1
|
Chen Q, Wang H, Liu Q, Luo C. CTHRC1: a key player in colorectal cancer progression and immune evasion. Front Immunol 2025; 16:1579661. [PMID: 40201173 PMCID: PMC11975584 DOI: 10.3389/fimmu.2025.1579661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
The multifunctional secreted protein, collagen triple helix repeat containing 1 (CTHRC1), has recently emerged as a significant focus within oncology research. CTHRC1 expression in tumors is governed by a complex interplay of regulatory signals, including methylation, glycosylation, and notably, non-coding RNAs, which constitute its predominant regulatory mechanism. Colorectal cancer (CRC), a highly prevalent epithelial malignancy, sees CTHRC1 influencing tumor progression and metastasis through its modulation of several downstream signaling cascades, such as Wnt/PCP, TGF-β/Smad, and MEK/ERK pathways. Furthermore, CTHRC1 contributes to immune evasion in CRC via diverse mechanisms. It is intricately associated with macrophage phenotypic switching within the tumor microenvironment (TME), favoring M2 macrophage polarization and facilitating the infiltration of T cells and neutrophils. CTHRC1 is also instrumental in immune escape by driving the remodeling of the extracellular matrix through interactions with cancer-associated fibroblasts. Additionally, CTHRC1's roles extend to the regulation of hypoxia-related pathways, metabolism of glycolysis and fatty acids, and involvement in tumor angiogenesis, all of which support tumor immune evasion. Considering its multifaceted activities, CTHRC1 emerges as a promising therapeutic target in CRC, with the potential to enhance the outcomes of existing radiotherapeutic and immunotherapeutic regimens. This review endeavors to delineate the mechanistic and therapeutic landscapes of CTHRC1 in CRC. Through a comprehensive discussion of CTHRC1's diverse functions, we aim to provide insights that could pave the way for innovative approaches in cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Changjiang Luo
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
2
|
Cero C, House JS, Verdi V, Ferguson JL, Jima DD, Selmek AA, Patania OM, Dwyer JE, Wei BR, Lloyd DT, Shive HR. Profiling the cancer-prone microenvironment in a zebrafish model for MPNST. Oncogene 2025; 44:179-191. [PMID: 39511408 PMCID: PMC11725499 DOI: 10.1038/s41388-024-03210-1] [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: 01/11/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Microenvironmental contributions to soft tissue sarcoma progression are relatively undefined, particularly during sarcoma onset. Use of animal models to reveal these contributions is impeded by difficulties in discriminating between microenvironmental, precancerous, and cancer cells, and challenges in defining a precancerous microenvironment. We developed a zebrafish model that allows segregation of microenvironmental, precancerous, and cancerous cell populations by fluorescence-activated cell sorting. This model has high predilection for malignant peripheral nerve sheath tumor (MPNST), a type of soft tissue sarcoma that exhibits rapid, aggressive growth. Using RNA-seq, we profiled the transcriptomes of microenvironmental, precancerous, and cancer cells from our zebrafish MPNST model. We show broad activation of inflammation/immune-associated signaling networks, describe gene expression patterns that uniquely characterize the transition from precancerous to cancer ME, and identify macrophages as potential contributors to microenvironmental phenotypes. We identify conserved gene expression changes and candidate genes of interest by comparative genomics analysis of MPNST versus benign lesions in both humans and zebrafish. Finally, we functionally validate a candidate extracellular matrix protein, periostin (POSTN), in human MPNST. This work provides insight into how the microenvironment may regulate MPNST initiation and progression.
Collapse
Affiliation(s)
- Cheryl Cero
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Cancer Biology, Cancer Cell Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John S House
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, USA
| | - Vincenzo Verdi
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jordan L Ferguson
- State Laboratory of Public Health, North Carolina Department of Health and Human Services, Raleigh, NC, USA
| | - Dereje D Jima
- Center of Human Health and the Environment and Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - Aubrie A Selmek
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Jennifer E Dwyer
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dillon T Lloyd
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, USA
| | - Heather R Shive
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
3
|
Diaz MJ, Tran JT, Samia AM, Forouzandeh M, Grant-Kels JM, Montanez-Wiscovich ME. Integrated Analysis of Single-Cell and Bulk RNA Data Reveals Complexity and Significance of the Melanoma Interactome. Cancers (Basel) 2025; 17:148. [PMID: 39796775 PMCID: PMC11720022 DOI: 10.3390/cancers17010148] [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: 11/07/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Despite significant strides in anti-melanoma therapies, resistance and recurrence remain major challenges. A deeper understanding of the underlying biology of these challenges is necessary for developing more effective treatment paradigms. Methods: Melanoma single-cell data were retrieved from the Broad Single Cell Portal (SCP11). High-dimensional weighted gene co-expression network analysis (hdWGCNA), CellChat, and ligand-receptor relative crosstalk (RC) scoring were employed to evaluate intercellular and intracellular signaling. The prognostic value of key regulatory genes was assessed via Kaplan-Meier (KM) survival analysis using the 'SKCM-TCGA' dataset. Results: Twenty-seven (27) gene co-expression modules were identified via hdWGCNA. Notable findings include NRAS Q61L melanomas being enriched for modules involving C19orf10 and ARF4, while BRAF V600E melanomas were enriched for modules involving ALAS1 and MYO1B. Additionally, CellChat analysis highlighted several dominant signaling pathways, namely MHC-II, CD99, and Collagen-receptor signaling, with numerous significant ligand-receptor interactions from melanocytes, including CD99-CD99 communications with cancer-associated fibroblasts, endothelial cells, NK cells, and T-cells. KM analysis revealed that higher expression of SELL, BTLA, IL2RG, PDGFA, CLDN11, ITGB3, and SPN improved overall survival, while higher FGF5 expression correlated with worse survival. Protein-protein interaction network analysis further indicated significant interconnectivity among the identified prognostic genes. Conclusions: Overall, these insights underscore critical immune interactions and potential therapeutic targets to combat melanoma resistance, paving the way for more personalized and effective treatment strategies.
Collapse
Affiliation(s)
- Michael J. Diaz
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jasmine T. Tran
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
| | - Arthur M. Samia
- Department of Dermatology, University of Florida College of Medicine, Gainesville, FL 32606, USA; (A.M.S.)
| | - Mahtab Forouzandeh
- Department of Dermatology, University of Florida College of Medicine, Gainesville, FL 32606, USA; (A.M.S.)
| | - Jane M. Grant-Kels
- Department of Dermatology, University of Florida College of Medicine, Gainesville, FL 32606, USA; (A.M.S.)
- Department of Dermatology, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | | |
Collapse
|
4
|
Silkwood K, Dollinger E, Gervin J, Atwood S, Nie Q, Lander AD. Leveraging gene correlations in single cell transcriptomic data. BMC Bioinformatics 2024; 25:305. [PMID: 39294560 PMCID: PMC11411778 DOI: 10.1186/s12859-024-05926-z] [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: 11/15/2023] [Accepted: 09/09/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Many approaches have been developed to overcome technical noise in single cell RNA-sequencing (scRNAseq). As researchers dig deeper into data-looking for rare cell types, subtleties of cell states, and details of gene regulatory networks-there is a growing need for algorithms with controllable accuracy and fewer ad hoc parameters and thresholds. Impeding this goal is the fact that an appropriate null distribution for scRNAseq cannot simply be extracted from data in which ground truth about biological variation is unknown (i.e., usually). RESULTS We approach this problem analytically, assuming that scRNAseq data reflect only cell heterogeneity (what we seek to characterize), transcriptional noise (temporal fluctuations randomly distributed across cells), and sampling error (i.e., Poisson noise). We analyze scRNAseq data without normalization-a step that skews distributions, particularly for sparse data-and calculate p values associated with key statistics. We develop an improved method for selecting features for cell clustering and identifying gene-gene correlations, both positive and negative. Using simulated data, we show that this method, which we call BigSur (Basic Informatics and Gene Statistics from Unnormalized Reads), captures even weak yet significant correlation structures in scRNAseq data. Applying BigSur to data from a clonal human melanoma cell line, we identify thousands of correlations that, when clustered without supervision into gene communities, align with known cellular components and biological processes, and highlight potentially novel cell biological relationships. CONCLUSIONS New insights into functionally relevant gene regulatory networks can be obtained using a statistically grounded approach to the identification of gene-gene correlations.
Collapse
Affiliation(s)
- Kai Silkwood
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Emmanuel Dollinger
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
| | - Joshua Gervin
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Scott Atwood
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Qing Nie
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
| | - Arthur D Lander
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
5
|
Cao M, Ke D, Zhou H. The role and molecular mechanism of CTHRC1 in fibrosis. Life Sci 2024; 350:122745. [PMID: 38834096 DOI: 10.1016/j.lfs.2024.122745] [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: 04/15/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Fibrosis, a pathological state characterized by the excessive accumulation of extracellular matrix components, is primarily driven by the overactivation of fibroblasts. This condition becomes particularly pronounced under chronic inflammatory conditions. Fibrosis can occur in several tissues throughout the body. Among the notable discoveries in the study of fibrosis is the role of Collagen Triple Helix Repeat Containing-1 (CTHRC1), a protein that has emerged as a critical regulator in the fibrotic process. CTHRC1 is rapidly expressed on the outer membrane of fibroblasts and intimal smooth muscle cells following vascular injury, such as that induced by balloon angioplasty. This expression denotes the organism efforts to repair and restructure compromised tissue, signifying a critical component of the tissue repair mechanism in reaction to fibrosis. It plays a pivotal role in promoting cell migration and aiding tissue repair post-injury, contributing significantly to various pathophysiological processes including revascularization, bone formation, developmental morphological changes, inflammatory arthritis, and the progression of cancer. Significantly, researchers have observed marked expression of CTHRC1 across a variety of fibrotic conditions, closely associating it with the progression of the disease. Intervention with CTHRC1 can affect the occurrence and progression of fibrosis. This review aims to comprehensively explore the role and underlying mechanisms of CTHRC1 in fibrotic diseases, highlighting its potential as a key target for therapeutic interventions.
