1
|
Li J, Yang X, Yin C, Li S, Xu Y, Liu B. CDKN2A, a key gene in copper-induced cell death model, influencing melanoma invasion and apoptosis. Discov Oncol 2025; 16:246. [PMID: 40014167 PMCID: PMC11867994 DOI: 10.1007/s12672-025-01992-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
Skin cutaneous melanoma (SKCM) is one of the most lethal cancers translating into 75% of skin cancer-related deaths. Despite the advances in SKCM management and treatment strategies, the overall survival of patients remains unsatisfactory due to the metastatic properties of SKCM as well as the absence of effective prognostic biomarkers. Recent studies have shown that overload copper renders accumulation of mitochondrial proteins and fuels a form of cell death at odds with known death mechanisms and is hinged on mitochondrial respiration, the so-called cuproptosis. However, the exact role of cuproptosis in SKCM development and progression is unknown, and painting a clear picture of its functions in SKCM is fraught with challenges. A more systematic investigation is justified. In this study, we were posed to dissect the clout and latent regulatory mechanisms of cuproptosis-related genes (CRGs) in reining in SKCM progression. Also, we identified three CRGs that stood out were used to construct a prognostic model, which could be employed to predict the prognosis of patients with SKCM. Finally, through pan-cancer analysis, we found that the four cuproptosis key genes play a role in multiple tumors, suggesting that cuproptosis may impact tumor progression at the pan-cancer level. Taken together, these findings may not only contribute to the development of treatment strategies but also provide clues for treatment decision-making.
Collapse
Affiliation(s)
- Jing Li
- Sichuan Cancer Hospital &Institute, School of Medicine, Sichuan Cancer Center, University of Electronic Science and Technology of China, People's South Road, Section 4, Number 55, Chengdu, 610041, China
| | - Xi Yang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cunli Yin
- Department of Oncology Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Siru Li
- Department of Oncology Sichuan Cancer Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Xu
- Genecast Biotechnology Co., Ltd., Wu Xi, 214105, China
| | - Bin Liu
- Sichuan Cancer Hospital &Institute, School of Medicine, Sichuan Cancer Center, University of Electronic Science and Technology of China, People's South Road, Section 4, Number 55, Chengdu, 610041, China.
| |
Collapse
|
2
|
Luo WF, Song DM, Shen T, He YB, Du HY, Si MJ, Fang LW. Exploring new mechanisms in cancer molecular pathways and pathogenic cell transformation: PIP4K2A as a prognostic marker and therapeutic target in cutaneous malignant melanoma. Discov Oncol 2024; 15:697. [PMID: 39579298 PMCID: PMC11585527 DOI: 10.1007/s12672-024-01555-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/07/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Cutaneous malignant melanoma is a very aggressive and metastatic form of skin cancer, typically linked with poor outcomes. Advances in genomic analysis have underscored the crucial role of T cells in tumor immunity. Immune checkpoint inhibitors have notably transformed melanoma treatment by boosting T cell activity. Studies of gene expression have found that the phosphatidylinositol-4-phosphate kinase 2A (PIP4K2A) gene is abnormally expressed in various tumors, indicating its potential role in tumor progression. Utilizing single-cell sequencing and machine learning, researchers can now explore the complex interactions between T cells and melanoma cells at a genomic level. This study aimed to investigate the role of the PIP4K2A gene in cutaneous malignant melanoma, with a focus on its influence on T cell-mediated immune responses. METHODS Samples from cutaneous melanoma patients were analysed by single-cell transcriptome for differentially expressed genes and signalling pathways associated with cutaneous melanoma. Then, genes were identified and predictive models were built based on the transcriptomic data using machine learning models to assess whether the expression level of PIP4K2A could effectively predict the malignancy and prognosis of cutaneous melanoma. In addition, we also performed drug therapy predictive analysis and immunotherapy analysis.Finally, the critical role of PIP4K2A in cutaneous melanoma was further confirmed by immunohistochemistry. RESULTS The PIP4K2A gene exhibited a significantly elevated expression level in cutaneous malignant melanoma, showing a strong correlation with the clinical stage and patient prognosis. At the therapeutic level, high PIP4K2A expression is less responsive to immunotherapy, and this gene is a risk factor for drug therapy in cutaneous malignant melanoma. Additionally, our experimental outcomes validated this observation. CONCLUSIONS The PIP4K2A gene could be a crucial prognostic marker for cutaneous malignant melanoma, as it significantly affects T cell activity within the tumor microenvironment. This study offers essential insights into melanoma pathogenesis and assists in pinpointing new early diagnostic markers and therapeutic targets. Utilizing advanced genomic tools and computational techniques, the research enhances our understanding of T cell dynamics in melanoma, facilitating the development of personalized medicine and more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Wen-Fei Luo
- Jinzhou Medical University, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ding-Ming Song
- Department of Urology, Jinzhou Medical University, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Tong Shen
- Department of Urology, Jinzhou Medical University, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yi-Bo He
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Hong-Yang Du
- Department of Dermatology, Jinzhou Medical University, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.
| | - Ming-Jue Si
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lu-Wei Fang
- Department of Dermatology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Rosenthal KJ, Gordan JD, Scott JD. Protein kinase A and local signaling in cancer. Biochem J 2024; 481:1659-1677. [PMID: 39540434 PMCID: PMC11975432 DOI: 10.1042/bcj20230352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Protein kinase A (PKA) is a basophilic kinase implicated in the modulation of many cell-signaling and physiological processes. PKA also contributes to cancer-relevant events such as growth factor action, cell cycle control, cell migration and tumor metabolism. Germline and somatic mutations in PKA, gene amplifications, and chromosome rearrangements that encode kinase fusions, are linked to a growing number of malignant neoplasms. Mislocalization of PKA by exclusion from A-Kinase Anchoring Protein (AKAP) signaling islands further underlies cancer progression. This article highlights the influence of AKAP signaling and local kinase action in selected hallmarks of cancer. We also feature the utility of kinase inhibitor drugs as frontline and future anti-cancer therapies.
Collapse
Affiliation(s)
- Kacey J. Rosenthal
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St., Box 357750, Seattle, WA 98195, U.S.A
| | - John D. Gordan
- Department of Medicine (Hematology/Oncology), Quantitative Biosciences Institute, UCSF Helen Diller Family Cancer Center, 1700 4th St., San Francisco, CA 94143, U.S.A
| | - John D. Scott
- Department of Pharmacology, University of Washington School of Medicine, 1959 NE Pacific St., Box 357750, Seattle, WA 98195, U.S.A
| |
Collapse
|
4
|
Hu M, Coleman S, Judson-Torres RL, Tan AC. The classification of melanocytic gene signatures. Pigment Cell Melanoma Res 2024; 37:854-863. [PMID: 39072997 PMCID: PMC11518649 DOI: 10.1111/pcmr.13189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/11/2024] [Accepted: 07/06/2024] [Indexed: 07/30/2024]
Abstract
Gene expression profiling technologies have revolutionized cell biology, enabling researchers to identify gene signatures linked to various biological attributes of melanomas, such as pigmentation status, differentiation state, proliferative versus invasive capacity, and disease progression. Although the discovery of gene signatures has significantly enhanced our understanding of melanocytic phenotypes, reconciling the numerous signatures reported across independent studies and different profiling platforms remains a challenge. Current methods for classifying melanocytic gene signatures depend on exact gene overlap and comparison with unstandardized baseline transcriptomes. In this study, we aimed to categorize published gene signatures into clusters based on their similar patterns of expression across clinical cutaneous melanoma specimens. We analyzed nearly 800 melanoma samples from six gene expression repositories and developed a classification framework for gene signatures that is resilient against biases in gene identification across profiling platforms and inconsistencies in baseline standards. Using 39 frequently cited published gene signatures, our analysis revealed seven principal classes of gene signatures that correlate with previously identified phenotypes: Differentiated, Mitotic/MYC, AXL, Amelanotic, Neuro, Hypometabolic, and Invasive. Each class is consistent with the phenotypes that the constituent gene signatures represent, and our classification method does not rely on overlapping genes between signatures. To facilitate broader application, we created WIMMS (what is my melanocytic signature, available at https://wimms.tanlab.org/), a user-friendly web application. WIMMS allows users to categorize any gene signature, determining its relationship to predominantly cited signatures and its representation within the seven principal classes.
Collapse
Affiliation(s)
- Min Hu
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Samuel Coleman
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, USA
| | - Robert L Judson-Torres
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Aik Choon Tan
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| |
Collapse
|
5
|
Zhang N, Yang M, Yang JM, Zhang CY, Guo AY. A Predictive Network-Based Immune Checkpoint Blockade Immunotherapeutic Signature Optimizing Patient Selection and Treatment Strategies. SMALL METHODS 2024; 8:e2301685. [PMID: 38546036 DOI: 10.1002/smtd.202301685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/01/2024] [Indexed: 10/18/2024]
Abstract
Immune checkpoint blockade (ICB) therapy has brought significant advancements to the field of oncology. However, the diverse responses among patients highlight the need for more accurate predictive tools. In this study, insights are drawn from tumor-immunology pathways, and a novel network-based ICB immunotherapeutic signature, termed ICBnetIS, is constructed. The signature is derived from advanced biological network-based computational strategies involving co-expression networks and molecular interactions networks. The efficacy of ICBnetIS is established through its association with enhanced patient survival and a robust immune response characterized by diverse immune cell infiltration and active anti-tumor immune pathways. The validation process positions ICBnetIS as an effective tool in predicting responses to ICB therapy, analyzing ICB data from a broad collection of over 700 samples from multiple cancer types of more than 15 datasets. It achieves an aggregated prediction AUC of 0.784, which outperforms the other nine renowned immunotherapeutic signatures, indicating the superior predictive capability of ICBnetIS. To sum up, the findings suggest ICBnetIS as a potent tool in predicting ICB therapy responses, offering significant implications for patient selection and treatment optimization in oncology. The study highlights the role of ICBnetIS in advancing personalized treatment strategies, potentially transforming the clinical landscape of ICB therapy.
Collapse
Affiliation(s)
- Nan Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, 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
| | - Mei Yang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, 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
| | - Jing-Min Yang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, 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
| | - Chu-Yu Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, 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
| | - An-Yuan Guo
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, 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
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
6
|
Khan DA, Adhikary T, Sultana MT, Toukir IA. A comprehensive identification of potential molecular targets and small drugs candidate for melanoma cancer using bioinformatics and network-based screening approach. J Biomol Struct Dyn 2024; 42:7349-7369. [PMID: 37534476 DOI: 10.1080/07391102.2023.2240409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023]
Abstract
Melanoma is the third most common malignant skin tumor and has increased in morbidity and mortality over the previous decade due to its rapid spread into the bloodstream or lymphatic system. This study used integrated bioinformatics and network-based methodologies to reliably identify molecular targets and small molecular medicines that may be more successful for Melanoma diagnosis, prognosis and treatment. The statistical LIMMA approach utilized for bioinformatics analysis in this study found 246 common differentially expressed genes (cDEGs) between case and control samples from two microarray gene-expression datasets (GSE130244 and GSE15605). Protein-protein interaction network study revealed 15 cDEGs (PTK2, STAT1, PNO1, CXCR4, WASL, FN1, RUNX2, SOCS3, ITGA4, GNG2, CDK6, BRAF, AGO2, GTF2H1 and AR) to be critical in the development of melanoma (KGs). According to regulatory network analysis, the most important transcriptional and post-transcriptional regulators of DEGs and hub-DEGs are ten transcription factors and three miRNAs. We discovered the pathogenetic mechanisms of MC by studying DEGs' biological processes, molecular function, cellular components and KEGG pathways. We used molecular docking and dynamics modeling to select the four most expressed genes responsible for melanoma malignancy to identify therapeutic candidates. Then, utilizing the Connectivity Map (CMap) database, we analyzed the top 4-hub-DEGs-guided repurposable drugs. We validated four melanoma cancer drugs (Fisetin, Epicatechin Gallate, 1237586-97-8 and PF 431396) using molecular dynamics simulation with their target proteins. As a result, the results of this study may provide resources to researchers and medical professionals for the wet-lab validation of MC diagnosis, prognosis and treatments.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Dhrubo Ahmed Khan
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Tonmoy Adhikary
- Department of Mathematics, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Mst Tania Sultana
- Department of Mathematics, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Imran Ahamed Toukir
- Department of Chemical Engineering, Jashore University of Science and Technology, Jashore, Bangladesh
| |
Collapse
|
7
|
Cui L, Yang L, Lai B, Luo L, Deng H, Chen Z, Wang Z. Integrative and comprehensive pan-cancer analysis of ubiquitin specific peptidase 11 ( USP11) as a prognostic and immunological biomarker. Heliyon 2024; 10:e34523. [PMID: 39114046 PMCID: PMC11305246 DOI: 10.1016/j.heliyon.2024.e34523] [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: 02/03/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
The significance of USP11 as a critical regulator in cancer has garnered substantial attention, primarily due to its catalytic activity as a deubiquitinating enzyme. Nonetheless, a thorough evaluation of USP11 across various cancer types in pan-cancer studies remains absent. Our analysis integrates data from a variety of sources, including five immunotherapy cohorts, thirty-three cohorts from The Cancer Genome Atlas (TCGA), and sixteen cohorts from the Gene Expression Omnibus (GEO), two of which involve single-cell transcriptomic data. Our findings indicate that aberrant USP11 expression is predictive of survival outcomes across various cancer types. The highest frequency of genomic alterations was observed in uterine corpus endometrial carcinoma (UCEC), with single-cell transcriptome analysis revealing significantly higher USP11 expression in plasmacytoid dendritic cells and mast cells. Notably, USP11 expression was associated with the infiltration levels of CD8+ T cells and natural killer (NK) activated cells. Additionally, in the skin cutaneous melanoma (SKCM) phs000452 cohort, patients with higher USP11 mRNA levels during immunotherapy experienced a significantly shorter median progression-free survival. USP11 emerges as a promising molecular biomarker with significant potential for predicting patient prognosis and immunoreactivity across various cancer types.
