1
|
Erin N, Tavşan E, Haksever S, Yerlikaya A, Riganti C. Targeting BMP-1 enhances anti-tumoral effects of doxorubicin in metastatic mammary cancer: common and distinct features of TGF-β inhibition. Breast Cancer Res Treat 2025; 210:563-574. [PMID: 39792296 PMCID: PMC11953206 DOI: 10.1007/s10549-024-07592-4] [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: 10/20/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE Mammary carcinoma is comprised heterogeneous groups of cells with different metastatic potential. 4T1 mammary carcinoma cells metastasized to heart (4THM), liver (4TLM) and brain (4TBM) and demonstrate cancer-stem cell phenotype. Using these cancer cells we found thatTGF-β is the top upstream regulator of metastatic process. In addition, secretion of bone morphogenetic protein 1 (BMP-1), which is crucial for the proteolytic release of TGF-β, was markedly high in metastatic mammary cancer cells compared to non-metastatic cells. Although TGF-β inhibitors are in clinical trials, systemic inhibition of TGF-β may produce heavy side effects. We here hypothesize that inhibition of BMP-1 proteolytic activity inhibits TGF-β activity and induces anti-tumoral effects. METHOD AND RESULTS Effects of specific BMP-1 inhibitor on liver and brain metastatic murine mammary cancer cells (4TLM and 4TBM), as well as on human mammary cancer MDA-MB-231 and MCF-7 cells, were examined and compared with the results of TGF-β inhibition. Inhibition of BMP-1 activity markedly suppressed proliferation of cancer cells and enhanced anti-tumoral effects of doxorubicin. Inhibition of BMP-1 activity but not of TGF-β activity decreased colony and spheroid formation. Differential effects of BMP-1 and TGF-β inhibitors on TGF-β secretion was also observed. CONCLUSIONS These results demonstrated for the first time that the inhibition of BMP-1 activity has therapeutic potential for treatment of metastatic mammary cancer and enhances the anti-tumoral effects of doxorubicin.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Medical Pharmacology, Faculty of Medicine, Akdeniz University, Antalya, Turkey.
| | - Esra Tavşan
- Department of Medical Pharmacology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Seren Haksever
- Department of Medical Pharmacology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Azmi Yerlikaya
- Department of Medical Biology, Faculty of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Nizza 44, 10126, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", Via Nizza 44, 10126, Turin, Italy
- Interdepartmental Center "G.Scansetti" for the Study of Asbestos and Other Toxic Particulates, University of Torino, 10126, Turin, Italy
| |
Collapse
|
2
|
Napoli M, Bauer J, Bonod C, Vadon-Le Goff S, Moali C. PCPE-2 (procollagen C-proteinase enhancer-2): The non-identical twin of PCPE-1. Matrix Biol 2024; 134:59-78. [PMID: 39251075 DOI: 10.1016/j.matbio.2024.09.001] [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: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
PCPE-2 was discovered at the beginning of this century, and was soon identified as a close homolog of PCPE-1 (procollagen C-proteinase enhancer 1). After the demonstration that it could also stimulate the proteolytic maturation of fibrillar procollagens by BMP-1/tolloid-like proteinases (BTPs), PCPE-2 did not attract much attention as it was thought to fulfill the same functions as PCPE-1 which was already well-described. However, the tissue distribution of PCPE-2 shows both common points and significant differences with PCPE-1, suggesting that their activities are not fully overlapping. Also, the recently established connections between PCPE-2 (gene name PCOLCE2) and several important diseases such as atherosclerosis, inflammatory diseases and cancer have highlighted the need for a thorough reappraisal of the in vivo roles of this regulatory protein. In this context, the recent finding that, while retaining the ability to bind fibrillar procollagens and to activate their C-terminal maturation, PCPE-2 can also bind BTPs and inhibit their activity has substantially extended its potential functions. In this review, we describe the current knowledge about PCPE-2 with a focus on collagen fibrillogenesis, lipid metabolism and inflammation, and discuss how we could further advance our understanding of PCPE-2-dependent biological processes.