Collapse
Affiliation(s)
- Mingzhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, People's Republic of China
| | - Da Ke
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, People's Republic of China
| | - Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, People's Republic of China.
| |
Collapse
|
6
|
Ai C, Huang Z, Rong T, Shen W, Yang F, Li Q, Bi L, Li W. The impact of SOX4-activated CTHRC1 transcriptional activity regulating DNA damage repair on cisplatin resistance in lung adenocarcinoma. Electrophoresis 2024; 45:1408-1417. [PMID: 38629299 DOI: 10.1002/elps.202300255] [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: 11/02/2023] [Revised: 02/08/2024] [Accepted: 03/02/2024] [Indexed: 08/22/2024]
Abstract
Lung adenocarcinoma (LUAD) is the predominant subtype within the spectrum of lung malignancies. CTHRC1 has a pro-oncogenic role in various cancers. Here, we observed the upregulation of CTHRC1 in LUAD, but its role in cisplatin resistance in LUAD remains unclear. Bioinformatics analysis was employed to detect CTHRC1 and SRY-related HMG-box 4 (SOX4) expression in LUAD. Gene Set Enrichment Analysis predicted the enriched pathways related to CTHRC1. JASPAR and MotifMap databases predicted upstream transcription factors of CTHRC1. Pearson analysis was conducted to analyze the correlation between genes of interest. The interaction and binding relationship between CTHRC1 and SOX4 were validated through dual-luciferase and chromatin immunoprecipitation assays. Quantitative real-time polymerase chain reaction determined the expression of CTHRC1 and SOX4 genes. CCK-8 was performed to assess cell viability and calculate IC50 value. Flow cytometry examined the cell cycle. Comet assay and western blot assessed DNA damage. CTHRC1 and SOX4 were upregulated in LUAD. CTHRC1 exhibited higher expression in cisplatin-resistant A549 cells compared to cisplatin-sensitive A549 cells. Knockdown of CTHRC1 enhanced DNA damage during cisplatin treatment and increased the sensitivity of LUAD cells to cisplatin. Additionally, SOX4 modulated DNA damage repair (DDR) by activating CTHRC1 transcriptional activity, promoting cisplatin resistance in LUAD cells. SOX4 regulated DDR by activating CTHRC1, thereby enhancing cisplatin resistance in LUAD cells. The finding provides a novel approach to address clinical cisplatin resistance in LUAD, with CTHRC1 possibly serving as a candidate for targeted therapies in addressing cisplatin resistance within LUAD.
Collapse
Affiliation(s)
- Cheng Ai
- Department of Cardiothoracic Surgery, Bishan Hospital of Chongqing, Medical University, Chongqing, P. R. China
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The affiliated Hospital of Nanjing University Medical School, Nanjing, P. R. China
| | - Zhenhao Huang
- Department of Cardiothoracic Surgery, Bishan Hospital of Chongqing, Medical University, Chongqing, P. R. China
| | - Tenghao Rong
- Department of Cardiothoracic Surgery, Bishan Hospital of Chongqing, Medical University, Chongqing, P. R. China
| | - Wang Shen
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, P. R. China
| | - Fuyu Yang
- Department of Cardiothoracic Surgery, Bishan Hospital of Chongqing, Medical University, Chongqing, P. R. China
| | - Qiang Li
- Department of Cardiothoracic Surgery, Bishan Hospital of Chongqing, Medical University, Chongqing, P. R. China
| | - Lei Bi
- Department of Cardiothoracic Surgery, Bishan Hospital of Chongqing, Medical University, Chongqing, P. R. China
| | - Wen Li
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, P. R. China
| |
Collapse
|
7
|
Singh CK, Fernandez S, Chhabra G, Zaemisch GR, Nihal A, Swanlund J, Ansari N, Said Z, Chang H, Ahmad N. The role of collagen triple helix repeat containing 1 (CTHRC1) in cancer development and progression. Expert Opin Ther Targets 2024; 28:419-435. [PMID: 38686865 PMCID: PMC11189736 DOI: 10.1080/14728222.2024.2349686] [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: 12/27/2023] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Collagen triple helix repeat containing 1 (CTHRC1) is a protein that has been implicated in pro-migratory pathways, arterial tissue-repair processes, and inhibition of collagen deposition via the regulation of multiple signaling cascades. Studies have also demonstrated an upregulation of CTHRC1 in multiple cancers where it has been linked to enhanced proliferation, invasion, and metastasis. However, the understanding of the exact role and mechanisms of CTHRC1 in cancer is far from complete. AREAS COVERED This review focuses on analyzing the role of CTHRC1 in cancer as well as its associations with clinicopathologies and cancer-related processes and signaling. We have also summarized the available literature information regarding the role of CTHRC1 in tumor microenvironment and immune signaling. Finally, we have discussed the mechanisms associated with CTHRC1 regulations, and opportunities and challenges regarding the development of CTHRC1 as a potential target for cancer management. EXPERT OPINION CTHRC1 is a multifaceted protein with critical roles in cancer progression and other pathological conditions. Its association with lower overall survival in various cancers, and impact on the tumor immune microenvironment make it an intriguing target for further research and potential therapeutic interventions in cancer.
Collapse
Affiliation(s)
- Chandra K. Singh
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | - Sofia Fernandez
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | | | - Ayaan Nihal
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | - Jenna Swanlund
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | - Naveed Ansari
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | - Zan Said
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | - Hao Chang
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
- William S. Middleton VA Medical Center, Madison, Wisconsin, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
- William S. Middleton VA Medical Center, Madison, Wisconsin, USA
| |
Collapse
|
8
|
Kang B, Camps J, Fan B, Jiang H, Ibrahim MM, Hu X, Qin S, Kirchhoff D, Chiang DY, Wang S, Ye Y, Shen Z, Bu Z, Zhang Z, Roider HG. Parallel single-cell and bulk transcriptome analyses reveal key features of the gastric tumor microenvironment. Genome Biol 2022; 23:265. [PMID: 36550535 PMCID: PMC9773611 DOI: 10.1186/s13059-022-02828-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The tumor microenvironment (TME) has been shown to strongly influence treatment outcome for cancer patients in various indications and to influence the overall survival. However, the cells forming the TME in gastric cancer have not been extensively characterized. RESULTS We combine bulk and single-cell RNA sequencing from tumors and matched normal tissue of 24 treatment-naïve GC patients to better understand which cell types and transcriptional programs are associated with malignant transformation of the stomach. Clustering 96,623 cells of non-epithelial origin reveals 81 well-defined TME cell types. We find that activated fibroblasts and endothelial cells are most prominently overrepresented in tumors. Intercellular network reconstruction and survival analysis of an independent cohort imply the importance of these cell types together with immunosuppressive myeloid cell subsets and regulatory T cells in establishing an immunosuppressive microenvironment that correlates with worsened prognosis and lack of response in anti-PD1-treated patients. In contrast, we find a subset of IFNγ activated T cells and HLA-II expressing macrophages that are linked to treatment response and increased overall survival. CONCLUSIONS Our gastric cancer single-cell TME compendium together with the matched bulk transcriptome data provides a unique resource for the identification of new potential biomarkers for patient stratification. This study helps further to elucidate the mechanism of gastric cancer and provides insights for therapy.
Collapse
Affiliation(s)
- Boxi Kang
- BIOPIC, Beijing Advanced Innovation Centre for Genomics, and School of Life Sciences, Peking University, Beijing, China
| | - Jordi Camps
- Biomedical Data Science, Research & Early Development Oncology, Bayer AG, Berlin, Germany
| | - Biao Fan
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Hongpeng Jiang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Mahmoud M Ibrahim
- Biomedical Data Science, Research & Early Development preMed, Bayer AG, Wuppertal, Germany
| | - Xueda Hu
- BIOPIC, Beijing Advanced Innovation Centre for Genomics, and School of Life Sciences, Peking University, Beijing, China
| | - Shishang Qin
- BIOPIC, Beijing Advanced Innovation Centre for Genomics, and School of Life Sciences, Peking University, Beijing, China
| | - Dennis Kirchhoff
- Immuno Oncology, Research & Early Development Oncology, Bayer AG, Berlin, Germany
| | - Derek Y Chiang
- Biomedical Data Science, Research & Early Development Oncology, Bayer US, Cambridge, MA, USA
| | - Shan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Beijing, China
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
- Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Beijing, China
| | - Zhanlong Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China.
- Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, China.
- Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Beijing, China.
| | - Zhaode Bu
- Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Zemin Zhang
- BIOPIC, Beijing Advanced Innovation Centre for Genomics, and School of Life Sciences, Peking University, Beijing, China.