Collapse
Affiliation(s)
- Lijuan Cui
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Ling Yang
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Boan Lai
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Lingzhi Luo
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Haoyue Deng
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Zhongyi Chen
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| | - Zixing Wang
- Pathology Department, Suining Central Hospital, Suining, Sichuan, 629000, China
| |
Collapse
|
8
|
Xiang H, Luo R, Wang Y, Yang B, Xu S, Huang W, Tang S, Fang R, Chen L, Zhu N, Yu Z, Akesu S, Wei C, Xu C, Zhou Y, Gu J, Zhao J, Hou Y, Ding C. Proteogenomic insights into the biology and treatment of pan-melanoma. Cell Discov 2024; 10:78. [PMID: 39039072 PMCID: PMC11263678 DOI: 10.1038/s41421-024-00688-7] [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: 08/04/2023] [Accepted: 05/03/2024] [Indexed: 07/24/2024] Open
Abstract
Melanoma is one of the most prevalent skin cancers, with high metastatic rates and poor prognosis. Understanding its molecular pathogenesis is crucial for improving its diagnosis and treatment. Integrated analysis of multi-omics data from 207 treatment-naïve melanomas (primary-cutaneous-melanomas (CM, n = 28), primary-acral-melanomas (AM, n = 81), primary-mucosal-melanomas (MM, n = 28), metastatic-melanomas (n = 27), and nevi (n = 43)) provides insights into melanoma biology. Multivariate analysis reveals that PRKDC amplification is a prognostic molecule for melanomas. Further proteogenomic analysis combined with functional experiments reveals that the cis-effect of PRKDC amplification may lead to tumor proliferation through the activation of DNA repair and folate metabolism pathways. Proteome-based stratification of primary melanomas defines three prognosis-related subtypes, namely, the ECM subtype, angiogenesis subtype (with a high metastasis rate), and cell proliferation subtype, which provides an essential framework for the utilization of specific targeted therapies for particular melanoma subtypes. The immune classification identifies three immune subtypes. Further analysis combined with an independent anti-PD-1 treatment cohort reveals that upregulation of the MAPK7-NFKB signaling pathway may facilitate T-cell recruitment and increase the sensitivity of patients to immunotherapy. In contrast, PRKDC may reduce the sensitivity of melanoma patients to immunotherapy by promoting DNA repair in melanoma cells. These results emphasize the clinical value of multi-omics data and have the potential to improve the understanding of melanoma treatment.
Collapse
Affiliation(s)
- Hang Xiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunzhi Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bing Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sha Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wen Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shaoshuai Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rundong Fang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lingli Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Na Zhu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zixiang Yu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sujie Akesu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuanyuan Wei
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianying Gu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital (Xiamen), Fudan University, Shanghai, China.
| | - Jianyuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Guo Y, Kasai Y, Tanaka Y, Ohashi‐Kumagai Y, Sakamoto T, Ito T, Murakami Y. IGSF3 is a homophilic cell adhesion molecule that drives lung metastasis of melanoma by promoting adhesion to vascular endothelium. Cancer Sci 2024; 115:1936-1947. [PMID: 38590281 PMCID: PMC11145127 DOI: 10.1111/cas.16166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
The immunoglobulin superfamily (IgSF) is one of the largest families of cell-surface molecules involved in various cell-cell interactions, including cancer-stromal interactions. In this study, we undertook a comprehensive RT-PCR-based screening for IgSF molecules that promote experimental lung metastasis in mice. By comparing the expression of 325 genes encoding cell-surface IgSF molecules between mouse melanoma B16 cells and its highly metastatic subline, B16F10 cells, we found that expression of the immunoglobulin superfamily member 3 gene (Igsf3) was significantly enhanced in B16F10 cells than in B16 cells. Knockdown of Igsf3 in B16F10 cells significantly reduced lung metastasis following intravenous injection into C57BL/6 mice. IGSF3 promoted adhesion of B16F10 cells to vascular endothelial cells and functioned as a homophilic cell adhesion molecule between B16F10 cells and vascular endothelial cells. Notably, the knockdown of IGSF3 in either B16F10 cells or vascular endothelial cells suppressed the transendothelial migration of B16F10 cells. Moreover, IGSF3 knockdown suppressed the extravasation of B16F10 cells into the lungs after intravenous injection. These results suggest that IGSF3 promotes the metastatic potential of B16F10 cells in the lungs by facilitating their adhesion to vascular endothelial cells.
Collapse
Affiliation(s)
- Yue Guo
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yutaka Kasai
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yuto Tanaka
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yuki Ohashi‐Kumagai
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Takeharu Sakamoto
- Department of Cancer BiologyInstitute of Biomedical Science, Kansai Medical UniversityHirakataJapan
| | - Takeshi Ito
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Yoshinori Murakami
- Division of Molecular PathologyThe Institute of Medical Science, The University of TokyoTokyoJapan
| |
Collapse
|
10
|
Sahin Y, Pei J, Baldwin DA, Mansoor N, Koslosky L, Abdelmessieh P, Wang YL, Nejati R, Testa JR. Acute myeloid leukemia with a novel AKAP9::PDGFRA fusion transformed from essential thrombocythemia: A case report and mini review. Leuk Res Rep 2024; 21:100465. [PMID: 38952949 PMCID: PMC11215950 DOI: 10.1016/j.lrr.2024.100465] [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: 02/07/2024] [Revised: 05/07/2024] [Accepted: 05/30/2024] [Indexed: 07/03/2024] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy associated with various combinations of gene mutations, epigenetic abnormalities, and chromosome rearrangement-related gene fusions. Despite the significant degree of heterogeneity in its pathogenesis, many gene fusions and point mutations are recurrent in AML and have been employed in risk stratification over the last several decades. Gene fusions have long been recognized for understanding tumorigenesis and their proven roles in clinical diagnosis and targeted therapies. Advances in DNA sequencing technologies and computational biology have contributed significantly to the detection of known fusion genes as well as for the discovery of novel ones. Several recurring gene fusions in AML have been linked to prognosis, treatment response, and disease progression. In this report, we present a case with a long history of essential thrombocythemia and hallmark CALR mutation transforming to AML characterized by a previously unreported AKAP9::PDGFRA fusion gene. We propose mechanisms by which this fusion may contribute to the pathogenesis of AML and its potential as a molecular target for tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Yavuz Sahin
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jianming Pei
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Don A. Baldwin
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Nashwa Mansoor
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Lori Koslosky
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Peter Abdelmessieh
- Department of Bone Marrow Transplant and Cellular Therapies, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Y. Lynn Wang
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Reza Nejati
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Joseph. R. Testa
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| |
Collapse
|
11
|
Agrawal P, Chen S, de Pablos A, Jame-Chenarboo F, Miera Saenz de Vega E, Darvishian F, Osman I, Lujambio A, Mahal LK, Hernando E. Integrated in vivo functional screens and multi-omics analyses identify α-2,3-sialylation as essential for melanoma maintenance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584072. [PMID: 38559078 PMCID: PMC10979837 DOI: 10.1101/2024.03.08.584072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Glycosylation is a hallmark of cancer biology, and altered glycosylation influences multiple facets of melanoma growth and progression. To identify glycosyltransferases, glycans, and glycoproteins essential for melanoma maintenance, we conducted an in vivo growth screen with a pooled shRNA library of glycosyltransferases, lectin microarray profiling of benign nevi and melanoma patient samples, and mass spectrometry-based glycoproteomics. We found that α-2,3 sialyltransferases ST3GAL1 and ST3GAL2 and corresponding α-2,3-linked sialosides are upregulated in melanoma compared to nevi and are essential for melanoma growth in vivo and in vitro. Glycoproteomics revealed that glycoprotein targets of ST3GAL1 and ST3GAL2 are enriched in transmembrane proteins involved in growth signaling, including the amino acid transporter Solute Carrier Family 3 Member 2 (SLC3A2/CD98hc). CD98hc suppression mimicked the effect of ST3GAL1 and ST3GAL2 silencing, inhibiting melanoma cell proliferation. We found that both CD98hc protein stability and its pro-survival effect in melanoma are dependent upon α-2,3 sialylation mediated by ST3GAL1 and ST3GAL2. In summary, our studies reveal that α-2,3-sialosides functionally contribute to melanoma maintenance, supporting ST3GAL1 and ST3GAL2 as novel therapeutic targets in these tumors.
Collapse
Affiliation(s)
- Praveen Agrawal
- Department of Pathology, NYU Grossman School of Medicine, New York
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| | - Shuhui Chen
- Department of Chemistry, New York University
| | - Ana de Pablos
- Department of Pathology, NYU Grossman School of Medicine, New York
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
- Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
| | | | | | | | - Iman Osman
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
- Department of Dermatology, NYU Grossman School of Medicine, New York
| | | | - Lara K. Mahal
- Department of Chemistry, New York University
- Department of Chemistry, University of Alberta, Edmonton, Canada
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health
| |
Collapse
|
12
|
Ramos-Rodriguez AJ, McFadden JR, Momtahen S, LeBlanc RE, Yan S, Chaudhari AS, Cloutier JM, Stevanovic M, Barney R, Syku M, Lozano-Franco M, Hughes E, Sriharan A. A novel method to assess copy number variations in melanocytic neoplasms: Droplet digital PCR for precise quantitation of MYC and MYB genes. J Cutan Pathol 2024; 51:146-154. [PMID: 37795541 PMCID: PMC10863652 DOI: 10.1111/cup.14540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023]
Abstract
INTRODUCTION While most melanocytic neoplasms can be classified as either benign or malignant by histopathology alone, ancillary molecular diagnostic tests can be necessary to establish the correct diagnosis in challenging cases. Currently, the detection of copy number variations (CNVs) by fluorescence in situ hybridization and chromosomal microarray (CMA) are the most popular methods, but remain expensive and inaccessible. We aim to develop a relatively inexpensive, fast, and accessible molecular assay to detect CNVs relevant to melanoma using droplet digital polymerase chain reaction (ddPCR) technology. METHODS In this proof-of-concept study, we evaluated CNVs in MYC and MYB genes from 73 cases of benign nevi, borderline melanocytic lesions, and primary and metastatic melanoma at our institution from 2015 to 2022. A multiplexed ddPCR assay and CMA were performed on each sample, and the results were compared. RESULTS Concordance analysis of ddPCR with CMA for quantification of MYC and MYB CNVs revealed a sensitivity and specificity of 89% and 86% for MYC and 83% and 74% for MYB, respectively. CONCLUSION We demonstrate the first use of a multiplexed ddPCR assay to identify CNVs in melanocytic neoplasms. With further improvement and validation, ddPCR may represent a low-cost and rapid tool to aid in the diagnosis of histopathologically ambiguous melanocytic tumors.
Collapse
Affiliation(s)
- Alvaro J. Ramos-Rodriguez
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766
| | | | - Shabnam Momtahen
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766
| | - Robert E. LeBlanc
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766
| | - Shaofeng Yan
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766
| | | | - Jeffrey M. Cloutier
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766
| | | | - Rachael Barney
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766
- Clinical Genomics and Advanced Technology Laboratory, Dartmouth-Hitchcock Medical Center, Lebanon NH 03766
| | - Marie Syku
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766
| | | | - Edward Hughes
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766
- Clinical Genomics and Advanced Technology Laboratory, Dartmouth-Hitchcock Medical Center, Lebanon NH 03766
| | - Aravindhan Sriharan
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766
| |
Collapse
|
13
|
Berico P, Nogaret M, Cigrang M, Lallement A, Vand-Rajabpour F, Flores-Yanke A, Gambi G, Davidson G, Seno L, Obid J, Vokshi BH, Le Gras S, Mengus G, Ye T, Cordero CF, Dalmasso M, Compe E, Bertolotto C, Hernando E, Davidson I, Coin F. Super-enhancer-driven expression of BAHCC1 promotes melanoma cell proliferation and genome stability. Cell Rep 2023; 42:113363. [PMID: 37924516 DOI: 10.1016/j.celrep.2023.113363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 07/27/2023] [Accepted: 10/16/2023] [Indexed: 11/06/2023] Open
Abstract
Super-enhancers (SEs) are stretches of enhancers ensuring a high level of expression of key genes associated with cell function. The identification of cancer-specific SE-driven genes is a powerful means for the development of innovative therapeutic strategies. Here, we identify a MITF/SOX10/TFIIH-dependent SE promoting the expression of BAHCC1 in a broad panel of melanoma cells. BAHCC1 is highly expressed in metastatic melanoma and is required for tumor engraftment, growth, and dissemination. Integrative genomics analyses reveal that BAHCC1 is a transcriptional regulator controlling expression of E2F/KLF-dependent cell-cycle and DNA-repair genes. BAHCC1 associates with BRG1-containing remodeling complexes at the promoters of these genes. BAHCC1 silencing leads to decreased cell proliferation and delayed DNA repair. Consequently, BAHCC1 deficiency cooperates with PARP inhibition to induce melanoma cell death. Our study identifies BAHCC1 as an SE-driven gene expressed in melanoma and demonstrates how its inhibition can be exploited as a therapeutic target.
