifications, such as tyrosine phosphorylation, regulate protein–protein interactions (PPIs) critical for signal processing and cellular phenotypes. We extended an established yeast two-hybrid system employing human protein kinases for the analyses of phospho-tyrosine (pY)-dependent PPIs in a direct experimental, large-scale approach. We identified 292 mostly novel pY-dependent PPIs which showed high specificity with respect to kinases and interacting proteins and validated a large fraction in co-immunoprecipitation experiments from mammalian cells. About one-sixth of the interactions are mediated by known linear sequence binding motifs while the majority of pY-PPIs are mediated by other linear epitopes or governed by alternative recognition modes. Network analysis revealed that pY-mediated recognition events are tied to a highly connected protein module dedicated to signaling and cell growth pathways related to cancer. Using binding assays, protein complementation and phenotypic readouts to characterize the pY-dependent interactions of TSPAN2 (tetraspanin 2) and GRB2 or PIK3R3 (p55γ), we exemplarily provide evidence that the two pY-dependent PPIs dictate cellular cancer phenotypes.
Collapse
Affiliation(s)
- Arndt Grossmann
- Otto-Warburg Laboratory, Max-Planck Institute for Molecular Genetics (MPIMG), Berlin, Germany
| | - Nouhad Benlasfer
- Otto-Warburg Laboratory, Max-Planck Institute for Molecular Genetics (MPIMG), Berlin, Germany
| | - Petra Birth
- Otto-Warburg Laboratory, Max-Planck Institute for Molecular Genetics (MPIMG), Berlin, Germany
| | - Anna Hegele
- Otto-Warburg Laboratory, Max-Planck Institute for Molecular Genetics (MPIMG), Berlin, Germany
| | - Franziska Wachsmuth
- Otto-Warburg Laboratory, Max-Planck Institute for Molecular Genetics (MPIMG), Berlin, Germany
| | - Luise Apelt
- Otto-Warburg Laboratory, Max-Planck Institute for Molecular Genetics (MPIMG), Berlin, Germany
| | - Ulrich Stelzl
- Otto-Warburg Laboratory, Max-Planck Institute for Molecular Genetics (MPIMG), Berlin, Germany
| |
Collapse
|
29
|
TSPAN12 is a critical factor for cancer-fibroblast cell contact-mediated cancer invasion. Proc Natl Acad Sci U S A 2014; 111:18691-6. [PMID: 25512506 DOI: 10.1073/pnas.1412062112] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Communication between cancer cells and their microenvironment controls cancer progression. Although the tumor suppressor p53 functions in a cell-autonomous manner, it has also recently been shown to function in a non-cell-autonomous fashion. Although functional defects have been reported in p53 in stromal cells surrounding cancer, including mutations in the p53 gene and decreased p53 expression, the role of p53 in stromal cells during cancer progression remains unclear. We herein show that the expression of α-smooth muscle actin (α-SMA), a marker of cancer-associated fibroblasts (CAFs), was increased by the ablation of p53 in lung fibroblasts. CAFs enhanced the invasion and proliferation of lung cancer cells when cocultured with p53-depleted fibroblasts and required contact between cancer and stromal cells. A comprehensive analysis using a DNA chip revealed that tetraspanin 12 (TSPAN12), which belongs to the tetraspanin protein family, was derepressed by p53 knockdown. TSPAN12 knockdown in p53-depleted fibroblasts inhibited cancer cell proliferation and invasion elicited by coculturing with p53-depleted fibroblasts in vitro, and inhibited tumor growth in vivo. It also decreased CXC chemokine ligand 6 (CXCL6) secretion through the β-catenin signaling pathway, suggesting that cancer cell contact with TSPAN12 in fibroblasts transduced β-catenin signaling into fibroblasts, leading to the secretion of CXCL6 to efficiently promote invasion. These results suggest that stroma-derived p53 plays a pivotal role in epithelial cancer progression and that TSPAN12 and CXCL6 are potential targets for lung cancer therapy.
Collapse
|