- Peking-Tsinghua Centre for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| | - Helge G Roider
- Oncology Precision Medicine, Research & Early Development Oncology, Bayer AG, Berlin, Germany.
| |
Collapse
|
9
|
A pan-cancer analysis of matrisome proteins reveals CTHRC1 and a related network as major ECM regulators across cancers. PLoS One 2022; 17:e0270063. [PMID: 36190948 PMCID: PMC9529084 DOI: 10.1371/journal.pone.0270063] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/02/2022] [Indexed: 11/07/2022] Open
Abstract
The extracellular matrix in the tumour microenvironment can regulate cancer cell growth and progression. A pan-cancer analysis of TCGA data from 30 cancer types, identified the top 5% of matrisome genes with amplifications or deletions in their copy number, that affect their expression and cancer survival. A similar analysis of matrisome genes in individual cancers identified CTHRC1 to be significantly altered. CTHRC1, a regulator of collagen synthesis, was identified as the most prominently upregulated matrisome gene of interest across cancers. Differential gene expression analysis identified 19 genes whose expression is increased with CTHRC1. STRING analysis of these genes classified them as ‘extracellular’, involved most prominently in ECM organization and cell adhesion. KEGG analysis showed their involvement in ECM-receptor and growth factor signalling. Cytohubba analysis of these genes revealed 13 hub genes, of which MMP13, POSTN, SFRP4, ADAMTS16 and FNDC1 were significantly altered in their expression with CTHRC1 and seen to affect survival across cancers. This could in part be mediated by their overlapping roles in regulating ECM (collagen or fibronectin) expression and organisation. In breast cancer tumour samples CTHRC1 protein levels are significantly upregulated with POSTN and MMP13, further supporting the need to evaluate their crosstalk in cancers.
Collapse
|
10
|
Dermawan JK, Vanoli F, Herviou L, Sung YS, Zhang L, Singer S, Tap WD, Benayed R, Bale TA, Benhamida JK, Dickson BC, Antonescu CR. Comprehensive genomic profiling of EWSR1/FUS::CREB translocation-associated tumors uncovers prognostically significant recurrent genetic alterations and methylation-transcriptional correlates. Mod Pathol 2022; 35:1055-1065. [PMID: 35347249 PMCID: PMC9329182 DOI: 10.1038/s41379-022-01023-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/22/2022]
Abstract
To elucidate the mechanisms underlying the divergent clinicopathologic spectrum of EWSR1/FUS::CREB translocation-associated tumors, we performed a comprehensive genomic analysis of fusion transcript variants, recurrent genetic alterations (mutations, copy number alterations), gene expression, and methylation profiles across a large cohort of tumor types. The distribution of the EWSR1/FUS fusion partners-ATF1, CREB1, and CREM-and exon involvement was significantly different across different tumor types. Our targeted sequencing showed that secondary genetic events are associated with tumor type rather than fusion type. Of the 39 cases that underwent targeted NGS testing, 18 (46%) had secondary OncoKB mutations or copy number alterations (29 secondary genetic events in total), of which 15 (52%) were recurrent. Secondary recurrent, but mutually exclusive, TERT promoter and CDKN2A mutations were identified only in clear cell sarcoma (CCS) and associated with worse overall survival. CDKN2A/B homozygous deletions were recurrent in angiomatoid fibrous histiocytoma (AFH) and restricted to metastatic cases. mRNA upregulation of MITF, CDH19, PARVB, and PFKP was found in CCS, compared to AFH, and correlated with a hypomethylated profile. In contrast, S100A4 and XAF1 were differentially upregulated and hypomethylated in AFH but not CCS. Unsupervised clustering of methylation profiles revealed that CREB family translocation-associated tumors form neighboring but tight, distinct clusters. A sarcoma methylation classifier was able to accurately match 100% of CCS cases to the correct methylation class; however, it was suboptimal when applied to other histologies. In conclusion, our comprehensive genomic profiling of EWSR1/FUS::CREB translocation-associated tumors uncovered mostly histotype, rather than fusion-type associated correlations in transcript variants, prognostically significant secondary genetic alterations, and gene expression and methylation patterns.
Collapse
Affiliation(s)
| | - Fabio Vanoli
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laurie Herviou
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yun-Shao Sung
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lei Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William D. Tap
- Department of Medicine, Sarcoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tejus A. Bale
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jamal K. Benhamida
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brendan C. Dickson
- Department of Pathology and Laboratory Medicine, Sinai Health System, Toronto, Ontario, Canada
| | | |
Collapse
|
11
|
Yang C, Huang T, Liang Y, Xue Y, Liang Y, Wei X, Meng F, Wei Q. CTHRC1 targeted by miR-30a-5p regulates cell adhesion, invasion and migration in lung adenocarcinoma. J Cardiothorac Surg 2022; 17:46. [PMID: 35313900 PMCID: PMC8935819 DOI: 10.1186/s13019-022-01788-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 03/13/2022] [Indexed: 12/29/2022] Open
Abstract
The morbidity of lung cancer ranks first among all cancers. Lung adenocarcinoma (LUAD) is a classification of lung cancer, and cell invasion and migration of LUAD are the main causes for its high mortality. Therefore, further exploring the potential mechanism of LUAD metastasis may provide bases for following targeted drug development and treatment of LUAD. In this study, clinical data as well as gene expression profiles were obtained from TCGA-LUAD and GEO to analyze CTHRC1 expression. The result found that CTHRC1 was significantly high in LUAD. Similar results were also discovered in 4 cancer cell lines. Moreover, overexpressed/knock-down CTHRC1 cell lines were constructed. It was uncovered that overexpressing CTHRC1 promoted LUAD cell migration and invasion, and inhibited cell adhesion, while knocked down CTHRC1 had the opposite effect. Afterward, the upstream miRNAs that regulated CTHRC1 were predicted by several bioinformatics websites. It was testified by dual-luciferase method that CTHRC1 was negatively mediated by miR-30a-5p. Overexpressed miR-30a-5p suppressed cell invasion/migration, and increased cell adhesion, while overexpressing CTHRC1 as well reversed such impacts. In conclusion, it was disclosed in this study that CTHRC1 worked as a cancer promoter in LUAD, and miR-30a-5p could target and downregulate CTHRC1 to regulate cell adhesion, and inhibited LUAD cell invasion and migration. These results elucidated at cellular level that upregulated CTHRC1 may be a marker protein for LUAD metastasis.
Collapse
|
12
|
Ding Y, Li M, Tayier T, Zhang M, Chen L, Feng S. Bioinformatics analysis of lncRNA‑associated ceRNA network in melanoma. J Cancer 2021; 12:2921-2932. [PMID: 33854593 PMCID: PMC8040875 DOI: 10.7150/jca.51851] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 02/15/2021] [Indexed: 01/06/2023] Open
Abstract
Melanoma is an extremely malignant tumor with early metastasis and high mortality. Little is known about the process of by which melanoma occurs, as its mechanism is very complex and only limited data are available on its long non-coding RNA (lncRNA)-associated competing endogenous RNAs (ceRNAs). The purpose of this study was to screen out potential prognostic molecules and identify a ceRNA network related to the occurrence of melanoma. We screened 169 differentially expressed mRNAs (DEmRNAs) from E-MTAB-1862 and GSE3189; gene ontology (GO) enrichment analysis showed that these genes were closely related to the development of skin. In the protein-protein interaction network, we screened out a total of 19 hub genes. Furthermore, we predicted the microRNAs (miRNAs) that regulate hub genes using the miRWalk database and then intersected these with GSE35579, resulting in nine DEmiRNAs. We also predicted the lncRNAs that regulate the miRNAs using the LncBasev.2 database. According to the ceRNA hypothesis, and based on the intersection of the DElncRNAs with merged GTEx and TCGA data, we obtained 20 DElncRNAs. A total of four DEmRNAs, nine DEmiRNAs, and 20 DElncRNAs were included in the ceRNA network. Based on Cox stepwise regression and survival analysis, we identified five biomarkers, ZSCAN16-AS1, LINC00520, XIST, DTL, and let-7a-5p, and obtained risk scores. The results showed that most of the differentially expressed genes were related to epithelial-mesenchymal transition (EMT) in melanoma. Finally, we obtained a LINC00520/let-7a-5p/DTL molecular regulatory network. These results suggest that ceRNA networks have an important role in evaluating the prognosis of patients with melanoma and provide a new experimental basis for exploring the EMT process in the development of melanoma.
Collapse
Affiliation(s)
- Yi Ding
- Department of Histology and Embryology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Min Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tuersong Tayier
- Department of Pharmacology, Pharmacy College, Xinjiang Medical University, Urumqi, China
| | - MeiLin Zhang
- Xinjiang Urumqi City Center Blood Station, Urumqi, China
| | - Long Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - ShuMei Feng
- Department of Histology and Embryology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
13
|
Blackburn NB, Leandro AC, Nahvi N, Devlin MA, Leandro M, Martinez Escobedo I, Peralta JM, George J, Stacy BA, deMaar TW, Blangero J, Keniry M, Curran JE. Transcriptomic Profiling of Fibropapillomatosis in Green Sea Turtles ( Chelonia mydas) From South Texas. Front Immunol 2021; 12:630988. [PMID: 33717164 PMCID: PMC7943941 DOI: 10.3389/fimmu.2021.630988] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/01/2021] [Indexed: 12/19/2022] Open
Abstract
Sea turtle fibropapillomatosis (FP) is a tumor promoting disease that is one of several threats globally to endangered sea turtle populations. The prevalence of FP is highest in green sea turtle (Chelonia mydas) populations, and historically has shown considerable temporal growth. FP tumors can significantly affect the ability of turtles to forage for food and avoid predation and can grow to debilitating sizes. In the current study, based in South Texas, we have applied transcriptome sequencing to FP tumors and healthy control tissue to study the gene expression profiles of FP. By identifying differentially expressed turtle genes in FP, and matching these genes to their closest human ortholog we draw on the wealth of human based knowledge, specifically human cancer, to identify new insights into the biology of sea turtle FP. We show that several genes aberrantly expressed in FP tumors have known tumor promoting biology in humans, including CTHRC1 and NLRC5, and provide support that disruption of the Wnt signaling pathway is a feature of FP. Further, we profiled the expression of current targets of immune checkpoint inhibitors from human oncology in FP tumors and identified potential candidates for future studies.