Collapse
Affiliation(s)
- Pietro Berico
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France; Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Maguelone Nogaret
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Max Cigrang
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Antonin Lallement
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Fatemeh Vand-Rajabpour
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Amanda Flores-Yanke
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Giovanni Gambi
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Guillaume Davidson
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Leane Seno
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Julian Obid
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Bujamin H Vokshi
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Stephanie Le Gras
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Gabrielle Mengus
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Tao Ye
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Carlos Fernandez Cordero
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Mélanie Dalmasso
- Université Côte d'Azur, Nice, France; INSERM, Biology and Pathologies of Melanocytes, Equipe labellisée "Ligue contre le Cancer 2020" and Equipe labellisée "Fondation ARC 2022", Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Emmanuel Compe
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Corine Bertolotto
- Université Côte d'Azur, Nice, France; INSERM, Biology and Pathologies of Melanocytes, Equipe labellisée "Ligue contre le Cancer 2020" and Equipe labellisée "Fondation ARC 2022", Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Eva Hernando
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Irwin Davidson
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France.
| | - Frédéric Coin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Equipe Labéllisée, "Ligue contre le Cancer 2022", BP 163, 67404 Illkirch Cedex, C.U. Strasbourg, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U1258, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France.
| |
Collapse
|
14
|
Singh N, Khan FM, Bala L, Vera J, Wolkenhauer O, Pützer B, Logotheti S, Gupta SK. Logic-based modeling and drug repurposing for the prediction of novel therapeutic targets and combination regimens against E2F1-driven melanoma progression. BMC Chem 2023; 17:161. [PMID: 37993971 PMCID: PMC10666365 DOI: 10.1186/s13065-023-01082-2] [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: 06/12/2023] [Accepted: 11/08/2023] [Indexed: 11/24/2023] Open
Abstract
Melanoma presents increasing prevalence and poor outcomes. Progression to aggressive stages is characterized by overexpression of the transcription factor E2F1 and activation of downstream prometastatic gene regulatory networks (GRNs). Appropriate therapeutic manipulation of the E2F1-governed GRNs holds the potential to prevent metastasis however, these networks entail complex feedback and feedforward regulatory motifs among various regulatory layers, which make it difficult to identify druggable components. To this end, computational approaches such as mathematical modeling and virtual screening are important tools to unveil the dynamics of these signaling networks and identify critical components that could be further explored as therapeutic targets. Herein, we integrated a well-established E2F1-mediated epithelial-mesenchymal transition (EMT) map with transcriptomics data from E2F1-expressing melanoma cells to reconstruct a core regulatory network underlying aggressive melanoma. Using logic-based in silico perturbation experiments of a core regulatory network, we identified that simultaneous perturbation of Protein kinase B (AKT1) and oncoprotein murine double minute 2 (MDM2) drastically reduces EMT in melanoma. Using the structures of the two protein signatures, virtual screening strategies were performed with the FDA-approved drug library. Furthermore, by combining drug repurposing and computer-aided drug design techniques, followed by molecular dynamics simulation analysis, we identified two potent drugs (Tadalafil and Finasteride) that can efficiently inhibit AKT1 and MDM2 proteins. We propose that these two drugs could be considered for the development of therapeutic strategies for the management of aggressive melanoma.
Collapse
Affiliation(s)
- Nivedita Singh
- Department of Biochemistry, BBDCODS, BBD University, Lucknow, Uttar Pradesh, India
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Faiz M Khan
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
| | - Lakshmi Bala
- Department of Biochemistry, BBDCODS, BBD University, Lucknow, Uttar Pradesh, India
| | - Julio Vera
- Department of Dermatology, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany
- Leibniz Institute for Food Systems Biology, Technical University of Munich, Munich, Germany
- Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India
- Stellenbosch Institute of Advanced Study, Wallenberg Research Centre, Stellenbosch University, Stellenbosch, South Africa
| | - Brigitte Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
| | - Stella Logotheti
- Institute of Experimental Gene Therapy and Cancer Research, Rostock University Medical Center, Rostock, Germany
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, Athens, Greece
| | - Shailendra K Gupta
- Department of Systems Biology and Bioinformatics, University of Rostock, Rostock, Germany.
- Chhattisgarh Swami Vivekanand Technical University, Bhilai, Chhattisgarh, India.
| |
Collapse
|
15
|
Liu LP, Liu N, Zhang HN, Li D. Exploring and validating the metastasis mechanism of pathenolide interfering with cutaneous melanoma through ER stress-dependent apoptosis based on the network pharmacology. PHYTOCHEMICAL ANALYSIS : PCA 2023; 34:745-754. [PMID: 36477977 DOI: 10.1002/pca.3193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/06/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND This study aimed to explore the mechanism of parthenolide in inhibiting melanoma metastasis through network pharmacology and cell experiment. MATERIALS AND METHODS This research obtained the targets of the drug from the HERB database and PubChem database, the differential expression gene of metastatic cutaneous melanoma was obtained by differentially expression gene analysis of four Gene Expression Omnibus (GEO) datasets. The intersection of drug targets and differentially expression genes were considered to be related to drugs that inhibit metastasis of cutaneous melanoma. The STRING database was used to construct the protein-protein interaction (PPI) network, and cytohubba package in Cytoscape software was used to rank the PPI network targets. The enrichment analysis was used to screen out the relevance Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and gene ontology to explain the underlying mechanism of drug inhibiting the cutaneous melanoma metastatic; cell viability, apoptosis, cell migration and protein levels were assessed using cell counting kit-8 (CCK-8) assay, Annexin V-FITC/PI assay, wound healing assay, respectively. Finally, combining pathway maps and literature, we detected ATF4 and proteins upstream and downstream of ATF4 through Western blot. RESULTS A total of 87 targets were screened out from the drug databases, and a total of 1635 differentially expression genes was obtained from the differentially expression genes analysis of GEO datasets, a total of nine targets (VEGFA, ANXA5, ICAM1, SELE, NFKBIA, ATF4, CTNNB1, SELP and HPGDS) were considered to be related to drugs that inhibit metastasis of cutaneous melanoma. The result of enrichment analysis showed that the drug inhibits the metastatic of cutaneous melanoma through multiple pathways such as TNF signalling pathway, lipid and atherosclerosis and fluid shear stress and atherosclerosis, relevance multiple biological processes, cellular components and molecular function; cell experiments showed that parthenolide could inhibit tumour cell migration and induce a decrease of cell viability. Flow cytometry results showed that parthenolide induced tumour cell apoptosis. Western blot results suggested that parthenolide exerted therapeutic effects by regulating ATF4 protein and its upstream and downstream proteins, namely endoplasmic reticulum (ER) stress signalling pathway. CONCLUSION Parthenolide induces ER stress-dependent apoptosis in melanoma cells.
Collapse
Affiliation(s)
- Li-Ping Liu
- Institute of Integrated Medicine, Medical College, Qingdao University, Qingdao, Shandong, China
| | - Ning Liu
- Department of Dermatology, the Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong, China
| | - Han-Ni Zhang
- Department of Acupuncture, Wuhan Puren Hospital, Wuhan, Hubei, China
| | - Ding Li
- Institute of Integrated Medicine, Medical College, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
16
|
Song L, Wei X, Zhang X, Lu Y. Combining single-cell and transcriptomic analysis revealed the immunomodulatory effect of GOT2 on a glutamine-dependent manner in cutaneous melanoma. Front Pharmacol 2023; 14:1241454. [PMID: 37693904 PMCID: PMC10483140 DOI: 10.3389/fphar.2023.1241454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023] Open
Abstract
Background: Reprogramming in glutamine metabolism is a hallmark of cancers, while its role in cutaneous melanoma has not been studied at great length. Methods: Here, we constructed a glutamine metabolism-related prognostic signature in cutaneous melanoma with a variety of bioinformatics methods according to the glutamine metabolism regulatory molecules. Moreover, experimental verification was carried out for the key gene. Results: We have identified two subgroups of cutaneous melanoma patients, each with different prognoses, immune characteristics, and genetic mutations. GOT2 was the most concerned key gene among the model genes. We verified its role in promoting tumor cell proliferation by CCK-8 and clone formation assays. Conclusion: Our study cast new light on the prognosis of cutaneous melanoma, and the internal mechanism regulating glutamine metabolism of GOT2 may provide a new avenue for treating the cutaneous melanoma disease precisely.
Collapse
Affiliation(s)
- Lebin Song
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiyi Wei
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xi Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Lu
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
McFadden JR, Syku M, Barney RE, Stevanovic M, Chaudhari AS, O’Hern KJ, Chambers M, Baker CM, LeBlanc RE, Doan L, Tsongalis GJ, Hughes EG, Sriharan A. A Novel Method to Detect Copy Number Variation in Melanoma: Droplet Digital PCR for Quantitation of the CDKN2A Gene, a Proof-of-Concept Study. Am J Dermatopathol 2023; 45:454-462. [PMID: 37130203 PMCID: PMC10993871 DOI: 10.1097/dad.0000000000002436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
ABSTRACT A definitive diagnosis of nevus or melanoma is not always possible for histologically ambiguous melanocytic neoplasms. In such cases, ancillary molecular testing can support a diagnosis of melanoma if certain chromosomal aberrations are detected. Current technologies for copy number variation (CNV) detection include chromosomal microarray analysis (CMA) and fluorescence in situ hybridization. Although CMA and fluorescence in situ hybridization are effective, their utilization can be limited by cost, turnaround time, and inaccessibility outside of large reference laboratories. Droplet digital polymerase chain reaction (ddPCR) is a rapid, automated, and relatively inexpensive technology for CNV detection. We investigated the ability of ddPCR to quantify CNV in cyclin-dependent kinase inhibitor 2A ( CDKN2A ), the most commonly deleted tumor suppressor gene in melanoma. CMA data were used as the gold standard. We analyzed 57 skin samples from 52 patients diagnosed with benign nevi, borderline lesions, primary melanomas, and metastatic melanomas. In a training cohort comprising 29 randomly selected samples, receiver operator characteristic curve analysis revealed an optimal ddPCR cutoff value of 1.73 for calling CDKN2A loss. In a validation cohort comprising the remaining 28 samples, ddPCR detected CDKN2A loss with a sensitivity and specificity of 94% and 90%, respectively. Significantly, ddPCR could also identify whether CDKN2A losses were monoallelic or biallelic. These pilot data suggest that ddPCR can detect CDKN2A deletions in melanocytic tumors with accuracy comparable with CMA. With further validation, ddPCR could provide an additional CNV assay to aid in the diagnosis of challenging melanocytic neoplasms.
Collapse
Affiliation(s)
- Jason R. McFadden
- Department of Biological Sciences, Dartmouth College, Hanover, NH, NH
| | - Marie Syku
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Rachael E. Barney
- Clinical Genomics and Advanced Technologies (CGAT) Laboratory, Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Mirjana Stevanovic
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH
| | | | - Keegan J. O’Hern
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Meagan Chambers
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Catherine M. Baker
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Robert E. LeBlanc
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Linda Doan
- Department of Dermatology, University of California, Irvine Health (UCI Health), Irvine, CA
- Department of Pathology & Laboratory Services, University of California, Irvine Health (UCI Health), Irvine, CA
| | - Gregory J. Tsongalis
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH
- Clinical Genomics and Advanced Technologies (CGAT) Laboratory, Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Edward G. Hughes
- Clinical Genomics and Advanced Technologies (CGAT) Laboratory, Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Aravindhan Sriharan
- Department of Pathology and Laboratory Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| |
Collapse
|
18
|
Zia A, Litvin Y, Voskoboynik R, Klein A, Shachaf C. Transcriptome Analysis Identifies Oncogenic Tissue Remodeling during Progression from Common Nevi to Early Melanoma. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:995-1004. [PMID: 37146966 DOI: 10.1016/j.ajpath.2023.03.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/09/2023] [Accepted: 03/27/2023] [Indexed: 05/07/2023]
Abstract
Early detection and treatment of melanoma, the most aggressive skin cancer, improves the median 5-year survival rate of patients from 25% to 99%. Melanoma development involves a stepwise process during which genetic changes drive histologic alterations within nevi and surrounding tissue. Herein, a comprehensive analysis of publicly available gene expression data sets of melanoma, common or congenital nevi (CN), and dysplastic nevi (DN), assessed molecular and genetic pathways leading to early melanoma. The results demonstrate several pathways reflective of ongoing local structural tissue remodeling activity likely involved during the transition from benign to early-stage melanoma. These processes include the gene expression of cancer-associated fibroblasts, collagens, extracellular matrix, and integrins, which assist early melanoma development and the immune surveillance that plays a substantial role at this early stage. Furthermore, genes up-regulated in DN were also overexpressed in melanoma tissue, supporting the notion that DN may serve as a transitional phase toward oncogenesis. CN collected from healthy individuals exhibited different gene signatures compared with histologically benign nevi tissue located adjacent to melanoma (adjacent nevi). Finally, the expression profile of microdissected adjacent nevi tissue was more similar to melanoma compared with CN, revealing the melanoma influence on this annexed tissue.
Collapse
Affiliation(s)
- Amin Zia
- Orlucent, Inc., Los Gatos, California
| | | | | | - Amit Klein
- Department of Bioengineering: Bioinformatics, University of California, San Diego, San Diego, California
| | | |
Collapse
|
19
|
Azimi A, Patrick E, Teh R, Kim J, Fernandez-Penas P. Proteomic profiling of cutaneous melanoma explains the aggressiveness of distant organ metastasis. Exp Dermatol 2023. [PMID: 37082900 DOI: 10.1111/exd.14814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/22/2023]
Abstract
Despite recent developments in managing metastatic melanomas, patients' overall survival remains low. Therefore, the current study aims to understand better the proteome-wide changes associated with melanoma metastasis that will assist with identifying targeted therapies. The latest development in mass spectrometry-based proteomics, together with extensive bioinformatics analysis, was used to investigate the molecular changes in 60 formalin-fixed and paraffin-embedded samples of primary and lymph nodes (LN) and distant organ metastatic melanomas. A total of 4631 proteins were identified, of which 72 and 453 were significantly changed between the LN and distant organ metastatic melanomas compared to the primary lesions (adj. p-value <0.05). An increase in proteins such as SLC9A3R1, CD20 and GRB2 and a decrease in CST6, SERPINB5 and ARG1 were associated with regional LN metastasis. By contrast, increased metastatic activities in distant organ metastatic melanomas were related to higher levels of CEACAM1, MC1R, AKT1 and MMP3-9 and decreased levels of CDKN2A, SDC1 and SDC4 proteins. Furthermore, machine learning analysis classified the lesions with up to 92% accuracy based on their metastatic status. The findings from this study provide up to date proteome-level information about the progression of melanomas to regional LN and distant organs, leading to the identification of protein signatures with potential for clinical translation.