Collapse
Affiliation(s)
- Nicholas B. Blackburn
- Department of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Ana Cristina Leandro
- Department of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Nina Nahvi
- Sea Turtle Inc., South Padre Island, TX, United States
| | | | - Marcelo Leandro
- Department of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | | | - Juan M. Peralta
- Department of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Jeff George
- Sea Turtle Inc., South Padre Island, TX, United States
| | - Brian A. Stacy
- National Marine Fisheries Service, Office of Protected Resources, University of Florida, Gainesville, FL, United States
| | | | - John Blangero
- Department of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Megan Keniry
- Department of Biology, College of Sciences, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Joanne E. Curran
- Department of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| |
Collapse
|
14
|
The Role of CTHRC1 in Regulation of Multiple Signaling and Tumor Progression and Metastasis. Mediators Inflamm 2020; 2020:9578701. [PMID: 32848510 PMCID: PMC7441421 DOI: 10.1155/2020/9578701] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
Collagen triple helix repeat containing-1 (CTHRC1) has been identified as cancer-related protein. CTHRC1 expresses mainly in adventitial fibroblasts and neointimal smooth muscle cells of balloon-injured vessels and promotes cell migration and tissue repair in response to injury. CTHRC1 plays a pivotal role in some pathophysiological processes, including increasing bone mass, preventing myelination, and reversing collagen synthesis in many tumor cells. The ascended expression of CTHRC1 is related to tumorigenesis, proliferation, invasion, and metastasis in various human malignancies, including gastric cancer, pancreatic cancer, hepatocellular carcinoma, keloid, breast cancer, colorectal cancer, epithelial ovarian cancer, esophageal squamous cell carcinoma, cervical cancer, non-small-cell lung carcinoma, and melanoma. And molecules that regulate the expression of CTHRC1 include miRNAs, lncRNAs, WAIF1, and DPAGT1. Many reports have pointed that CTHRC1 could exert different effects through several signaling pathways such as TGF-β, Wnt, integrin β/FAK, Src/FAK, MEK/ERK, PI3K/AKT/ERK, HIF-1α, and PKC-δ/ERK signaling pathways. As a participant in tissue remodeling or immune response, CTHRC1 may promote early-stage cancer. Several recent studies have identified CTHRC1 as an effectual prognostic biomarker for predicting tumor recurrence or metastasis. It is worth noting that CTHRC1 has different cellular localization and mechanisms of action in different cells and different microenvironments. In this article, we focus on the advances in the signaling pathways mediated by CTHRC1 in tumors.
Collapse
|
15
|
Gene Expression Signature of BRAF Inhibitor Resistant Melanoma Spheroids. Pathol Oncol Res 2020; 26:2557-2566. [PMID: 32613561 PMCID: PMC7471197 DOI: 10.1007/s12253-020-00837-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
In vitro cell cultures are frequently used to define the molecular background of drug resistance. The majority of currently available data have been obtained from 2D in vitro cultures, however, 3D cell culture systems (spheroids) are more likely to behave similarly to in vivo conditions. Our major aim was to compare the gene expression signature of 2D and 3D cultured BRAFV600E mutant melanoma cell lines. We successfully developed BRAF-drug resistant cell lines from paired primary/metastatic melanoma cell lines in both 2D and 3D in vitro cultures. Using Affymetrix Human Gene 1.0 ST arrays, we determined the gene expression pattern of all cell lines. Our analysis revealed 1049 genes (562 upregulated and 487 downregulated) that were differentially expressed between drug-sensitive cells grown under different cell cultures. Pathway analysis showed that the differently expressed genes were mainly associated with the cell cycle, p53, and other cancer-related pathways. The number of upregulated genes (72 genes) was remarkably fewer when comparing the resistant adherent cells to cells that grow in 3D, and were associated with cell adhesion molecules and IGF1R signalling. Only 1% of the upregulated and 5.6% of the downregulated genes were commonly altered between the sensitive and the resistant spheroids. Interestingly, we found several genes (BNIP3, RING1 and ABHD4) with inverse expression signature between sensitive and resistant spheroids, which are involved in anoikis resistance and cell cycle regulation. In summary, our study highlights gene expression alterations that might help to understand the development of acquired resistance in melanoma cells in tumour tissue.
Collapse
|
16
|
Zhang L, Wang Q, Wang L, Xie L, An Y, Zhang G, Zhu W, Li Y, Liu Z, Zhang X, Tang P, Huo X, Guo X. OSskcm: an online survival analysis webserver for skin cutaneous melanoma based on 1085 transcriptomic profiles. Cancer Cell Int 2020; 20:176. [PMID: 32467670 PMCID: PMC7236197 DOI: 10.1186/s12935-020-01262-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022] Open
Abstract
Background Cutaneous melanoma is one of the most aggressive and lethal skin cancers. It is greatly important to identify prognostic biomarkers to guide the clinical management. However, it is technically challenging for untrained researchers to process high dimensional profiling data and identify potential prognostic genes in profiling datasets. Methods In this study, we developed a webserver to analyze the prognostic values of genes in cutaneous melanoma using data from TCGA and GEO databases. The webserver is named Online consensus Survival webserver for Skin Cutaneous Melanoma (OSskcm) which includes 1085 clinical melanoma samples. The OSskcm is hosted in a windows tomcat server. Server-side scripts were developed in Java script. The database system is managed by a SQL Server, which integrates gene expression data and clinical data. The Kaplan–Meier (KM) survival curves, Hazard ratio (HR) and 95% confidence interval (95%CI) were calculated in a univariate Cox regression analysis. Results In OSskcm, by inputting official gene symbol and selecting proper options, users could obtain KM survival plot with log-rank P value and HR on the output web page. In addition, clinical characters including race, stage, gender, age and type of therapy could also be included in the prognosis analysis as confounding factors to constrain the analysis in a subgroup of melanoma patients. Conclusion The OSskcm is highly valuable for biologists and clinicians to perform the assessment and validation of new or interested prognostic biomarkers for melanoma. OSskcm can be accessed online at: http://bioinfo.henu.edu.cn/Melanoma/MelanomaList.jsp.
Collapse
Affiliation(s)
- Lu Zhang
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Qiang Wang
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Lijie Wang
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Longxiang Xie
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Yang An
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Guosen Zhang
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Wan Zhu
- 3Department of Anesthesia, Stanford University, Stanford, CA 94305 USA
| | - Yongqiang Li
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Zhihui Liu
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Xiaochen Zhang
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Panpan Tang
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Xiaozheng Huo
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China
| | - Xiangqian Guo
- 1Department of Preventive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, School of Software, School of Basic Medical Sciences, Henan University, Kaifeng, 475004 Henan China.,2Henan Provincial Engineering Centre for Tumor Molecular Medicine, Henan University, Kaifeng, 475004 Henan China
| |
Collapse
|
17
|
Eriksson J, Le Joncour V, Jahkola T, Juteau S, Laakkonen P, Saksela O, Hölttä E. Prolyl 4-hydroxylase subunit alpha 1 (P4HA1) is a biomarker of poor prognosis in primary melanomas, and its depletion inhibits melanoma cell invasion and disrupts tumor blood vessel walls. Mol Oncol 2020; 14:742-762. [PMID: 32053263 PMCID: PMC7138405 DOI: 10.1002/1878-0261.12649] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 01/15/2020] [Accepted: 02/11/2020] [Indexed: 12/19/2022] Open
Abstract
Melanoma is an unpredictable, highly metastatic malignancy, and treatment of advanced melanoma remains challenging. Novel molecular markers based on the alterations in gene expression and the molecular pathways activated or deactivated during melanoma progression are needed for predicting the course of the disease already in primary tumors and for providing new targets for therapy. Here, we sought to identify genes whose expression in primary melanomas correlate with patient disease‐specific survival using global gene expression profiling. Many of the identified potential markers of poor prognosis were associated with the epithelial–mesenchymal transition, extracellular matrix formation, and angiogenesis. We studied further the significance of one of the genes, prolyl 4‐hydroxylase subunit alpha 1 (P4HA1), in melanoma progression. P4HA1 depletion in melanoma cells reduced cell adhesion, invasion, and viability in vitro. In melanoma xenograft assays, we found that P4HA1 knockdown reduced melanoma tumor invasion as well as the deposition of collagens, particularly type IV collagen, in the interstitial extracellular matrix and in the basement membranes of tumor blood vessels, leading to vessel wall rupture and hemorrhages. Further, P4HA1 knockdown reduced the secretion of collagen triple helix repeat containing 1 (CTHRC1), an important mediator of melanoma cell migration and invasion, in vitro and its deposition around tumor blood vessels in vivo. Taken together, P4HA1 is an interesting potential prognostic marker and therapeutic target in primary melanomas, influencing many aspects of melanoma tumor progression.