Collapse
Affiliation(s)
- Ali Azimi
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Department of Dermatology, Westmead Hospital, Westmead, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Ellis Patrick
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Camperdown, New South Wales, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Rachel Teh
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Department of Dermatology, Westmead Hospital, Westmead, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Jennifer Kim
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Pablo Fernandez-Penas
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Department of Dermatology, Westmead Hospital, Westmead, New South Wales, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| |
Collapse
|
20
|
Santamaria-Barria JA, Matsuba C, Khader A, Scholar AJ, Garland-Kledzik M, Fischer TD, Essner R, Salomon MP, Mammen JMV, Goldfarb M. Age-related next-generation sequencing mutational analysis in 1196 melanomas. J Surg Oncol 2023; 127:1187-1195. [PMID: 36938777 DOI: 10.1002/jso.27239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/11/2023] [Accepted: 03/07/2023] [Indexed: 03/21/2023]
Abstract
BACKGROUND AND OBJECTIVES Melanoma mutational burden is high and approximately 50% have oncogenic mutations in BRAF. We sought to evaluate age-related mutational differences in melanoma. METHODS We analyzed melanoma samples in the Genomics Evidence Neoplasia Information Exchange database. Targetable mutations were identified using the Precision Oncology Knowledge Base (OncoKB). RESULTS We found 1194 patients with a common set of 30 genes. The top mutated genes in patients <40 years old (y/o) (n = 98) were BRAF (59%), TP53 (31%), NRAS (17%), and PTEN (14%); in 40-59 y/o (n = 354) were BRAF (51%), NRAS (30%), TP53 (26%), and APC (13%); and in ≥60 y/o (n = 742) were BRAF (38%), NRAS (33%), TP53 (26%), and KDR (19%). BRAF mutations were almost mutually exclusive from NRAS mutations in <40 y/o (58/59). Mutational burden increased with age, with means of 2.39, 2.92, and 3.67 mutations per sample in patients <40, 40-59, and ≥60 y/o, respectively (p < 0.0001). There were 10 targetable mutations meeting OncoKB criteria for melanoma: BRAF (level 1), RET (level 1), KIT (level 2), NRAS (level 3A), TP53 (level 3A), and FGFR2, MET, PTEN, PIK3CA, and KRAS (level 4). CONCLUSIONS Mutations in melanoma have age-related differences and demonstrates potential targetable mutations for personalized therapies.
Collapse
Affiliation(s)
- Juan A Santamaria-Barria
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Chikako Matsuba
- Computational Biology Division, Saint John's Cancer Institute at Providence St. John's Health Center, Santa Monica, California, USA
| | - Adam Khader
- Division of Surgical Oncology, Department of Surgery, Hunter Holmes McGuire Veterans Affair Medical Center, Richmond, Virginia, USA
| | - Anthony J Scholar
- Division of Surgical Oncology, University of South Carolina School of Medicine, Greenville, South Carolina, USA
| | - Mary Garland-Kledzik
- Division of Surgical Oncology, West Virginia University, Morgantown, West Virginia, USA
| | - Trevan D Fischer
- Department of Surgery, Saint John's Cancer Institute at Providence St. John's Health Center, Santa Monica, California, USA
| | - Richard Essner
- Department of Surgery, Saint John's Cancer Institute at Providence St. John's Health Center, Santa Monica, California, USA
| | - Matthew P Salomon
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Joshua M V Mammen
- Division of Surgical Oncology, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Melanie Goldfarb
- Department of Surgery, Saint John's Cancer Institute at Providence St. John's Health Center, Santa Monica, California, USA
| |
Collapse
|
21
|
Xu X, Ju Y, Zhao X, Yang P, Zhu F, Fang B. SMG7-AS1 as a prognostic biomarker and predictor of immunotherapy responses for skin cutaneous melanoma. Genomics 2023; 115:110614. [PMID: 36931476 DOI: 10.1016/j.ygeno.2023.110614] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/14/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Skin cutaneous melanoma (SKCM) is the most life-threatening skin cancer and lacks early detection and effective treatment strategies. Many long noncoding RNAs are associated with the development of tumors and may serve as potential immunotherapeutic targets. In this study, microarray analysis was performed to screen for differentially expressed lncRNAs between SKCM and normal tissues, and SMG7-AS1 was identified as an upregulated lncRNA in SKCM. Subsequently, bioinformatic analysis revealed that dysregulation of SMG7-AS1 influences metastasis and immune infiltration. qRT-PCR of clinical samples demonstrated that the expression of SMG7-AS1 was higher in melanoma tissues. Flow cytometry showed that SMG7-AS1 plays a vital role in the cell cycle. Additionally, SMG7-AS1 was found to be associated with immunotherapy responses. To the best of our knowledge, this study is the first to report that SMG7-AS1 is associated with SKCM and may serve as a prognostic biomarker and predictor of immunotherapy responses in SKCM.
Collapse
Affiliation(s)
- Xuezheng Xu
- Department of Orthopaedics, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, People's Republic of China
| | - Yikun Ju
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Xueheng Zhao
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410078, People's Republic of China
| | - Pu Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Fang Zhu
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha 410078, People's Republic of China
| | - Bairong Fang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China.
| |
Collapse
|
22
|
Wang JX, Cao B, Ma N, Wu KY, Chen WB, Wu W, Dong X, Liu CF, Gao YF, Diao TY, Min XY, Yong Q, Li ZF, Zhou W, Li K. Collectin-11 promotes cancer cell proliferation and tumor growth. JCI Insight 2023; 8:e159452. [PMID: 36883567 PMCID: PMC10077485 DOI: 10.1172/jci.insight.159452] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 01/25/2023] [Indexed: 03/09/2023] Open
Abstract
Collectin-11 (CL-11) is a recently described soluble C-type lectin that has distinct roles in embryonic development, host defence, autoimmunity, and fibrosis. Here we report that CL-11 also plays an important role in cancer cell proliferation and tumor growth. Melanoma growth was found to be suppressed in Colec11-/- mice in a s.c. B16 melanoma model. Cellular and molecular analyses revealed that CL-11 is essential for melanoma cell proliferation, angiogenesis, establishment of more immunosuppressive tumor microenvironment, and the reprogramming of macrophages to M2 phenotype within melanomas. In vitro analysis revealed that CL-11 can activate tyrosine kinase receptors (EGFR, HER3) and ERK, JNK, and AKT signaling pathways and has a direct stimulatory effect on murine melanoma cell proliferation. Furthermore, blockade of CL-11 (treatment with L-fucose) inhibited melanoma growth in mice. Analysis of open data sets revealed that COLEC11 gene expression is upregulated in human melanomas and that high COLEC11 expression has a trend toward poor survival. CL-11 also had direct stimulatory effects on human tumor cell proliferation in melanoma and several other types of cancer cells in vitro. Overall, our findings provide the first evidence to our knowledge that CL-11 is a key tumor growth-promoting protein and a promising therapeutic target in tumor growth.
Collapse
Affiliation(s)
- Jia-Xing Wang
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Bo Cao
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Ning Ma
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Kun-Yi Wu
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Wan-Bing Chen
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Weiju Wu
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Xia Dong
- Department of Ophthalmology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Cheng-Fei Liu
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Ya-Feng Gao
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Teng-Yue Diao
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xiao-Yun Min
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qing Yong
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zong-Fang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
| | - Wuding Zhou
- Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Ke Li
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
23
|
Zhai Z, Yamauchi T, Shangraw S, Hou V, Matsumoto A, Fujita M. Ethanol Metabolism and Melanoma. Cancers (Basel) 2023; 15:1258. [PMID: 36831600 PMCID: PMC9954650 DOI: 10.3390/cancers15041258] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Malignant melanoma is the deadliest form of skin cancer. Despite significant efforts in sun protection education, melanoma incidence is still rising globally, drawing attention to other socioenvironmental risk factors for melanoma. Ethanol and acetaldehyde (AcAH) are ubiquitous in our diets, medicines, alcoholic beverages, and the environment. In the liver, ethanol is primarily oxidized to AcAH, a toxic intermediate capable of inducing tumors by forming adducts with proteins and DNA. Once in the blood, ethanol and AcAH can reach the skin. Although, like the liver, the skin has metabolic mechanisms to detoxify ethanol and AcAH, the risk of ethanol/AcAH-associated skin diseases increases when the metabolic enzymes become dysfunctional in the skin. This review highlights the evidence linking cutaneous ethanol metabolism and melanoma. We summarize various sources of skin ethanol and AcAH and describe how the reduced activity of each alcohol metabolizing enzyme affects the sensitivity threshold to ethanol/AcAH toxicity. Data from the Gene Expression Omnibus database also show that three ethanol metabolizing enzymes (alcohol dehydrogenase 1B, P450 2E1, and catalase) and an AcAH metabolizing enzyme (aldehyde dehydrogenase 2) are significantly reduced in melanoma tissues.
Collapse
Affiliation(s)
- Zili Zhai
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Takeshi Yamauchi
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarah Shangraw
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Vincent Hou
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Akiko Matsumoto
- Department of Social Medicine, School of Medicine, Saga University, Saga 849-8501, Japan
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO 80045, USA
| |
Collapse
|
24
|
Shoji KF, Bayet E, Leverrier-Penna S, Le Devedec D, Mallavialle A, Marionneau-Lambot S, Rambow F, Perret R, Joussaume A, Viel R, Fautrel A, Khammari A, Constantin B, Tartare-Deckert S, Penna A. The mechanosensitive TRPV2 calcium channel promotes human melanoma invasiveness and metastatic potential. EMBO Rep 2023; 24:e55069. [PMID: 36744297 PMCID: PMC10074106 DOI: 10.15252/embr.202255069] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 12/21/2022] [Accepted: 01/10/2023] [Indexed: 02/07/2023] Open
Abstract
Melanoma is a highly aggressive cancer endowed with a unique capacity of rapidly metastasizing, which is fundamentally driven by aberrant cell motility behaviors. Discovering "migrastatics" targets, specifically controlling invasion and dissemination of melanoma cells during metastasis, is therefore of primary importance. Here, we uncover the prominent expression of the plasma membrane TRPV2 calcium channel as a distinctive feature of melanoma tumors, directly related to melanoma metastatic dissemination. In vitro as well as in vivo, TRPV2 activity is sufficient to confer both migratory and invasive potentials, while conversely TRPV2 silencing in highly metastatic melanoma cells prevents aggressive behavior. In invasive melanoma cells, TRPV2 channel localizes at the leading edge, in dynamic nascent adhesions, and regulates calcium-mediated activation of calpain and the ensuing cleavage of the adhesive protein talin, along with F-actin organization. In human melanoma tissues, TRPV2 overexpression correlates with advanced malignancy and poor prognosis, evoking a biomarker potential. Hence, by regulating adhesion and motility, the mechanosensitive TRPV2 channel controls melanoma cell invasiveness, highlighting a new therapeutic option for migrastatics in the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Kenji F Shoji
- Inserm, EHESP, IRSET, UMR_S 1085, Université de Rennes 1, Rennes, France
| | - Elsa Bayet
- Inserm, EHESP, IRSET, UMR_S 1085, Université de Rennes 1, Rennes, France.,CNRS, 4CS, Université de Poitiers, Poitiers, France
| | | | - Dahiana Le Devedec
- Inserm, EHESP, IRSET, UMR_S 1085, Université de Rennes 1, Rennes, France
| | - Aude Mallavialle
- INSERM, C3M, team 'labellisée Ligue Contre le Cancer 2022, Université Côte d'Azur, Nice, France
| | | | - Florian Rambow
- Department of Applied Computational Cancer Research, Institute for AI in Medicine (IKIM), University Hospital Essen, Essen, Germany.,University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site Essen, Essen, Germany
| | - Raul Perret
- Service de Dermatologie, CHU Nantes, CIC 1413, INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes Université, Nantes, France
| | - Aurélie Joussaume
- Inserm, EHESP, IRSET, UMR_S 1085, Université de Rennes 1, Rennes, France
| | - Roselyne Viel
- CNRS, Inserm UMS Biosit, H2P2 Core Facility, Université de Rennes 1, Rennes, France
| | - Alain Fautrel
- CNRS, Inserm UMS Biosit, H2P2 Core Facility, Université de Rennes 1, Rennes, France
| | - Amir Khammari
- Service de Dermatologie, CHU Nantes, CIC 1413, INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes Université, Nantes, France
| | | | - Sophie Tartare-Deckert
- INSERM, C3M, team 'labellisée Ligue Contre le Cancer 2022, Université Côte d'Azur, Nice, France
| | - Aubin Penna
- Inserm, EHESP, IRSET, UMR_S 1085, Université de Rennes 1, Rennes, France.,CNRS, 4CS, Université de Poitiers, Poitiers, France
| |
Collapse
|
25
|
Bonamy C, Pesnel S, Ben Haddada M, Gorgette O, Schmitt C, Morel AL, Sauvonnet N. Impact of Green Gold Nanoparticle Coating on Internalization, Trafficking, and Efficiency for Photothermal Therapy of Skin Cancer. ACS OMEGA 2023; 8:4092-4105. [PMID: 36743010 PMCID: PMC9893490 DOI: 10.1021/acsomega.2c07054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/29/2022] [Indexed: 06/18/2023]
Abstract
Skin cancer is a global health issue and mainly composed of melanoma and nonmelanoma cancers. For the first clinical proof of concept on humans, we decided to study good prognosis skin cancers, i.e., carcinoma basal cell. In UE, the first-line treatment remains surgical resection, healing most of the tumors, but presents aesthetic disadvantages with a high reoccurrence rate on exposed areas. Moreover, the therapeutic indications could extend to melanoma and metastasis, which is a different medical strategy that could combine this treatment. Indeed, patients with late-stage melanoma are in a therapeutic impasse, despite recent targeted and immunological therapies. Photothermal therapy using gold nanoparticles is the subject of many investigations due to their strong potential to treat cancers by physical, thermal destruction. We developed gold nanoparticles synthesized by green chemistry (gGNPs), using endemic plant extract from Reunion Island, which have previously showed their efficiency at a preclinical stage. Here, we demonstrate that these gGNPs are less cytotoxic than gold nanoparticles synthesized by Turkevich's method. Furthermore, our work describes the optimization of gGNP coating and stabilization, also taking into consideration the gGNP path in cells (endocytosis, intracellular trafficking, and exocytosis), their specificity toward cancerous cells, their cytotoxicity, and their in vivo efficiency. Finally, based on the metabolic switch of cancerous cells overexpressing Glut transporters in skin cancers, we demonstrated that glucose-stabilized gGNP (gGNP@G) enables a quick internalization, fourfold higher in cancerous cells in contrast to healthy cells with no side cytotoxicity, which is particularly relevant to target and treat cancer.
Collapse
Affiliation(s)
- Clément Bonamy
- Torskal, 2 rue Maxime Rivière, 97490 Sainte-Clotilde, France
- Group
Intracellular Trafficking and Tissue Homeostasis, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Sabrina Pesnel
- Torskal, 2 rue Maxime Rivière, 97490 Sainte-Clotilde, France
| | | | - Olivier Gorgette
- Ultrastructural
BioImaging, Institut Pasteur, Université
Paris Cité, 75015 Paris, France
| | - Christine Schmitt
- Ultrastructural
BioImaging, Institut Pasteur, Université
Paris Cité, 75015 Paris, France
| | | | - Nathalie Sauvonnet
- Group
Intracellular Trafficking and Tissue Homeostasis, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| |
Collapse
|
26
|
Jung-Garcia Y, Maiques O, Monger J, Rodriguez-Hernandez I, Fanshawe B, Domart MC, Renshaw MJ, Marti RM, Matias-Guiu X, Collinson LM, Sanz-Moreno V, Carlton JG. LAP1 supports nuclear adaptability during constrained melanoma cell migration and invasion. Nat Cell Biol 2023; 25:108-119. [PMID: 36624187 PMCID: PMC9859759 DOI: 10.1038/s41556-022-01042-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/04/2022] [Indexed: 01/11/2023]
Abstract
Metastasis involves dissemination of cancer cells away from a primary tumour and colonization at distal sites. During this process, the mechanical properties of the nucleus must be tuned since they pose a challenge to the negotiation of physical constraints imposed by the microenvironment and tissue structure. We discovered increased expression of the inner nuclear membrane protein LAP1 in metastatic melanoma cells, at the invasive front of human primary melanoma tumours and in metastases. Human cells express two LAP1 isoforms (LAP1B and LAP1C), which differ in their amino terminus. Here, using in vitro and in vivo models that recapitulate human melanoma progression, we found that expression of the shorter isoform, LAP1C, supports nuclear envelope blebbing, constrained migration and invasion by allowing a weaker coupling between the nuclear envelope and the nuclear lamina. We propose that LAP1 renders the nucleus highly adaptable and contributes to melanoma aggressiveness.
Collapse
Affiliation(s)
- Yaiza Jung-Garcia
- Organelle Dynamics Laboratory, The Francis Crick Institute, London, UK
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Oscar Maiques
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Joanne Monger
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK
| | - Irene Rodriguez-Hernandez
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Bruce Fanshawe
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK
| | - Marie-Charlotte Domart
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Matthew J Renshaw
- Advanced Light Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Rosa M Marti
- Department of Dermatology, Hospital Universitari Arnau de Vilanova, University of Lleida, IRB Lleida, CIBERONC, Lleida, Spain
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, University of Lleida, IRB Lleida, CIBERONC, Lleida, Spain
| | - Lucy M Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Victoria Sanz-Moreno
- Sanz-Moreno Group, Centre for the Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, UK.
- Randall Division of Cell and Molecular Biophysics, King's College London, London, UK.
| | - Jeremy G Carlton
- Organelle Dynamics Laboratory, The Francis Crick Institute, London, UK.
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK.
| |
Collapse
|
27
|
Miñoza JMA, Rico JA, Zamora PRF, Bacolod M, Laubenbacher R, Dumancas GG, de Castro R. Biomarker Discovery for Meta-Classification of Melanoma Metastatic Progression Using Transfer Learning. Genes (Basel) 2022; 13:2303. [PMID: 36553569 PMCID: PMC9777873 DOI: 10.3390/genes13122303] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Melanoma is considered to be the most serious and aggressive type of skin cancer, and metastasis appears to be the most important factor in its prognosis. Herein, we developed a transfer learning-based biomarker discovery model that could aid in the diagnosis and prognosis of this disease. After applying it to the ensemble machine learning model, results revealed that the genes found were consistent with those found using other methodologies previously applied to the same TCGA (The Cancer Genome Atlas) data set. Further novel biomarkers were also found. Our ensemble model achieved an AUC of 0.9861, an accuracy of 91.05, and an F1 score of 90.60 using an independent validation data set. This study was able to identify potential genes for diagnostic classification (C7 and GRIK5) and diagnostic and prognostic biomarkers (S100A7, S100A7, KRT14, KRT17, KRT6B, KRTDAP, SERPINB4, TSHR, PVRL4, WFDC5, IL20RB) in melanoma. The results show the utility of a transfer learning approach for biomarker discovery in melanoma.
Collapse
Affiliation(s)
- Jose Marie Antonio Miñoza
- System Modeling and Simulation Laboratory, Department of Computer Science, University of the Philippines Diliman, Quezon City 1101, Philippines
| | - Jonathan Adam Rico
- Center for Informatics, University of San Agustin, Iloilo City 5000, Philippines
| | | | - Manny Bacolod
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Gerard G. Dumancas
- Center for Informatics, University of San Agustin, Iloilo City 5000, Philippines
- Loyola Science Center, Department of Chemistry, The University of Scranton, Scranton, PA 18510, USA
| | - Romulo de Castro
- Center for Informatics, University of San Agustin, Iloilo City 5000, Philippines
- 3R Biosystems, Long Beach, CA 90840, USA
| |
Collapse
|
28
|
Wagstaff W, Mwamba RN, Grullon K, Armstrong M, Zhao P, Hendren-Santiago B, Qin KH, Li AJ, Hu DA, Youssef A, Reid RR, Luu HH, Shen L, He TC, Haydon RC. Melanoma: Molecular genetics, metastasis, targeted therapies, immunotherapies, and therapeutic resistance. Genes Dis 2022; 9:1608-1623. [PMID: 36157497 PMCID: PMC9485270 DOI: 10.1016/j.gendis.2022.04.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/29/2022] [Accepted: 04/10/2022] [Indexed: 02/07/2023] Open
Abstract
Cutaneous melanoma is a common cancer and cases have steadily increased since the mid 70s. For some patients, early diagnosis and surgical removal of melanomas is lifesaving, while other patients typically turn to molecular targeted therapies and immunotherapies as treatment options. Easy sampling of melanomas allows the scientific community to identify the most prevalent mutations that initiate melanoma such as the BRAF, NRAS, and TERT genes, some of which can be therapeutically targeted. Though initially effective, many tumors acquire resistance to the targeted therapies demonstrating the need to investigate compensatory pathways. Immunotherapies represent an alternative to molecular targeted therapies. However, inter-tumoral immune cell populations dictate initial therapeutic response and even tumors that responded to treatment develop resistance in the long term. As the protocol for combination therapies develop, so will our scientific understanding of the many pathways at play in the progression of melanoma. The future direction of the field may be to find a molecule that connects all of the pathways. Meanwhile, noncoding RNAs have been shown to play important roles in melanoma development and progression. Studying noncoding RNAs may help us to understand how resistance - both primary and acquired - develops; ultimately allow us to harness the true potential of current therapies. This review will cover the basic structure of the skin, the mutations and pathways responsible for transforming melanocytes into melanomas, the process by which melanomas metastasize, targeted therapeutics, and the potential that noncoding RNAs have as a prognostic and treatment tool.
Collapse
Affiliation(s)
- William Wagstaff
- The Pritzker School of Medicine, and the Medical Scientist Training Program, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rimel N. Mwamba
- The Pritzker School of Medicine, and the Medical Scientist Training Program, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Karina Grullon
- The Pritzker School of Medicine, and the Medical Scientist Training Program, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Mikhayla Armstrong
- The Pritzker School of Medicine, and the Medical Scientist Training Program, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Piao Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Bryce Hendren-Santiago
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Kevin H. Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alexander J. Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Daniel A. Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Andrew Youssef
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Le Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
29
|
Georgoulias G, Zaravinos A. Genomic landscape of the immunogenicity regulation in skin melanomas with diverse tumor mutation burden. Front Immunol 2022; 13:1006665. [PMID: 36389735 PMCID: PMC9650672 DOI: 10.3389/fimmu.2022.1006665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/10/2022] [Indexed: 08/27/2023] Open
Abstract
Skin melanoma cells are tightly interconnected with their tumor microenvironment (TME), which influences their initiation, progression, and sensitivity/resistance to therapeutic interventions. An immune-active TME favors patient response to immune checkpoint inhibition (ICI), but not all patients respond to therapy. Here, we assessed differential gene expression in primary and metastatic tumors from the TCGA-SKCM dataset, compared to normal skin samples from the GTEx project and validated key findings across 4 independent GEO datasets, as well as using immunohistochemistry in independent patient cohorts. We focused our attention on examining the expression of various immune receptors, immune-cell fractions, immune-related signatures and mutational signatures across cutaneous melanomas with diverse tumor mutation burdens (TMB). Globally, the expression of most immunoreceptors correlated with patient survival, but did not differ between TMBhigh and TMBlow tumors. Melanomas were enriched in "naive T-cell", "effector memory T-cell", "exhausted T-cell", "resting Treg T-cell" and "Th1-like" signatures, irrespective of their BRAF, NF1 or RAS mutational status. Somatic mutations in IDO1 and HLA-DRA were frequent and could be involved in hindering patient response to ICI therapies. We finally analyzed transcriptome profiles of ICI-treated patients and associated their response with high levels of IFNγ, Merck18, CD274, CD8, and low levels of myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs) and M2 macrophages, irrespective of their TMB status. Overall, our findings highlight the importance of pre-existing T-cell immunity in ICI therapeutic outcomes in skin melanoma and suggest that TMBlow patients could also benefit from such therapies.
Collapse
Affiliation(s)
- George Georgoulias
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
- Cancer Genetics, Genomics and Systems Biology laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus
| |
Collapse
|
30
|
Yang J, Xu Y, Xie K, Gao L, Zhong W, Liu X. CEBPB is associated with active tumor immune environment and favorable prognosis of metastatic skin cutaneous melanoma. Front Immunol 2022; 13:991797. [PMID: 36353635 PMCID: PMC9637891 DOI: 10.3389/fimmu.2022.991797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/03/2022] [Indexed: 12/24/2023] Open
Abstract
Metastatic skin cutaneous melanoma (SKCM) is a common malignancy that accounts for low morbidity but high mortality of skin cancer. SKCM is characterized by high lymphocytic infiltration, whereas the states of infiltrated cells are variable in patients leading to a heterogeneous prognosis and hindering appropriate clinical decisions. It is therefore urgent to identify markers associated with lymphocytic infiltration, cellular conditions, and the prognosis of SKCM. In this study, we report that CEBPB, a transcriptional factor, is mainly expressed in macrophages in metastatic SKCM and associated with an active tumor immune environment and a favorable prognosis through integrated analysis of single-cell and bulk RNA-seq datasets. High CEBPB expression is significantly associated with active inflammation and immune response pathways in both macrophages and bulk SKCM tumor tissues. A signature based on CEBPB-associated genes that are specifically expressed in macrophages could robustly and prognostically separate different metastatic SKCM patients. In addition, the associations between the metastatic SKCM tumor signature and microenvironment with respect to T-cell recruitment and state, inflammation response, angiogenesis, and so on were also determined. In conclusion, we present here the first report on CEBPB in tumor immune environment and prognosis regulation in metastatic SKCM and construct a reliable signature, which should provide a useful biomarker for stratification of the patient's prognosis and therapeutic selection.
Collapse
Affiliation(s)
- Jingrun Yang
- Department of General Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Key Laboratory of Bio-inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Yang Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Kuixia Xie
- Department of Dermatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Ling Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Wenying Zhong
- Department of Dermatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xinhua Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
31
|
Exploration and validation of metastasis-associated genes for skin cutaneous melanoma. Sci Rep 2022; 12:13002. [PMID: 35906389 PMCID: PMC9338051 DOI: 10.1038/s41598-022-17468-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/26/2022] [Indexed: 12/04/2022] Open
Abstract
Skin cutaneous melanoma is a malignant and highly metastatic skin tumor, and its morbidity and mortality are still rising worldwide. However, the molecular mechanisms that promote melanoma metastasis are unclear. Two datasets (GSE15605 and GSE46517) were retrieved to identify the differentially expressed genes (DEGs), including 23 normal skin tissues (N), 77 primary melanoma tissues (T) and 85 metastatic melanoma tissues (M). Gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed to explore the functions of the DEGs. We constructed protein–protein interaction network using the STRING database and Cytoscape software. Using the cytoHubba plugin of Cytoscape, we identified the most significant hub genes by five analytical methods (Degree, Bottleneck, MCC, MNC, and EPC). Hub gene expression was validated using the UALCAN website. Clinical relevance was investigated using The Cancer Genome Atlas resources. Finally, we explored the association between metastasis-associated genes and immune infiltrates through the Tumor Immune Estimation Resource (TIMER) database and performed drug–gene interaction analysis using the Drug-Gene Interaction database. A total of 294 specific genes were related to melanoma metastasis and were mainly involved in the positive regulation of locomotion, mitotic cell cycle process, and epithelial cell differentiation. Four hub genes (CDK1, FOXM1, KIF11, and RFC4) were identified from the cytoHubba plugin of Cytoscape. CDK1 was significantly upregulated in metastatic melanoma compared with primary melanoma, and high CDK1 expression was positively correlated with worse overall survival. Immune infiltration analysis revealed that CDK1 expression negatively correlated with macrophage infiltration (Rho = − 0.164, P = 2.02e−03) and positively correlated with neutrophil cells (Rho = 0.269, P = 2.72e−07) in SKCM metastasis. In addition, we identified that CDK1 had a close interaction with 10 antitumor drugs. CDK1 was identified as a hub gene involved in the progression of melanoma metastasis and may be regarded as a therapeutic target for melanoma patients to improve prognosis and prevent metastasis in the future.
Collapse
|
32
|
Identification of a Seven-Differentially Expressed Gene-Based Recurrence-Free Survival Model for Melanoma Patients. DISEASE MARKERS 2022; 2022:3915112. [PMID: 35872694 PMCID: PMC9303152 DOI: 10.1155/2022/3915112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022]
Abstract
Melanoma is a malignant tumor that originates in melanocytes of the skin or mucous membrane, which has a high mortality rate and worse prognosis. Therefore, perspective prognosis evaluation seems more important for patients' treatment. Gene expression profiles of melanoma were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, respectively. 130 consistent differentially expressed genes (DEGs) were identified between melanoma and nevus tissues from two GEO cohorts. Prognostic genes were identified by univariate analysis, and 20 of them were regarded to be associated with the recurrence-free survival (RFS) of melanoma patients. Then, the LASSO Cox regression analysis chose seven of them to establish a seven-DEG-based RFS predicting signature. We demonstrated that this model was more powerful to predict RFS risk than other individual clinical features and was able to independently predict the RFS outcomes in different subsets of patients. We attempted to search for the underlying mechanisms by analyzing the coexpression genes of the seven candidates, and the pathway enrichment analyses indicated that immune response-related pathways might play a critical role in melanoma progression. Finally, we establish a robust seven-DEG-based RFS predicting signature, which will facilitate the personalized treatment of melanoma patients.