Collapse
Affiliation(s)
| | - Vadim Le Joncour
- Faculty of Medicine, Translational Cancer Medicine Research Program, University of Helsinki, Finland
| | - Tiina Jahkola
- Department of Plastic Surgery, Helsinki University Hospital, Finland
| | - Susanna Juteau
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Finland
| | - Pirjo Laakkonen
- Faculty of Medicine, Translational Cancer Medicine Research Program, University of Helsinki, Finland.,Laboratory Animal Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Finland
| | - Olli Saksela
- Department of Dermatology, Helsinki University Hospital, Finland
| | - Erkki Hölttä
- Department of Pathology, University of Helsinki, Finland
| |
Collapse
|
18
|
Xu Y, Mu Y, Wang L, Zhang X. Detailed Analysis of Molecular Mechanisms in Primary and Metastatic Melanoma. J Comput Biol 2020; 27:9-19. [PMID: 31424282 DOI: 10.1089/cmb.2019.0197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yangchun Xu
- Department of Dermatology and Second Hospital of Jilin University, Changchun, China
| | - Yan Mu
- Department of Dermatology and Second Hospital of Jilin University, Changchun, China
| | - Ling Wang
- Department of Gynecology, Second Hospital of Jilin University, Changchun, China
| | - Xuan Zhang
- Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Hiroshima Y, Kasajima R, Kimura Y, Komura D, Ishikawa S, Ichikawa Y, Bouvet M, Yamamoto N, Oshima T, Morinaga S, Singh SR, Hoffman RM, Endo I, Miyagi Y. Novel targets identified by integrated cancer-stromal interactome analysis of pancreatic adenocarcinoma. Cancer Lett 2020; 469:217-227. [PMID: 31669204 DOI: 10.1016/j.canlet.2019.10.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/03/2019] [Accepted: 10/18/2019] [Indexed: 12/27/2022]
|
20
|
Rittler D, Baranyi M, Molnár E, Garay T, Jalsovszky I, Varga IK, Hegedűs L, Aigner C, Tóvári J, Tímár J, Hegedűs B. The Antitumor Effect of Lipophilic Bisphosphonate BPH1222 in Melanoma Models: The Role of the PI3K/Akt Pathway and the Small G Protein Rheb. Int J Mol Sci 2019; 20:ijms20194917. [PMID: 31623406 PMCID: PMC6801414 DOI: 10.3390/ijms20194917] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/24/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
Malignant melanoma is one of the most metastatic cancer types, and despite recent success with novel treatment strategies, there is still a group of patients who do not respond to any therapies. Earlier, the prenylation inhibitor hydrophilic bisphosphonate zoledronic acid (ZA) was found to inhibit melanoma growth in vitro, but only a weaker effect was observed in vivo due to its hydrophilic properties. Recently, lipophilic bisphosphonates (such as BPH1222) were developed. Accordingly, for the first time, we compared the effect of BPH1222 to ZA in eight melanoma lines using viability, cell-cycle, clonogenic and spheroid assays, videomicroscopy, immunoblot, and xenograft experiments. Based on 2D and spheroid assays, the majority of cell lines were more sensitive to BPH. The activation of Akt and S6 proteins, but not Erk, was inhibited by BPH. Additionally, BPH had a stronger apoptotic effect than ZA, and the changes of Rheb showed a correlation with apoptosis. In vitro, only M24met cells were more sensitive to ZA than to BPH; however, in vivo growth of M24met was inhibited more strongly by BPH. Here, we present that lipophilic BPH is more effective on melanoma cells than ZA and identify the PI3K pathway, particularly Rheb as an important mediator of growth inhibition.
Collapse
Affiliation(s)
- Dominika Rittler
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
| | - Marcell Baranyi
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
| | - Eszter Molnár
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
| | - Tamás Garay
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
- Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, H-1083 Budapest, Hungary.
- Oncology Center, Semmelweis University, H-1091 Budapest, Hungary.
| | - István Jalsovszky
- Eötvös Loránd University, Faculty of Science, Institute of Chemistry, Department of Organic Chemistry; H-1117 Budapest, Hungary.
| | - Imre Károly Varga
- Eötvös Loránd University, Faculty of Science, Institute of Chemistry, Department of Organic Chemistry; H-1117 Budapest, Hungary.
| | - Luca Hegedűs
- Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, D-45239 Essen, Germany.
| | - Clemens Aigner
- Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, D-45239 Essen, Germany.
| | - József Tóvári
- Department of Experimental Pharmacology, National Institute of Oncology, H-1122 Budapest, Hungary.
| | - József Tímár
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
| | - Balázs Hegedűs
- Department of Pathology, Semmelweis University, H-1091 Budapest, Hungary.
- Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, D-45239 Essen, Germany.
| |
Collapse
|
21
|
Perotti V, Baldassari P, Molla A, Nicolini G, Bersani I, Grazia G, Benigni F, Maurichi A, Santinami M, Anichini A, Mortarini R. An actionable axis linking NFATc2 to EZH2 controls the EMT-like program of melanoma cells. Oncogene 2019; 38:4384-4396. [PMID: 30710146 PMCID: PMC6756060 DOI: 10.1038/s41388-019-0729-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/03/2018] [Accepted: 01/21/2019] [Indexed: 01/30/2023]
Abstract
Discovery of new actionable targets and functional networks in melanoma is an urgent need as only a fraction of metastatic patients achieves durable clinical benefit by targeted therapy or immunotherapy approaches. Here we show that NFATc2 expression is associated with an EMT-like transcriptional program and with an invasive melanoma phenotype, as shown by analysis of melanoma cell lines at the mRNA and protein levels, interrogation of the TCGA melanoma dataset and characterization of melanoma lesions by immunohistochemistry. Gene silencing or pharmacological inhibition of NFATc2 downregulated EMT-related genes and AXL, and suppressed c-Myc, FOXM1, and EZH2. Targeting of c-Myc suppressed FOXM1 and EZH2, while targeting of FOXM1 suppressed EZH2. Inhibition of c-Myc, or FOXM1, or EZH2 downregulated EMT-related gene expression, upregulated MITF and suppressed migratory and invasive activity of neoplastic cells. Stable silencing of NFATc2 impaired melanoma cell proliferation in vitro and tumor growth in vivo in SCID mice. In NFATc2+ EZH2+ melanoma cell lines pharmacological co-targeting of NFATc2 and EZH2 exerted strong anti-proliferative and pro-apoptotic activity, irrespective of BRAF or NRAS mutations and of BRAF inhibitor resistance. These results provide preclinical evidence for a role of NFATc2 in shaping the EMT-like melanoma phenotype and reveal a targetable vulnerability associated with NFATc2 and EZH2 expression in melanoma cells belonging to different mutational subsets.
Collapse
Affiliation(s)
- Valentina Perotti
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Paola Baldassari
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Alessandra Molla
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | | | - Ilaria Bersani
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Giulia Grazia
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Fabio Benigni
- HuMabs Biomed, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Andrea Maurichi
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Mario Santinami
- Melanoma and Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Anichini
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy
| | - Roberta Mortarini
- Department of Research, Human Tumors Immunobiology Unit, Milan, Italy.
| |
Collapse
|
22
|
Zheng M, Zhou Q, Liu X, Wang C, Liu G. CTHRC1 overexpression promotes cervical carcinoma progression by activating the Wnt/PCP signaling pathway. Oncol Rep 2019; 41:1531-1538. [PMID: 30628714 PMCID: PMC6365690 DOI: 10.3892/or.2019.6963] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/07/2019] [Indexed: 12/15/2022] Open
Abstract
The tumorigenesis and metastasis of tumors are associated with human collagen triple helix repeats containing 1 (CTHRC1). To study the effects and possible impacting mechanisms of CTHRC1 on human cervical carcinoma development, samples of paraffin‑embedded cervical carcinoma and HeLa cells were examined. Immunofluorescence, cell wound scratch assay, western blot analysis and Transwell invasion assay were used to evaluate HeLa cells in response to silencing of the CTHRC1 gene in cervical carcinoma. The expression levels of gap‑associated proteins of the Wnt/PCP pathway in paraffin‑embedded cervical carcinoma samples were also evaluated by immunohistochemical staining. CTHRC1 promoted the migration and invasion of HeLa cells in vitro, downregulated Ror2 and p‑c‑Jun and activated the Wnt/PCP pathway. Furthermore, the expression of p‑c‑Jun, Ror2 and Wnt5a was increased after overexpression of CTHRC1 as revealed in HeLa cells compared to control group. The present experiments revealed that CTHRC1 promoted HeLa cell progression by activating the Wnt/PCP signaling pathway and may play a key role in the invasion and metastasis of cervical carcinoma.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Traditional Chinese Medicine, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Qin Zhou
- School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, Shandong 250012, P.R. China
| | - Xingguang Liu
- School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, Shandong 250012, P.R. China
| | - Chengze Wang
- School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, Shandong 250012, P.R. China
| | - Gangli Liu
- School of Stomatology, Shandong University, Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
23
|
Kielosto M, Eriksson J, Nummela P, Yin M, Hölttä E. Divergent roles of lysyl oxidase family members in ornithine decarboxylase- and RAS-transformed mouse fibroblasts and human melanoma cells. Oncotarget 2018; 9:37733-37752. [PMID: 30701028 PMCID: PMC6340875 DOI: 10.18632/oncotarget.26508] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 12/13/2018] [Indexed: 12/12/2022] Open
Abstract
We have previously shown that proto-oncoprotein c-Jun is activated in ornithine decarboxylase (ODC)- and RAS-transformed mouse fibroblasts, and that the transformed morphology of these cells can be reversed by expressing the transactivation domain deletion mutant of c-Jun (TAM67). Here, we found that lysyl oxidase (Lox), encoding an extracellular matrix-modifying enzyme, is downregulated in a c-Jun-dependent manner in ODC-transformed fibroblasts (Odc cells). In addition to Lox, the Lox family members Lox-like 1 and 3 (Loxl1 and Loxl3) were found to be downregulated in Odc as well as in RAS-transformed fibroblasts (E4), whereas Lox-like 4 (Loxl4) was upregulated in Odc and downregulated in E4 cells compared to normal N1 fibroblasts. Tetracycline-regulatable LOX re-expression in Odc cells led to inhibition of cell growth and invasion in three-dimensional Matrigel in an activity-independent manner. On the contrary, LOX and especially LOXL2, LOXL3, and LOXL4 were found to be upregulated in several human melanoma cell lines, and LOX inhibitor B-aminopropionitrile inhibited the invasive growth of these cells particularly when co-cultured with fibroblasts in Matrigel. Knocking down the expression of LOX and especially LOXL2 in melanoma cells almost completely abrogated the invasive growth capability. Further, LOXL2 was significantly upregulated in clinical human primary melanomas compared to benign nevi, and high expression of LOXL2 in primary melanomas was associated with formation of metastases and shorter survival of patients. Thus, our studies reveal that inactive pro-LOX (together with Lox propeptide) functions as a tumor suppressor in ODC- and RAS-transformed murine fibroblasts by inhibiting cell growth and invasion, and active LOX and LOXL2 as tumor promoters in human melanoma cells by promoting their invasive growth.
Collapse
Affiliation(s)
- Mari Kielosto
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Johanna Eriksson
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Pirjo Nummela
- Department of Pathology, University of Helsinki, Helsinki, Finland.,Current address: University of Helsinki, Genome-Scale Biology Research Program, Helsinki, Finland
| | - Miao Yin
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Erkki Hölttä
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Ni S, Ren F, Xu M, Tan C, Weng W, Huang Z, Sheng W, Huang D. CTHRC1 overexpression predicts poor survival and enhances epithelial-mesenchymal transition in colorectal cancer. Cancer Med 2018; 7:5643-5654. [PMID: 30302922 PMCID: PMC6247052 DOI: 10.1002/cam4.1807] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/19/2018] [Accepted: 09/09/2018] [Indexed: 12/14/2022] Open
Abstract
Collagen triple helix repeat containing (CTHRC1), which was identified as a cancer-related factor, is a promigratory protein involved in multiple processes, including vascular remodeling, antifibrosis, metabolism, bone formation, and cancer. In this study, we aimed to investigate the clinical significance and possible role of CTHRC1 in the process of epithelial-mesenchymal transition (EMT) in colorectal cancer (CRC). Here, we revealed that CTHRC1 mRNA and protein levels are both upregulated in CRC tissues compared with those of paired noncancerous tissues. Moreover, the overexpression of CTHRC1 correlated with poor prognosis in patients with CRC (especially colon cancer). Furthermore, we showed that CTHRC1 induced EMT and promoted cell motility in CRC cells. Importantly, we demonstrated that CTHRC1 promoted EMT by activating transforming growth factor-β (TGF-β) signaling, revealing a possible effective therapeutic treatment for patients with CRC.
Collapse
Affiliation(s)
- Shujuan Ni
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
| | - Fei Ren
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
| | - Midie Xu
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
| | - Cong Tan
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
| | - Weiwei Weng
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
| | - Zhaohui Huang
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiChina
| | - Weiqi Sheng
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
| | - Dan Huang
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Institute of PathologyFudan UniversityShanghaiChina
| |
Collapse
|
25
|
Xu G, Fan W, Wang F, Lu H, Xing X, Zhang R, Jiang P. CTHRC1 as a novel biomarker in the diagnosis of cervical squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:847-854. [PMID: 31938174 PMCID: PMC6958054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 11/07/2017] [Indexed: 06/10/2023]
Abstract
To verify whether the collagen triple helix repeat containing 1 (CTHRC1) can be used as a potential biomarker in diagnosis of cervical cancer, by evaluating the expression level of CTHRC1 in cervical squamous cell carcinoma patients. In this study, CTHRC1 expression in cervical squamous cell carcinoma, CIN (cervical intraepithelial neoplasia) and healthy cervical squamous epithelium were measured by immunohistochemistry. The serum levels of CTHRC1 and SCC-Ag within the all three groups were performed using ELISA. In addition, the ROC curve of CTHRC1 and SCC-Ag as well as combined CTHRC1 and SCC-Ag was demonstrated and analyzed. CTHRC1 was significantly overexpressed in cervical squamous cell carcinoma compared with CIN and healthy control group. And CTHRC1 concentration in serum of cervical squamous cell carcinoma group was also remarkably higher than that in other two groups. The ROC curve showed AUC of CTHRC1 and SCC-Ag was 0.665±0.034 and 0.878±0.027, the sensitivity of them were 57.1% and 77.6%, and the specificity of them were 85.4% and 86%, respectively. Furthermore, AUC of combined CTHRC1 and SCC-Ag was 0.879±0.027, sensitivity was 87.2% and specificity were 84%. Our study indicated that CTHRC1 was highly upregulated not only in the tissue but also in the serum of cervical squamous cell carcinoma patients, which pointed out it can be used as a novel prognostic and metastatic biomarker of cervical squamous cell carcinoma. And combined SCC-Ag and CTHRC1 serological detection may have potential value in the early diagnosis of cervical squamous cell carcinoma and CIN.
Collapse
Affiliation(s)
- Guangxu Xu
- Department of Obstetrics and Gynecology, Fengxian Hospital, Southern Medical UniversityShanghai, P. R. China
| | - Weirong Fan
- Department of Obstetrics and Gynecology, Fengxian Hospital, Southern Medical UniversityShanghai, P. R. China
- Department of Obstetrics and Gynecology, Jinhua Maternity and Child Care HospitalJinhua, Zhejiang, P. R. China
| | - Fengmian Wang
- Department of Obstetrics and Gynecology, Fengxian Hospital, Southern Medical UniversityShanghai, P. R. China
| | - Huan Lu
- Department of Obstetrics and Gynecology, Fengxian Hospital, Southern Medical UniversityShanghai, P. R. China
| | - Xin Xing
- Department of Obstetrics and Gynecology, Fengxian Hospital, Southern Medical UniversityShanghai, P. R. China
| | - Rong Zhang
- Department of Obstetrics and Gynecology, Fengxian Hospital, Southern Medical UniversityShanghai, P. R. China
| | - Pengcheng Jiang
- Department of Obstetrics and Gynecology, The Affiliated Changzhou No.2 People’s Hospital of Nanjing Medical UniversityChangzhou, Jiangsu, P. R. China
| |
Collapse
|
26
|
Dai Y, Wang Q, Gonzalez Lopez A, Anders M, Malfertheiner P, Vieth M, Kemmner W. Genome-Wide Analysis of Barrett's Adenocarcinoma. A First Step Towards Identifying Patients at Risk and Developing Therapeutic Paths. Transl Oncol 2017; 11:116-124. [PMID: 29223109 PMCID: PMC6002392 DOI: 10.1016/j.tranon.2017.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/12/2017] [Accepted: 10/23/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND: Barrett's esophagus metaplasia is the key precursor lesion of esophageal adenocarcinoma. The aim of this study was to find a subset of markers that may allow the identification of patients at risk for esophageal adenocarcinoma, and to determine genes differentially expressed in esophageal squamous cell carcinoma. METHODS: Laser capture microdissection technique was applied to procure cells from defined regions. Genome-wide RNA profiling was performed on esophageal adenocarcinoma (n = 21), Barrett's esophagus (n = 20), esophageal squamous carcinoma (n = 9) and healthy esophageal biopsies (n = 18) using the Affymetrix Human Genome U133plus 2.0 array. Microarray results were validated by quantitative real-time polymerase chain reaction in a second and independent cohort and by immunohistochemistry of two putative markers in a third independent cohort. RESULTS: Through unsupervised hierarchical clustering and principal component analysis, samples were separated into four distinct groups that match perfectly with histology. Many genes down-regulated in esophageal cancers belong to the epidermal differentiation complex or the related GO-group “cornified envelope” of terminally differentiated keratinocytes. Similarly, retinol metabolism was strongly down-regulated. Genes showing strong overexpression in esophageal carcinomas belong to the GO groups extracellular region /matrix such as MMP1, CTHRC1, and INHBA. According to an analysis of genes strongly up-regulated in both esophageal adenocarcinoma and Barrett's esophagus, REG4 might be of particular interest as an early marker for esophageal adenocarcinoma. CONCLUSIONS: Our study provides high quality data, which could serve for identification of potential biomarkers of Barrett's esophagus at risk of esophageal adenocarcinoma progression.