Collapse
|
33
|
NRF3 Decreases during Melanoma Carcinogenesis and Is an Independent Prognostic Marker in Melanoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2240223. [PMID: 35378827 PMCID: PMC8976671 DOI: 10.1155/2022/2240223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 11/17/2022]
Abstract
The prognostic significance of the major redox regulator, nuclear factor erythroid-2-related factor 2 (NRF2), is recognized in many cancers, but the role of NRF3 is not studied. Analysis from the Gene Expression Omnibus datasets showed that NRF3 mRNA levels increased from benign to dysplastic naevi (p = 0.04). We characterized the immunohistochemical expression of NRF3 in 81 naevi, 67 primary skin melanomas, and 51 lymph node metastases. The immunohistochemical expression of cytoplasmic NRF3 decreased from benign to dysplastic naevi (p < 0.001) and further to primary melanomas (p < 0.001). High cytoplasmic NRF3 protein expression in pigment cells of the primary melanomas associated with worse melanoma-specific survival in multivariate analysis, specifically in the subgroup of patients with the lymph node metastases at the time of diagnosis (hazard ratio 3.179; 95% confidence interval 1.065-9.493; p = 0.038). Intriguingly, we did not observe associations between NRF3 and the traditional prognostic factors such as Breslow thickness, ulceration, or stage. Together, this data represents the primary description about the role of NRF3 in pigment tumours that is worthy of further explorations.
Collapse
|
34
|
Moubarak RS, de Pablos-Aragoneses A, Ortiz-Barahona V, Gong Y, Gowen M, Dolgalev I, Shadaloey SAA, Argibay D, Karz A, Von Itter R, Vega-Sáenz de Miera EC, Sokolova E, Darvishian F, Tsirigos A, Osman I, Hernando E. The histone demethylase PHF8 regulates TGFβ signaling and promotes melanoma metastasis. SCIENCE ADVANCES 2022; 8:eabi7127. [PMID: 35179962 PMCID: PMC8856617 DOI: 10.1126/sciadv.abi7127] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/14/2021] [Indexed: 05/17/2023]
Abstract
The contribution of epigenetic dysregulation to metastasis remains understudied. Through a meta-analysis of gene expression datasets followed by a mini-screen, we identified Plant Homeodomain Finger protein 8 (PHF8), a histone demethylase of the Jumonji C protein family, as a previously unidentified prometastatic gene in melanoma. Loss- and gain-of-function approaches demonstrate that PHF8 promotes cell invasion without affecting proliferation in vitro and increases dissemination but not subcutaneous tumor growth in vivo, thus supporting its specific contribution to the acquisition of metastatic potential. PHF8 requires its histone demethylase activity to enhance melanoma cell invasion. Transcriptomic and epigenomic analyses revealed that PHF8 orchestrates a molecular program that directly controls the TGFβ signaling pathway and, as a consequence, melanoma invasion and metastasis. Our findings bring a mechanistic understanding of epigenetic regulation of metastatic fitness in cancer, which may pave the way for improved therapeutic interventions.
Collapse
Affiliation(s)
- Rana S. Moubarak
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | | | | | - Yixiao Gong
- Applied Bioinformatics Laboratories, NYU School of Medicine, NY 10016, USA
| | - Michael Gowen
- NYU School of Medicine Institute for Computational Medicine, New York, NY 10016, USA
| | - Igor Dolgalev
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Applied Bioinformatics Laboratories, NYU School of Medicine, NY 10016, USA
| | - Sorin A. A. Shadaloey
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Diana Argibay
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Alcida Karz
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Richard Von Itter
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | | | - Elena Sokolova
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Farbod Darvishian
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
| | - Aristotelis Tsirigos
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Applied Bioinformatics Laboratories, NYU School of Medicine, NY 10016, USA
- NYU School of Medicine Institute for Computational Medicine, New York, NY 10016, USA
| | - Iman Osman
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
- Ronald O. Perelman Department of Dermatology, NYU School of Medicine, New York, NY 10016, USA
| | - Eva Hernando
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, NYU Cancer Institute, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
35
|
Identification of Keratinocyte Differentiation-Involved Genes for Metastatic Melanoma by Gene Expression Profiles. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2021:9652768. [PMID: 35003328 PMCID: PMC8728391 DOI: 10.1155/2021/9652768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/02/2021] [Indexed: 11/18/2022]
Abstract
Background Melanoma is the deadliest type of skin cancer. Until now, its pathological mechanisms, particularly the mechanism of metastasis, remain largely unknown. Our study on the identification of genes in association with metastasis for melanoma provides a novel understanding of melanoma. Methods From the Gene Expression Omnibus (GEO) database, the gene expression microarray datasets GSE46517, GSE7553, and GSE8401 were downloaded. We made use of R aiming at analyzing the differentially expressed genes (DEGs) between metastatic and nonmetastatic melanoma. R was also used in differentially expressed miRNA (DEM) data mining from GSE18509, GSE19387, GSE24996, GSE34460, GSE35579, GSE36236, and GSE54492 datasets referring to Li's study. Based on the DEG and DEM data, we performed functional enrichment analysis through the application of the DAVID database. Furthermore, we constructed the protein-protein interaction (PPI) network and established functional modules by making use of the STRING database. Through making use of Cytoscape, the PPI results were visualized. We predicted the targets of the DEMs through applying TargetScan, miRanda, and PITA databases and identified the overlapping genes between DEGs and predicted targets, followed by the construction of DEM-DEG pair network. The expressions of these keratinocyte differentiation-involved genes in Module 1 were identified based on the data from TCGA. Results 239 DEGs were screened out in all 3 datasets, which were inclusive of 21 positively regulated genes and 218 negatively regulated genes. Based on these 239 DEGs, we finished constructing the PPI network which was formed from 225 nodes and 846 edges. We finished establishing 3 functional modules. And we analyzed 92 overlapping genes and 26 miRNA, including 11 upregulated genes targeted by 11 negatively regulated DEMs and 81 downregulated genes targeted by 15 positively regulated DEMs. As proof of the differential expression of metastasis-associated genes, eleven keratinocyte differentiation-involved genes, including LOR, EVPL, SPRR1A, FLG, SPRR1B, SPRR2B, TGM1, DSP, CSTA, CDSN, and IVL in Module 1, were obviously downregulated in metastatic melanoma tissue in comparison with primary melanoma tissue based on the data from TCGA. Conclusion 239 melanoma metastasis-associated genes and 26 differentially expressed miRNA were identified in our study. The keratinocyte differentiation-involved genes may take part in melanoma metastasis, providing a latent molecular mechanism for this disease.
Collapse
|
36
|
Wu S, Shen D, Zhao L. AKAP9 Upregulation Predicts Unfavorable Prognosis in Pediatric Acute Myeloid Leukemia and Promotes Stemness Properties via the Wnt/β-Catenin Pathway. Cancer Manag Res 2022; 14:157-167. [PMID: 35046723 PMCID: PMC8760470 DOI: 10.2147/cmar.s343033] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/07/2021] [Indexed: 12/16/2022] Open
Abstract
Background PRKA kinase anchor protein 9 (AKAP9) is a scaffold protein involved in various cellular processes, including cell adhesion, proliferation, differentiation, and apoptosis. Although the oncogenic role of AKAP9 in solid tumors is well elucidated, the functions and mechanisms of AKAP9 in acute myeloid leukemia (AML) are still not understood. Methods We used the gene expression omnibus (GEO) database (GSE2191) to determine the mRNA expression of AKAP9 in the bone marrow of pediatric AML and healthy patients. We further used the therapeutically available research to generate effective treatments (TARGET) database to elucidate the relationship between AKAP9 expression and clinical outcomes in pediatric patients with AML. In addition, cell proliferation, cell cycle, apoptosis, RT-PCR, and Western blotting assays were applied to reveal the functions of AKAP9 and the underlying mechanisms of AKAP9 silencing in THP1 and HL60 cell lines. Results AKAP9 is overexpressed in the bone marrow of pediatric AML patients as compared with that of healthy patients. High expression of AKAP9 was found to be a predictor of poor overall survival (OS) and event-free survival (EFS). Using univariate and multivariate survival analyses, we found that high AKAP9 expression is an independent predictor of a worse OS and EFS. Functionally, AKAP9 silencing significantly inhibited AML cell proliferation, and cell cycle progression and promoted apoptosis. Moreover, AKAP9 silencing significantly downregulated the expression of stemness markers and β-catenin. Conclusion AKAP9 upregulation is a predictor of unfavorable prognosis, promotes stemness, and activates the Wnt/β-catenin pathway in AML patients. AKAP9 may act as a prognostic biomarker of AML in pediatric patients and a future therapeutic target.
Collapse
Affiliation(s)
- Shiwen Wu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
- Departments of Clinical Laboratory, The First Hospital of Lanzhou University, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Dongqin Shen
- Department of Medical Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Li Zhao
- Department of Central Laboratory, Gansu Key Laboratory of Genetic Study of Hematopathy, The First Hospital of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
- Correspondence: Li Zhao Department of Central Laboratory, Gansu Key Laboratory of Genetic Study of Hematopathy, The First Hospital of Lanzhou University, Lanzhou University, No. 1 Donggang West Road, Lanzhou, Gansu, 730000, People’s Republic of ChinaTel +8613919934053Fax +8609318356353 Email
| |
Collapse
|
37
|
Cherepakhin OS, Argenyi ZB, Moshiri AS. Genomic and Transcriptomic Underpinnings of Melanoma Genesis, Progression, and Metastasis. Cancers (Basel) 2021; 14:123. [PMID: 35008286 PMCID: PMC8750021 DOI: 10.3390/cancers14010123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022] Open
Abstract
Melanoma is a deadly skin cancer with rapidly increasing incidence worldwide. The discovery of the genetic drivers of melanomagenesis in the last decade has led the World Health Organization to reclassify melanoma subtypes by their molecular pathways rather than traditional clinical and histopathologic features. Despite this significant advance, the genomic and transcriptomic drivers of metastatic progression are less well characterized. This review describes the known molecular pathways of cutaneous and uveal melanoma progression, highlights recently identified pathways and mediators of metastasis, and touches on the influence of the tumor microenvironment on metastatic progression and treatment resistance. While targeted therapies and immune checkpoint blockade have significantly aided in the treatment of advanced disease, acquired drug resistance remains an unfortunately common problem, and there is still a great need to identify potential prognostic markers and novel therapeutic targets to aid in such cases.
Collapse
Affiliation(s)
| | - Zsolt B. Argenyi
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA;
| | - Ata S. Moshiri
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA;
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
38
|
Xuan X, Wang Y, Sun Y, Huang C. Identification of Genes Potentially Associated with Melanoma Tumorigenesis Through Co-Expression Network Analysis. Int J Gen Med 2021; 14:8495-8508. [PMID: 34824546 PMCID: PMC8610383 DOI: 10.2147/ijgm.s336295] [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: 08/26/2021] [Accepted: 11/02/2021] [Indexed: 12/24/2022] Open
Abstract
Background Melanoma is one of the most malignant and aggressive skin tumors, and its incidence is increasing worldwide. However, few studies have investigated the process of tumorigenesis from normal skin to melanoma. Methods Several bioinformatics analyses, including GEO databases, Oncomine database, TCGA database, STRING, MCODE and cytoHubba plug-ins, GEPIA, TIMER and TRRUST and DGIdb, were performed to disclose the hub genes and immunology implicated in primary melanoma tumorigenesis. Finally, immunohistochemistry (IHC) and quantitative real-time PCR (qPCR) were used to validate the results of bioinformatics analysis in vitro. Results A total of 295 overlapping DEGs (ODEGs) (157 upregulated and 138 downregulated) and 9 hub genes were identified between primary melanoma and normal skin tissues. Functional analysis of these 9 hub genes indicated that the genes were primarily enriched in cell chemotaxis, the chemokine-mediated signaling pathway, the extracellular region, the extracellular space, chemokine activity and CXCR3 chemokine receptor binding. KEGG pathway enrichment showed that these genes were primarily involved in the chemokine signaling pathway, cytokine–cytokine receptor interaction, the toll-like receptor signaling pathway, the cytosolic DNA-sensing pathway and the TNF signaling pathway. Upregulated CCL5, CCL4, CXCL9 and CXCL10 demonstrated good overall survival (OS), and most of them have a higher expression in stage 0 and 1 of melanoma. Moreover, immune infiltration analysis showed that the above hub genes showed a strong positive correlation between their expression and infiltration of the six immune cell subsets. Transcription factor regulation network suggested that RELA and NFKB1 are the transcription factors of CCL4, CCL5, CXCL10 and CXCL2, while IRF7, IRF3 and IRF1 are the transcription factors of CCL5 and CXCL10. Drug–gene interaction analysis identified 46 drug–gene interactions. In vitro data demonstrated that the level of CCL4, CCL5, CXCL9 and CXCL10 is higher in melanoma than that in normal skin tissues, either at tissue or cell lines level. Conclusion In summary, we identified 4 key chemokine members related to tumorigenesis and progression in primary melanoma, and these results may help to elucidate melanoma tumorigenesis and facilitate its treatment.