Collapse
Affiliation(s)
- Yiyang Dai
- Translational Oncology, Experimental Clinical Research Center (ECRC), Charité Campus Buch, 13125 Berlin, Germany
| | - Qing Wang
- Translational Oncology, Experimental Clinical Research Center (ECRC), Charité Campus Buch, 13125 Berlin, Germany
| | - Adrian Gonzalez Lopez
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité Campus Virchow-Klinikum, 13353 Berlin, Germany
| | - Mario Anders
- Gastroenterologie und Interdisziplinäre Endoskopie, Vivantes Wenckebach-Klinikum, 12099 Berlin, Germany
| | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, 95445 Bayreuth, Germany
| | - Wolfgang Kemmner
- Translational Oncology, Experimental Clinical Research Center (ECRC), Charité Campus Buch, 13125 Berlin, Germany.
| |
Collapse
|
27
|
Metri R, Mohan A, Nsengimana J, Pozniak J, Molina-Paris C, Newton-Bishop J, Bishop D, Chandra N. Identification of a gene signature for discriminating metastatic from primary melanoma using a molecular interaction network approach. Sci Rep 2017; 7:17314. [PMID: 29229936 PMCID: PMC5725601 DOI: 10.1038/s41598-017-17330-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/10/2017] [Indexed: 01/15/2023] Open
Abstract
Understanding the biological factors that are characteristic of metastasis in melanoma remains a key approach to improving treatment. In this study, we seek to identify a gene signature of metastatic melanoma. We configured a new network-based computational pipeline, combined with a machine learning method, to mine publicly available transcriptomic data from melanoma patient samples. Our method is unbiased and scans a genome-wide protein-protein interaction network using a novel formulation for network scoring. Using this, we identify the most influential, differentially expressed nodes in metastatic as compared to primary melanoma. We evaluated the shortlisted genes by a machine learning method to rank them by their discriminatory capacities. From this, we identified a panel of 6 genes, ALDH1A1, HSP90AB1, KIT, KRT16, SPRR3 and TMEM45B whose expression values discriminated metastatic from primary melanoma (87% classification accuracy). In an independent transcriptomic data set derived from 703 primary melanomas, we showed that all six genes were significant in predicting melanoma specific survival (MSS) in a univariate analysis, which was also consistent with AJCC staging. Further, 3 of these genes, HSP90AB1, SPRR3 and KRT16 remained significant predictors of MSS in a joint analysis (HR = 2.3, P = 0.03) although, HSP90AB1 (HR = 1.9, P = 2 × 10-4) alone remained predictive after adjusting for clinical predictors.
Collapse
Affiliation(s)
- Rahul Metri
- IISc Mathematics Initiative (IMI), Indian Institute of Science, Bangalore, Karnataka, India
| | - Abhilash Mohan
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Jérémie Nsengimana
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Joanna Pozniak
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Carmen Molina-Paris
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds, UK
| | - Julia Newton-Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - David Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Nagasuma Chandra
- IISc Mathematics Initiative (IMI), Indian Institute of Science, Bangalore, Karnataka, India.
- Department of Biochemistry, Indian Institute of Science, Bangalore, Karnataka, India.
| |
Collapse
|
28
|
Li Y, Zhang Y, Ma C, Wang S, Li N, Wang J, Ma G, Zhang L. Overexpression of CTHRC1 in human melanoma promotes tumorigenesis targeted by miRNA155. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8199-8210. [PMID: 31966671 PMCID: PMC6965430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/19/2017] [Indexed: 06/10/2023]
Abstract
Most of human malignant melanoma is metastatic and invasive, which has poor therapeutic response and poor prognosis as well as high incidence worldwide. CTHRC1 was overexpressed in metastatic primary melanomas. The relationship between CTHRC1 and miRNA in affecting the melanoma has not been studied. This study was aimed to explore the function of CTHRC1 in human melanoma and finding its regulator miRNA. CTHRC1 and miR155 expression in cancer tissues and paracancer tissues was examined. The cell proliferation, cell cycle, cell migration and cell invasion was evaluated in M21 cell transfected with CTHRC1 siRNA or miR155. The mRNA expression of miR155 in cancer tissues was lower than that in paracancer tissues. The mRNA expression of CTHRC1 in the cancer tissues was higher than that in corresponding paracancer tissues. The mRNA expression of CTHRC1 in the M21 cells was decreased by treated with miR155 mimic. CTHRC1 promoted the cell proliferation, cell cycle process, cell migration and invasion capacity and prevented the cell from apoptosis. miR155 inhibited the cell proliferation, cell migration and cell invasion, arrested the cell cycle process at G1 phase and enhanced the cell apoptosis. The luciferases reporter assay showed that miR155 bound to the 3'-UTR of CTHRC1 and regulated the expression of CTHRC1, further influenced the functions of CTHRC1 in human melanoma development. The study contributed to unearthing a novel potential predictor and therapeutic targets of human melanoma. CTHRC1 was a potential independent predictor and therapeutic target for human melanoma.
Collapse
Affiliation(s)
- Yuan Li
- Shandong UniversityJinan, Shandong, China
- Department of Dermatology, Binzhou Medical University HospitalBinzhou, Shandong, China
| | - Yujie Zhang
- Department of Dermatology, Binzhou Medical University HospitalBinzhou, Shandong, China
| | - Chong Ma
- Department Colorectal and General Surgery, Binzhou Medical University HospitalBinzhou, Shandong, China
| | - Shouzhong Wang
- Department of Dermatology, Linyi People’s HospitalLinyi, Shandong, China
| | - Na Li
- Department of Dermatology, Binzhou Medical University HospitalBinzhou, Shandong, China
| | - Jing Wang
- Department of Dermatology, Binzhou Medical University HospitalBinzhou, Shandong, China
| | - Guanghui Ma
- Department of Dermatology, Binzhou Medical University HospitalBinzhou, Shandong, China
| | - Li Zhang
- Department of Dermatology, Shandong Provincial Hospital Affiliated to Shandong UniversityJinan, Shandong, China
| |
Collapse
|
29
|
Liu X, Chang X, Liu R, Yu X, Chen L, Aihara K. Quantifying critical states of complex diseases using single-sample dynamic network biomarkers. PLoS Comput Biol 2017; 13:e1005633. [PMID: 28678795 PMCID: PMC5517040 DOI: 10.1371/journal.pcbi.1005633] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 07/19/2017] [Accepted: 06/19/2017] [Indexed: 02/04/2023] Open
Abstract
Dynamic network biomarkers (DNB) can identify the critical state or tipping point of a disease, thereby predicting rather than diagnosing the disease. However, it is difficult to apply the DNB theory to clinical practice because evaluating DNB at the critical state required the data of multiple samples on each individual, which are generally not available, and thus limit the applicability of DNB. In this study, we developed a novel method, i.e., single-sample DNB (sDNB), to detect early-warning signals or critical states of diseases in individual patients with only a single sample for each patient, thus opening a new way to predict diseases in a personalized way. In contrast to the information of differential expressions used in traditional biomarkers to “diagnose disease”, sDNB is based on the information of differential associations, thereby having the ability to “predict disease” or “diagnose near-future disease”. Applying this method to datasets for influenza virus infection and cancer metastasis led to accurate identification of the critical states or correct prediction of the immediate diseases based on individual samples. We successfully identified the critical states or tipping points just before the appearance of disease symptoms for influenza virus infection and the onset of distant metastasis for individual patients with cancer, thereby demonstrating the effectiveness and efficiency of our method for quantifying critical states at the single-sample level. The concept of dynamic network biomarkers (DNB) was proposed for detecting the critical state or tipping point of a complex disease (a pre-disease state immediately preceding the disease state), and has been applied to study the mechanism of cell fate decision and immune checkpoint blockade. But DNB cannot be used to identify the critical state or tipping point for a single patient because evaluating DNB for critical state required the data of multiple samples. The proposed method can identify the critical state of a complex disease for a single patient by implementing the concept of DNB. This method not only can be applied to detect the critical state or tipping point of a single sample, but also can be used to study the mechanism of complex disease at a single sample level. The ability of accurately and efficiently identifying the critical state for a single sample can benefit the development of personalized medicine.
Collapse
Affiliation(s)
- Xiaoping Liu
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- College of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu, Anhui Province, China
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Mathematics and Statistics, Shandong University at Weihai, Weihai, China
| | - Xiao Chang
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- College of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu, Anhui Province, China
| | - Rui Liu
- School of Mathematics, South China University of Technology, Guangzhou, China
| | - Xiangtian Yu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Luonan Chen
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- * E-mail: (LC); (KA)
| | - Kazuyuki Aihara
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- * E-mail: (LC); (KA)
| |
Collapse
|
30
|
Zhang Q, Wang Y, Liang J, Tian Y, Zhang Y, Tao K. Bioinformatics analysis to identify the critical genes, microRNAs and long noncoding RNAs in melanoma. Medicine (Baltimore) 2017; 96:e7497. [PMID: 28723760 PMCID: PMC5521900 DOI: 10.1097/md.0000000000007497] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Melanoma, which is usually induced by ultraviolet light exposure and the following DNA damage, is the most dangerous skin cancer. The purpose of the present study was to screen key molecules involved in melanoma.Microarray data of E-MTAB-1862 were downloaded from the ArrayExpress database, which included 21 primary melanoma samples and 11 benign nevus samples. In addition, the RNASeq version 2 and microRNA (miRNA) sequencing data of cutaneous melanoma were downloaded from The Cancer Genome Atlas database. After identifying the differentially expressed genes (DEGs) using Limma package, enrichment analysis and protein-protein interaction (PPI) network analysis were performed separately for them using DAVID software and Cytoscape software. In addition, survival analysis and regulatory network analysis were further performed by log-rank test and Cytoscape software, respectively. Moreover, real-time reverse transcription polymerase chain reaction (RT-PCR) was performed to further verify the expression patterns of several selected DEGs.A total of 382 DEGs were identified in primary melanoma samples, including 206 upregulated genes and 176 downregulated genes. Functional enrichment analysis showed that COL17A1 was enriched in epidermis development. In the PPI network, CXCL8 (degree = 29) and STAT1 (degree = 28) had higher degrees and could interact with each other. Survival analysis showed that 21 DEGs, 55 long noncoding RNAs (lncRNAs) and 32 miRNAs were found to be associated with prognosis. Furthermore, several regulatory relationships were found in the lncRNA-gene regulatory network (such as RP11-361L15.4 targeting COL17A1) and the miRNA-gene regulatory network (such as hsa-miR-375 targeting CCL27 and hsa-miR-375 targeting insulin-like growth factor 1 receptor [IGF1R]). Real-time RT-PCR results showed that the overall direction of differential expression was consistent except COL17A1.CXCL8 interacted with STAT1, CCL27, and IGF1R targeted by hsa-miR-375, and COL17A1 targeted by RP11-361L15.4 might function in the development and progression of melanoma, which should be verified by more detailed experiments.