Collapse
Affiliation(s)
- Xiuyun Xuan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People's Republic of China
| | - Yuqi Wang
- Department of Radiology, Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, 221002, People's Republic of China
| | - Yanhong Sun
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Changzheng Huang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, People's Republic of China
| |
Collapse
|
39
|
Maiques O, Fanshawe B, Crosas-Molist E, Rodriguez-Hernandez I, Volpe A, Cantelli G, Boehme L, Orgaz JL, Mardakheh FK, Sanz-Moreno V, Fruhwirth GO. A preclinical pipeline to evaluate migrastatics as therapeutic agents in metastatic melanoma. Br J Cancer 2021; 125:699-713. [PMID: 34172930 PMCID: PMC8405734 DOI: 10.1038/s41416-021-01442-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 03/23/2021] [Accepted: 05/13/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Metastasis is a hallmark of cancer and responsible for most cancer deaths. Migrastatics were defined as drugs interfering with all modes of cancer cell invasion and thus cancers' ability to metastasise. First anti-metastatic treatments have recently been approved. METHODS We used bioinformatic analyses of publicly available melanoma databases. Experimentally, we performed in vitro target validation (including 2.5D cell morphology analysis and mass spectrometric analysis of RhoA binding partners), developed a new traceable spontaneously metastasising murine melanoma model for in vivo validation, and employed histology (haematoxylin/eosin and phospho-myosin II staining) to confirm drug action in harvested tumour tissues. RESULTS Unbiased and targeted bioinformatic analyses identified the Rho kinase (ROCK)-myosin II pathway and its various components as potentially relevant targets in melanoma. In vitro validation demonstrated redundancy of several RhoGEFs upstream of RhoA and confirmed ROCK as a druggable target downstream of RhoA. The anti-metastatic effects of two ROCK inhibitors were demonstrated through in vivo melanoma metastasis tracking and inhibitor effects also confirmed ex vivo by digital pathology. CONCLUSIONS We proposed a migrastatic drug development pipeline. As part of the pipeline, we provide a new traceable spontaneous melanoma metastasis model for in vivo quantification of metastasis and anti-metastatic effects by non-invasive imaging.
Collapse
Affiliation(s)
- Oscar Maiques
- Centre for Tumour Microenvironment at Barts Cancer Institute, Queen Mary University of London, Charterhouse Square Campus, John Vane Science Centre, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK
| | - Bruce Fanshawe
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King's College London, Guy's Campus, London, UK
| | - Eva Crosas-Molist
- Centre for Tumour Microenvironment at Barts Cancer Institute, Queen Mary University of London, Charterhouse Square Campus, John Vane Science Centre, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK
| | - Irene Rodriguez-Hernandez
- Centre for Tumour Microenvironment at Barts Cancer Institute, Queen Mary University of London, Charterhouse Square Campus, John Vane Science Centre, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK
| | - Alessia Volpe
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King's College London, Guy's Campus, London, UK
- Molecular Imaging Group, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gaia Cantelli
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK
| | - Lena Boehme
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK
| | - Jose L Orgaz
- Centre for Tumour Microenvironment at Barts Cancer Institute, Queen Mary University of London, Charterhouse Square Campus, John Vane Science Centre, London, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK
- Instituto de Investigaciones Biomedicas 'Alberto Sols', CSIC-UAM, Madrid, Spain
| | - Faraz K Mardakheh
- Centre for Cancer Cell & Molecular Biology at Barts Cancer Institute, Queen Mary University of London, Charterhouse Square Campus, John Vane Science Centre, London, UK
| | - Victoria Sanz-Moreno
- Centre for Tumour Microenvironment at Barts Cancer Institute, Queen Mary University of London, Charterhouse Square Campus, John Vane Science Centre, London, UK.
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, UK.
| | - Gilbert O Fruhwirth
- Imaging Therapies and Cancer Group, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Studies, King's College London, Guy's Campus, London, UK.
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London, UK.
| |
Collapse
|
40
|
Scatolini M, Patel A, Grosso E, Mello-Grand M, Ostano P, Coppo R, Vitiello M, Venesio T, Zaccagna A, Pisacane A, Sarotto I, Taverna D, Poliseno L, Bergamaschi D, Chiorino G. GJB5 association with BRAF mutation and survival in cutaneous malignant melanoma. Br J Dermatol 2021; 186:117-128. [PMID: 34240406 DOI: 10.1111/bjd.20629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Gap junctional intercellular communication is crucial for epidermal cellular homeostasis. Inability to establish melanocyte-keratinocytes contacts and loss of intercellular junction's integrity may contribute to melanoma development. Connexins, laminins and desmocollins have been implicated in the control of melanoma growth, where their reduced expression has been reported in metastatic lesions. OBJECTIVES The aim of this study was to investigate Connexin 31.1 (GJB5) expression and identify any association with BRAF mutational status, melanoma patient prognosis and MAPK inhibitors (MAPKi) treatment. MATERIAL AND METHODS GJB5 expression was measured at RNA and protein level in melanoma clinical samples and established cell lines treated or not with BRAF and MEK inhibitors, as well as in cell lines which developed MAPK inhibitors resistance. Findings were further validated and confirmed by analysis of independent datasets. RESULTS Our analysis reveals significant downregulation of GJB5 expression in metastatic melanoma lesions compared to primary ones and in BRAF mutated versus BRAF wild-type melanomas. Likewise, GJB5 expression is significantly lower in BRAFV600E compared with BRAFWT cell lines and increases upon MAPKi treatment. MAPKi-resistant melanoma cells display a similar expression pattern compared to BRAFWT cells, with increased GJB5 expression associated with morphological changes. Enhancement of BRAFV600E expression in BRAFWT melanoma cells significantly upregulates miR-335-5p expression with consequent downregulation of GJB5, one of its targets. Furthermore, overexpression of miR-335-5p in two BRAFWT cell lines confirms specific GJB5 protein downregulation. RT-qPCR analysis also revealed upregulation of miR-335 in BRAFV600E melanoma cells, which is significantly downregulated in cells resistant to MEK inhibitors. Our data were further validated using the TCGA-SKCM dataset, where BRAF mutations associate with increased miR-335 expression and inversely correlate with GJB5 expression. In clinical samples, GJB5 underexpression is also associated with patient overall worse survival, especially at early stages. CONCLUSION We identified a significant association between metastases / BRAF mutation and low GJB5 expression in melanoma. Our results identify a novel mechanism of Gap-junctional protein regulation, suggesting a prognostic role for GJB5 in cutaneous melanoma.
Collapse
Affiliation(s)
- M Scatolini
- Molecular Oncology Laboratory, Fondazione Edo ed Elvo Tempia, 13875, Ponderano, BI, Italy
| | - A Patel
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London SMD, QMUL, London, E1 2AT, UK
| | - E Grosso
- Molecular Oncology Laboratory, Fondazione Edo ed Elvo Tempia, 13875, Ponderano, BI, Italy
| | - M Mello-Grand
- Cancer Genomics Laboratory, Fondazione Edo ed Elvo Tempia, 13900, Biella, Italy
| | - P Ostano
- Cancer Genomics Laboratory, Fondazione Edo ed Elvo Tempia, 13900, Biella, Italy
| | - R Coppo
- Molecular Biotechnology Centre, 10126, Torino, Italy.,Department of Clinical Bio-Resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - M Vitiello
- Oncogenomics Unit, Core Research Laboratory, Istituto Toscano Tumori, Institute of Clinical Physiology, CNR, 56124, Pisa, Italy
| | - T Venesio
- Pathology and Dermosurgery Units, Candiolo Cancer Institute (FPO-IRCCS), 10060, Candiolo, Turin, Italy
| | - A Zaccagna
- Pathology and Dermosurgery Units, Candiolo Cancer Institute (FPO-IRCCS), 10060, Candiolo, Turin, Italy
| | - A Pisacane
- Pathology and Dermosurgery Units, Candiolo Cancer Institute (FPO-IRCCS), 10060, Candiolo, Turin, Italy
| | - I Sarotto
- Pathology and Dermosurgery Units, Candiolo Cancer Institute (FPO-IRCCS), 10060, Candiolo, Turin, Italy
| | - D Taverna
- Molecular Biotechnology Centre, 10126, Torino, Italy
| | - L Poliseno
- Oncogenomics Unit, Core Research Laboratory, Istituto Toscano Tumori, Institute of Clinical Physiology, CNR, 56124, Pisa, Italy
| | - D Bergamaschi
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London SMD, QMUL, London, E1 2AT, UK
| | - G Chiorino
- Cancer Genomics Laboratory, Fondazione Edo ed Elvo Tempia, 13900, Biella, Italy
| |
Collapse
|
41
|
Saito RF, Rangel MC, Halman JR, Chandler M, de Sousa Andrade LN, Odete-Bustos S, Furuya TK, Carrasco AGM, Chaves-Filho AB, Yoshinaga MY, Miyamoto S, Afonin KA, Chammas R. Simultaneous silencing of lysophosphatidylcholine acyltransferases 1-4 by nucleic acid nanoparticles (NANPs) improves radiation response of melanoma cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 36:102418. [PMID: 34171470 DOI: 10.1016/j.nano.2021.102418] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 12/19/2022]
Abstract
Radiation induces the generation of platelet-activating factor receptor (PAF-R) ligands, including PAF and oxidized phospholipids. Alternatively, PAF is also synthesized by the biosynthetic enzymes lysophosphatidylcholine acyltransferases (LPCATs) which are expressed by tumor cells including melanoma. The activation of PAF-R by PAF and oxidized lipids triggers a survival response protecting tumor cells from radiation-induced cell death, suggesting the involvement of the PAF/PAF-R axis in radioresistance. Here, we investigated the role of LPCATs in the melanoma cell radiotherapy response. LPCAT is a family of four enzymes, LPCAT1-4, and modular nucleic acid nanoparticles (NANPs) allowed for the simultaneous silencing of all four LPCATs. We found that the in vitro simultaneous silencing of all four LPCAT transcripts by NANPs enhanced the therapeutic effects of radiation in melanoma cells by increasing cell death, reducing long-term cell survival, and activating apoptosis. Thus, we propose that NANPs are an effective strategy for improving radiotherapy efficacy in melanomas.
Collapse
Affiliation(s)
- Renata F Saito
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil.
| | - Maria Cristina Rangel
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil.
| | - Justin R Halman
- Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA.
| | - Morgan Chandler
- Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA.
| | - Luciana Nogueira de Sousa Andrade
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil.
| | - Silvina Odete-Bustos
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil.
| | - Tatiane Katsue Furuya
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil.
| | - Alexis Germán Murillo Carrasco
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil.
| | - Adriano B Chaves-Filho
- Laboratório de Lipídeos Modificados, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil.
| | - Marcos Y Yoshinaga
- Laboratório de Lipídeos Modificados, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil.
| | - Sayuri Miyamoto
- Laboratório de Lipídeos Modificados, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil.
| | - Kirill A Afonin
- Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA.
| | - Roger Chammas
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil; Nanoscale Science Program, Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA.
| |
Collapse
|
42
|
The Research on the Treatment of Metastatic Skin Cutaneous Melanoma by Huanglian Jiedu Decoction Based on the Analysis of Immune Infiltration Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9952060. [PMID: 34239596 PMCID: PMC8241506 DOI: 10.1155/2021/9952060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/22/2021] [Accepted: 06/07/2021] [Indexed: 11/18/2022]
Abstract
Objective To explore the potential mechanism of Huanglian Jiedu Decoction (HJD) treatment and prevention of metastatic Cutaneous Melanoma (CM) occurrence and metastasis based on network pharmacological methods and immune infiltration analysis. Methods The GEO database was used to obtain metastatic CM disease targets, the TCMSP database and the HERB database were used to obtain HJD action targets, core genes were screened by protein interaction network, and the potential mechanism of HJD in the treatment of metastatic CM was explored by enrichment analysis, prognostic analysis and immune infiltration analysis. Results HJD treatment of metastatic CM involved 60 targets, enrichment analysis showed that HJD treatment of metastatic CM involved Chemokine signaling pathway, NF-kappa B signaling pathway, and Fluid shear stress and atherosclerosis, etc. Prognostic analysis revealed that HJD had a certain ability to improve the prognosis of metastatic CM patients. Immune infiltration analysis showed that HJD could inhibit the immune cell infiltration of metastatic CM patients by acting on related targets. Conclusions Our study identified the potential mechanism of HJD in the treatment of metastatic CM through network pharmacology, and revealed the mechanism of HJD in the prevention of Skin Cutaneous Melanoma metastasis through immune infiltration analysis and prognostic analysis.
Collapse
|
43
|
Wang J. Prognostic score model-based signature genes for predicting the prognosis of metastatic skin cutaneous melanoma. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:5125-5145. [PMID: 34517481 DOI: 10.3934/mbe.2021261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
PURPOSE Cutaneous melanoma (SKCM) is the most invasive malignancy of skin cancer. Metastasis to distant lymph nodes or other system is an indicator of poor prognosis in melanoma patients. The aim of this study was to identify reliable prognostic biomarkers for SKCMs. METHODS Four RNA-sequencing datasets associated with SKCMs were downloaded from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) database as well as corresponding clinical information. Differentially expressed genes (DEGs) were screened between primary and metastatic samples by using MetaDE tool. Weighted gene co-expression network analysis (WGCNA) was conducted to screen functional modules. A prognostic score (PS)-based predictive model and nomogram model were constructed to identify signature genes and independent clinicopathologic factors. RESULTS Based on MetaDE analysis and WGCNA, a total of 456 overlapped genes were identified as hub genes related to SKCMs progression. Functional enrichment analysis revealed these genes were mainly involved in the hippo signaling pathway, signaling pathways regulating pluripotency of stem cells, pathways in cancer. In addition, eight optimal DEGs (RFPL1S, CTSV, EGLN3, etc.) were identified as signature genes by using PS model. Cox regression analysis revealed that pathologic stage T, N and recurrence were independent prognostic factors. Three clinical factors and PS status were incorporated to construct a nomogram predictive model for estimating the three years and five-year survival probability of individuals. CONCLUSIONS The prognosis prediction model of this study may provide a promising method for decision making in clinic and prognosis predicting of SKCM patients.
Collapse
Affiliation(s)
- Jiaping Wang
- Laboratory Medicine, Donghai County People's Hospital, Lianyungang City, Jiangsu 222300, China
| |
Collapse
|
44
|
Mou K, Zhang J, Mu X, Wang L, Liu W, Ge R. Zwint facilitates melanoma progression by promoting c-Myc expression. Exp Ther Med 2021; 22:818. [PMID: 34131441 PMCID: PMC8193213 DOI: 10.3892/etm.2021.10250] [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: 11/06/2020] [Accepted: 04/26/2021] [Indexed: 12/14/2022] Open
Abstract
ZW10 interactor (Zwint) is upregulated in various types of tumors and exerts a carcinogenic effect. However, little is known about the expression profile, function and molecular mechanisms of action of Zwint in melanoma. Therefore, the aim of the present study was to investigate the expression levels of Zwint in melanoma cell lines and tissues. It was revealed that Zwint was highly expressed in melanoma samples. Functional experiments indicated that Zwint knockdown suppressed the proliferation and migration of A375 melanoma cells. Further mechanistic studies demonstrated that Zwint knockdown decreased the protein expression levels of c-Myc, MMP-2, Slug, mTOR, phosphorylated (p)-mTOR, p-p38 and fibronectin, while it increased the protein expression levels of E-cadherin and MMP-9. Among these genes, c-Myc, MMP-2 and Slug were overexpressed to investigate their effects on cell proliferation following Zwint knockdown. The results demonstrated that overexpression of c-Myc, but not MMP-2 or Slug, rescued the effects of Zwint knockdown on melanoma cell proliferation and migration. Taken together, the results of the present study suggested that Zwint may act as an oncogene in melanoma by regulating c-Myc expression.
Collapse
Affiliation(s)
- Kuanhou Mou
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jian Zhang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xin Mu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lijuan Wang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenli Liu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Rui Ge
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
45
|
Tartey S, Neale G, Vogel P, Malireddi RKS, Kanneganti TD. A MyD88/IL1R Axis Regulates PD-1 Expression on Tumor-Associated Macrophages and Sustains Their Immunosuppressive Function in Melanoma. Cancer Res 2021; 81:2358-2372. [PMID: 33619117 PMCID: PMC11645125 DOI: 10.1158/0008-5472.can-20-3510] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/22/2020] [Accepted: 02/18/2021] [Indexed: 11/16/2022]
Abstract
Macrophages are critical mediators of tissue homeostasis, cell proliferation, and tumor metastasis. Tumor-associated macrophages (TAM) are generally associated with tumor-promoting immunosuppressive functions in solid tumors. Here, we examined the transcriptional landscape of adaptor molecules downstream of Toll-like receptors in human cancers and found that higher expression of MYD88 correlated with tumor progression. In murine melanoma, MyD88, but not Trif, was essential for tumor progression, angiogenesis, and maintaining the immunosuppressive phenotype of TAMs. In addition, MyD88 expression in myeloid cells drove melanoma progression. The MyD88/IL1 receptor (IL1R) axis regulated programmed cell death (PD)-1 expression on TAMs by promoting recruitment of NF-κBp65 to the Pdcd1 promoter. Furthermore, a combinatorial immunotherapy approach combining the MyD88 inhibitor with anti-PD-1 blockade elicited strong antitumor effects. Thus, the MyD88/IL1R axis maintains the immunosuppressive function of TAMs and promotes tumor growth by regulating PD-1 expression. SIGNIFICANCE: These findings indicate that MyD88 regulates TAM-immunosuppressive activity, suggesting that macrophage-mediated immunotherapy combining MYD88 inhibitors with PD-1 blockade could result in better treatment outcomes in a wide variety of cancers. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/9/2358/F1.large.jpg.
Collapse
Affiliation(s)
- Sarang Tartey
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Peter Vogel
- Animal Resources Center and the Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | | |
Collapse
|
46
|
L Antigen Family Member 3 Serves as a Prognostic Biomarker for the Clinical Outcome and Immune Infiltration in Skin Cutaneous Melanoma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6648182. [PMID: 33829062 PMCID: PMC8000545 DOI: 10.1155/2021/6648182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 12/31/2022]
Abstract
L Antigen Family Member 3 (LAGE3) is an important RNA modification-related protein. Whereas few studies have interrogated the LAGE3 protein, there is limited data on its role in tumors. Here, we analyzed and profiled the LAGE3 protein in skin cutaneous melanoma (CM) using TCGA, GTEx, or GEO databases. Our data showed an upregulation of LAGE3 in melanoma cell lines compared to normal skin cell lines. Besides, the Kaplan–Meier curves and Cox proportional hazard model revealed that LAGE3 was an independent survival indicator for CM, especially in metastatic CM. Moreover, LAGE3 was negatively associated with multiple immune cell infiltration levels in CM, especially CD8+ T cells in metastatic CM. Taken together, our study suggests that LAGE3 could be a potential prognostic biomarker and might be a potential target for the development of novel CM treatment strategies.
Collapse
|
47
|
Liu Y, He D, Xiao M, Zhu Y, Zhou J, Cao K. Long noncoding RNA LINC00518 induces radioresistance by regulating glycolysis through an miR-33a-3p/HIF-1α negative feedback loop in melanoma. Cell Death Dis 2021; 12:245. [PMID: 33664256 PMCID: PMC7933330 DOI: 10.1038/s41419-021-03523-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 01/31/2023]
Abstract
The long noncoding RNA, LINC00518, is highly expressed in various types of cancers and is involved in cancer progression. Although LINC00518 promotes the metastasis of cutaneous malignant melanoma (CMM), the mechanism underlaying its effects on CMM radiosensitivity remains unclear. In this study, LINC00518 expression was significantly upregulated in CMM samples, and LINC00518 levels were associated with poor prognosis of patients with CMM. Knockdown of LINC00518 in CMM cells significantly inhibited cell invasion, migration, proliferation, and clonogenicity. LINC00518-mediated invasion, migration, proliferation, and clonogenicity were negatively regulated by the microRNA, miR-33a-3p, in vitro, which increased sensitivity to radiotherapy via inhibition of the hypoxia-inducible factor 1α (HIF-1α)/lactate dehydrogenase A glycolysis axis. Additionally, HIF-1α recognized the miR-33a-3p promoter region and recruited histone deacetylase 2, which decreased the expression of miR-33a-3p and formed an LINC00518/miR-33a-3p/HIF-1α negative feedback loop. Furthermore, signaling with initially activated glycolysis and radioresistance in CMM cells was impaired by Santacruzamate A, a histone deacetylase inhibitor, and 2-deoxy-D-glucose, a glycolytic inhibitor. Lastly, knockdown of LINC00518 expression sensitized CMM cancer cells to radiotherapy in an in vivo subcutaneously implanted tumor model. In conclusion, LINC00518 was confirmed to be an oncogene in CMM, which induces radioresistance by regulating glycolysis through an miR-33a-3p/HIF-1α negative feedback loop. Our study, may provide a potential strategy to improve the treatment outcome of radiotherapy in CMM.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Databases, Genetic
- Feedback, Physiological
- Gene Expression Regulation, Neoplastic
- Glycolysis
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Male
- Melanoma/genetics
- Melanoma/metabolism
- Melanoma/pathology
- Melanoma/radiotherapy
- Mice, Inbred BALB C
- Mice, Nude
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Neoplasm Invasiveness
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Radiation Tolerance
- Signal Transduction
- Skin Neoplasms/genetics
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Skin Neoplasms/radiotherapy
- Tumor Hypoxia
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Yan Liu
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, PR China
- Department of Plastic Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, PR China
| | - Dong He
- Department of Respiration, the Second People's Hospital of Hunan Province of Hunan University of Chinese Medicine, Changsha, 410000, PR China
| | - Mengqing Xiao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, PR China
| | - Yuxing Zhu
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, PR China
| | - Jianda Zhou
- Department of Plastic Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, PR China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, PR China.
| |
Collapse
|
48
|
Wu X, Zhao J. Novel oxidative stress-related prognostic biomarkers for melanoma associated with tumor metastasis. Medicine (Baltimore) 2021; 100:e24866. [PMID: 33663112 PMCID: PMC7909214 DOI: 10.1097/md.0000000000024866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/26/2021] [Accepted: 01/30/2021] [Indexed: 01/05/2023] Open
Abstract
ABSTRACT Skin cutaneous melanoma (SKCM) is a prevalent skin cancer whose metastatic form is dangerous due to its high morbidity and mortality. Previous studies have systematically established the vital role of oxidative stress (OS) in melanoma progression. This study aimed to identify prognostic OS genes closely associated with SKCM and illustrate their potential mechanisms. Transcriptome data and corresponding clinical traits of patients with SKCM were retrieved from The Cancer Genome Atlas and Gene Expression Omnibus databases. A weighted gene co-expression network analysis was conducted to identify relationships between clinical features and OS genes in specific modules. Subsequently, Cox regression analysis was performed on candidate OS genes; four hub prognosis-associated OS genes (AKAP9, VPS13C, ACSL4, and HMOX2) were identified to construct a prognostic model. After a series of bioinformatics analysis, our prognostic model was identified significantly associated with the overall survival of patients with SKCM and metastatic ability of the cancer. Furthermore, our risk model demonstrated improved diagnostic accuracy in the Cancer Genome Atlas and Gene Expression Omnibus cohorts. In addition, we established 2 nomograms based on either risk score or hub genes, which displayed favorable discriminating ability for SKCM. Our results provide novel insight into the potential applications of OS-associated genes in SKCM.
Collapse
Affiliation(s)
- Xianpei Wu
- Department of Orthopedics Trauma and Hand Surgery
| | - Jinmin Zhao
- Department of Orthopedics Trauma and Hand Surgery
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration
- Guangxi Collaborative Innovation Center for Biomedicine
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| |
Collapse
|
49
|
Gomes-Lima CJ, Shobab L, Wu D, Ylli D, Bikas A, McCoy M, Feldman R, Lee W, Rao SN, Jensen K, Vasko V, Castro LC, Jonklaas J, Wartofsky L, Burman KD. Do Molecular Profiles of Primary Versus Metastatic Radioiodine Refractory Differentiated Thyroid Cancer Differ? Front Endocrinol (Lausanne) 2021; 12:623182. [PMID: 33716974 PMCID: PMC7949910 DOI: 10.3389/fendo.2021.623182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Management of metastatic radioiodine refractory differentiated thyroid cancer (DTC) can be a therapeutic challenge. Generally, little is known about the paired molecular profile of the primary tumor and the metastases and whether they harbor the same genetic abnormalities. The present study compared the molecular profile of paired tumor specimens (primary tumor/metastatic sites) from patients with radioiodine refractory DTC in order to gain insight into a possible basis for resistance to radioiodine. Twelve patients with radioiodine refractory metastases were studied; median age at diagnosis of 61 years (range, 25-82). Nine patients had papillary TC (PTC), one had follicular TC (FTC), and two had Hürthle cell TC (HTC). Distant metastases were present in the lungs (n = 10), bones (n = 4), and liver (n = 1). The molecular profiling of paired tumors was performed with a panel of 592 genes for Next Generation Sequencing, RNA-sequencing, and immunohistochemistry. Digital microfluidic PCR was used to investigate TERT promoter mutations. The genetic landscape of all paired sites comprised BRAF, NRAS, HRAS, TP53, ATM, MUTYH, POLE, and NTRK genes, including BRAF and NTRK fusions. BRAF V600E was the most common point mutation in the paired specimens (5/12). TERT promoter mutation C228T was detected in one case. PD-L1 expression at metastatic sites was highly positive (95%) for one patient with HTC. All specimens were stable for microsatellite instability testing, and the tumor mutation burden was low to intermediate. Therefore, the molecular profile of DTC primary and metastatic lesions can show heterogeneity, which may help explain some altered responses to therapeutic intervention.
Collapse
Affiliation(s)
- Cristiane J. Gomes-Lima
- Department of Internal Medicine, MedStar Clinical Research Center, MedStar Health Research Institute (MHRI), Washington, DC, United States
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
- University of Brasilia School of Health Sciences, Postgraduate Program, Brasilia, Brazil
| | - Leila Shobab
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| | - Di Wu
- Department of Internal Medicine, MedStar Clinical Research Center, MedStar Health Research Institute (MHRI), Washington, DC, United States
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| | - Dorina Ylli
- Department of Internal Medicine, MedStar Clinical Research Center, MedStar Health Research Institute (MHRI), Washington, DC, United States
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| | - Athanasios Bikas
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Matthew McCoy
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, United States
| | - Rebecca Feldman
- Caris Life Sciences, Medical Affairs, Phoenix, AZ, United States
| | - Wen Lee
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, United States
| | - Sarika N. Rao
- Division of Endocrinology, Mayo Clinic, Jacksonville, FL, United States
| | - Kirk Jensen
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Vasily Vasko
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Luiz Claudio Castro
- Department of Pediatrics, University of Brasilia School of Medicine, Brasilia, Brazil
| | - Jacqueline Jonklaas
- Department of Medicine, Georgetown University, Washington, DC, United States
| | - Leonard Wartofsky
- Department of Internal Medicine, MedStar Clinical Research Center, MedStar Health Research Institute (MHRI), Washington, DC, United States
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| | - Kenneth D. Burman
- Section of Endocrinology, MedStar Washington Hospital Center, Washington, DC, United States
| |
Collapse
|
50
|
Transcript levels of keratin 1/5/6/14/15/16/17 as potential prognostic indicators in melanoma patients. Sci Rep 2021; 11:1023. [PMID: 33441834 PMCID: PMC7806772 DOI: 10.1038/s41598-020-80336-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Keratins (KRTs), the intermediate filament-forming proteins of epithelial cells, are extensively used as diagnostic biomarkers in cancers and associated with tumorigenesis and metastasis in multiple cancers. However, the diverse expression patterns and prognostic values of KRTs in melanoma have yet to be elucidated. In the current study, we examined the transcriptional and clinical data of KRTs in patients with melanoma from GEO, TCGA, ONCOMINE, GEPIA, cBioPortal, TIMER and TISIDB databases. We found that the mRNA levels of KRT1/2/5/6/8/10/14/15/16/17 were significantly differential expressed between primary melanoma and metastatic melanoma. The expression levels of KRT1/2/5/6/10/14/15/16/17 were correlated with advanced tumor stage. Survival analysis revealed that the high transcription levels of KRT1/5/6/14/15/16/17 were associated with low overall survival in melanoma patients. GSEA analysis indicated that the most involved hallmarks pathways were P53 pathway, KRAS signaling, estrogen response early and estrogen response late. Furthermore, we found some correlations among the expression of KRTs and the infiltration of immune cells. Our study may provide novel insights for the selection of prognostic biomarkers for melanoma.
Collapse
|