Collapse
|
31
|
Wang C, Gu W, Sun B, Zhang Y, Ji Y, Xu X, Wen Y. CTHRC1 promotes osteogenic differentiation of periodontal ligament stem cells by regulating TAZ. J Mol Histol 2017. [PMID: 28647773 DOI: 10.1007/s10735-017-9729-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Collagen triple helix repeat containing 1 (CTHRC1) is associated with bone metabolism. Alveolar bone has an ability to rapidly remodel itself to adapt its biomechanical environment and function. However, whether CTHRC1 is expressed in alveolar bone tissue and the role of CTHRC1 in alveolar bone remodeling remain unclear. We used orthodontic tooth movement (OTM) rat model to study the effects of CHTRC1 in alveolar bone remodeling in vivo. We found that CTHRC1 was expressed in normal physiological condition of osteocytes, bone matrix, and periodontal ligament cells in rat. During the OTM, the expression of CTHRC1, Runx2 and TAZ were increased. We further studied the effects of CTHRC1 on osteogenic differentiation of human periodontal ligament stem cells in vitro. CTHRC1 can positively regulate the expression of TAZ and osteogenic differentiation markers like Col1, ALP, Runx2 and OCN. Overexpression of CHTRC1 increased osteogenic differentiation of PDLSCs, which could be abolished by TAZ siRNA. Our results suggest that CTHRC1 plays an important role in alveolar bone remodeling and osteogenic differentiation of PDLSCs.
Collapse
Affiliation(s)
- Chengze Wang
- School of Stomatology, Shandong University, No. 44-1, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, People's Republic of China
| | - Weiting Gu
- Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Baiyu Sun
- School of Stomatology, Shandong University, No. 44-1, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, People's Republic of China
| | - Yunpeng Zhang
- School of Stomatology, Shandong University, No. 44-1, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, People's Republic of China
| | - Yawen Ji
- School of Stomatology, Shandong University, No. 44-1, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, People's Republic of China
| | - Xin Xu
- School of Stomatology, Shandong University, No. 44-1, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China. .,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, People's Republic of China.
| | - Yong Wen
- School of Stomatology, Shandong University, No. 44-1, Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China. .,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, People's Republic of China.
| |
Collapse
|
32
|
E6/E7-P53-POU2F1-CTHRC1 axis promotes cervical cancer metastasis and activates Wnt/PCP pathway. Sci Rep 2017; 7:44744. [PMID: 28303973 PMCID: PMC5356195 DOI: 10.1038/srep44744] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 02/13/2017] [Indexed: 12/25/2022] Open
Abstract
Cervical cancer is an infectious cancer and the most common gynecologic cancer worldwide. E6/E7, the early genes of the high-risk mucosal human papillomavirus type, play key roles in the carcinogenic process of cervical cancer. However, little was known about its roles in modulating tumor microenvironment, particular extracellular matrix (ECM). In this study, we found that E6/E7 could regulate multiple ECM proteins, especially collagen triple helix repeat containing 1 (CTHRC1). CTHRC1 is highly expressed in cervical cancer tissue and serum and closely correlated with clinicopathological parameters. CTHRC1 promotes cervical cancer cell migration and invasion in vitro and metastasis in vivo. E6/E7 regulates the expression of CTHRC1 in cervical cancer by E6/E7-p53-POU2F1 (POU class 2 homeobox 1) axis. Futhermore, CTHRC1 activates Wnt/PCP signaling pathway. Take together, E6/E7-p53-POU2F1-CTHRC1 axis promotes cervical cancer cell invasion and metastasis and may act as a potential therapeutic target for interventions against cervical cancer invasion and metastasis.
Collapse
|
33
|
Jumper N, Hodgkinson T, Paus R, Bayat A. Site-specific gene expression profiling as a novel strategy for unravelling keloid disease pathobiology. PLoS One 2017; 12:e0172955. [PMID: 28257480 PMCID: PMC5336271 DOI: 10.1371/journal.pone.0172955] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022] Open
Abstract
Keloid disease (KD) is a fibroproliferative cutaneous tumour characterised by heterogeneity, excess collagen deposition and aggressive local invasion. Lack of a validated animal model and resistance to a multitude of current therapies has resulted in unsatisfactory clinical outcomes of KD management. In order to address KD from a new perspective, we applied for the first time a site-specific in situ microdissection and gene expression profiling approach, through combined laser capture microdissection and transcriptomic array. The aim here was to analyse the utility of this approach compared with established methods of investigation, including whole tissue biopsy and monolayer cell culture techniques. This study was designed to approach KD from a hypothesis-free and compartment-specific angle, using state-of-the-art microdissection and gene expression profiling technology. We sought to characterise expression differences between specific keloid lesional sites and elucidate potential contributions of significantly dysregulated genes to mechanisms underlying keloid pathobiology, thus informing future explorative research into KD. Here, we highlight the advantages of our in situ microdissection strategy in generating expression data with improved sensitivity and accuracy over traditional methods. This methodological approach supports an active role for the epidermis in the pathogenesis of KD through identification of genes and upstream regulators implicated in epithelial-mesenchymal transition, inflammation and immune modulation. We describe dermal expression patterns crucial to collagen deposition that are associated with TGFβ-mediated signalling, which have not previously been examined in KD. Additionally, this study supports the previously proposed presence of a cancer-like stem cell population in KD and explores the possible contribution of gene dysregulation to the resistance of KD to conventional therapy. Through this innovative in situ microdissection gene profiling approach, we provide better-defined gene signatures of distinct KD regions, thereby addressing KD heterogeneity, facilitating differential diagnosis with other cutaneous fibroses via transcriptional fingerprinting, and highlighting key areas for future KD research.
Collapse
Affiliation(s)
- N. Jumper
- Plastic and Reconstructive Surgery Research, University of Manchester, Oxford Rd, Manchester, United Kingdom
| | - T. Hodgkinson
- Plastic and Reconstructive Surgery Research, University of Manchester, Oxford Rd, Manchester, United Kingdom
- Centre for Tissue Injury and Repair, University of Manchester, and MAHSC, Manchester, United Kingdom
| | - R. Paus
- Centre for Dermatology Research, University of Manchester, and MAHSC, Manchester, United Kingdom
| | - A. Bayat
- Plastic and Reconstructive Surgery Research, University of Manchester, Oxford Rd, Manchester, United Kingdom
- Centre for Dermatology Research, University of Manchester, and MAHSC, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
34
|
Jiang N, Cui Y, Liu J, Zhu X, Wu H, Yang Z, Ke Z. Multidimensional Roles of Collagen Triple Helix Repeat Containing 1 (CTHRC1) in Malignant Cancers. J Cancer 2016; 7:2213-2220. [PMID: 27994657 PMCID: PMC5166530 DOI: 10.7150/jca.16539] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/04/2016] [Indexed: 12/12/2022] Open
Abstract
Tumor is one of the principal diseases that seriously threaten human health. Insight into sensitive cancer markers may open a new avenue for the early diagnosis and treatment of this disease. CTHRC1 has been identified as a cancer-related gene. It is a secretory glycoprotein that possesses multidimensional roles associated with wound repair, bone remodeling, hepatocytes fibrosis, adipose tissue formation, and so on. Our previous studies and numerous reports from other researchers have revealed that the ascended expression of CTHRC1 tends to go hand in hand with tumorigenesis, proliferation, invasion and metastasis in various human malignancies through a series of molecular mechanisms and signaling pathways. However, the detailed pathogenic mechanisms of CTHRC1 overexpression in human malignant cancers are not yet clear. Here, we shall focus our description on the functions, expression profile in several representative malignant tumors and a number of molecular mechanisms and signaling pathways involved with CTHRC1. This introductory discussion of CTHRC1 will serve as a reference for further research in understanding this intriguing cancer-related protein.
Collapse
Affiliation(s)
- Neng Jiang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - YongMei Cui
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - JunXiu Liu
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - XiaoLin Zhu
- Department of Otolaryngology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Province Guangdong, P.R. China
| | - Hui Wu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - Zheng Yang
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| | - ZunFu Ke
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Province Guangdong, P.R. China
| |
Collapse
|