Collapse
Affiliation(s)
- Manon Napoli
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Julien Bauer
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Christelle Bonod
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Sandrine Vadon-Le Goff
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Catherine Moali
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France.
| |
Collapse
|
3
|
Thomas MJ, Xu H, Wang A, Beg MA, Sorci-Thomas MG. PCPE2: Expression of multifunctional extracellular glycoprotein associated with diverse cellular functions. J Lipid Res 2024; 65:100664. [PMID: 39374805 PMCID: PMC11567036 DOI: 10.1016/j.jlr.2024.100664] [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: 03/31/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024] Open
Abstract
Procollagen C-endopeptidase enhancer 2, known as PCPE2 or PCOC2 (gene name, PCOLCE2) is a glycoprotein that resides in the extracellular matrix, and is similar in domain organization to PCPE1/PCPE, PCOC1 (PCOLCE1/PCOLCE). Due to the many similarities between the two related proteins, PCPE2 has been assumed to have biological functions similar to PCPE. PCPE is a well-established enhancer of procollagen processing activating the enzyme, BMP-1. However, reports show that PCPE2 has a strikingly different tissue expression profile compared to PCPE. With that in mind and given the paucity of published studies on PCPE2, this review examines the current literature citing PCPE2 and its association with specific cell types and signaling pathways. Additionally, this review will present a brief history of PCPE2's discovery, highlighting structural and functional similarities and differences compared to PCPE. Considering the widespread use of RNA sequencing techniques to examine associations between cell-specific gene expression and disease states, we will show that PCPE2 is repeatedly found as a differentially regulated gene (DEG) significantly associated with a number of cellular processes, well beyond the scope of procollagen fibril processing.
Collapse
Affiliation(s)
- Michael J Thomas
- Division of Endocrinology and Molecular Medicine, Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hao Xu
- Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Angela Wang
- Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mirza Ahmar Beg
- Division of Endocrinology and Molecular Medicine, Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mary G Sorci-Thomas
- Division of Endocrinology and Molecular Medicine, Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
4
|
Hong J, Jin HJ, Choi MR, Lim DWT, Park JE, Kim YS, Lim SB. Matrisomics: Beyond the extracellular matrix for unveiling tumor microenvironment. Biochim Biophys Acta Rev Cancer 2024; 1879:189178. [PMID: 39241895 DOI: 10.1016/j.bbcan.2024.189178] [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: 06/05/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
The matrisome, a group of proteins constituting or interacting with the extracellular matrix (ECM), has garnered attention as a potent regulator of cancer progression. An increasing number of studies have focused on cancer matrisome utilizing diverse -omics approaches. Here, we present diverse patterns of matrisomal populations within cancer tissues, exploring recent -omics studies spanning different '-omics' levels (epigenomics, genomics, transcriptomics, and proteomics), as well as newly developed sequencing techniques such as single-cell RNA sequencing and spatial transcriptomics. Some matrisome genes showed uniform patterns of upregulated or downregulated expression across various cancers, while others displayed different expression patterns according to the cancer types. This matrisomal dysregulation in cancer was further examined according to their originating cell type and spatial location in the tumor tissue. Experimental studies were also collected to demonstrate the identified roles of matrisome genes during cancer progression. Interestingly, many studies on cancer matrisome have suggested matrisome genes as effective biomarkers in cancer research. Although the specific mechanisms and clinical applications of cancer matrisome have not yet been fully elucidated, recent techniques and analyses on cancer matrisomics have emphasized their biological importance in cancer progression and their clinical implications in deciding the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Jiwon Hong
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Hyo Joon Jin
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Mi Ran Choi
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Darren Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre, Singapore 168583, Singapore
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - You-Sun Kim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
5
|
Kianmehr S, Vahabirad M, Seghatoleslam A, Sadeghi E, Kiani R, Ghasemi H. Prognostic Value of TGF-β Expression in Bladder Cancer: A Systematic Review and Meta-analysis. UROLOGY RESEARCH & PRACTICE 2024; 50:148-153. [PMID: 39495544 PMCID: PMC11562745 DOI: 10.5152/tud.2024.24024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/03/2024] [Indexed: 11/05/2024]
Abstract
Objective Transforming growth factor beta (TGF-β) is a member of the growth factor superfamily that clinical studies address its association with bladder cancer invasion, progression, and metastasis. The present systematic review and meta-analysis aimed to explore the prognostic significance of TGF-β expression in bladder cancer patients. Materials and Methods The major international databases, including PubMed, Web of Science, Embase, and Scopus, were searched for full-text literature citations. The hazard ratio (HR) with a 95% CI as the effect size was applied as the appropriate summarized statistic. We used a random-effects model using the DerSimonian and Laird method to estimate the pooled effect size. To assess the heterogeneity among trials, the I-square (I 2 ) statistic and Cochran's Q test were used. Forest and funnel plots were drawn to respectively demonstrate the findings and detect any existing publication bias. Results This meta-analysis included 3 studies that met the criteria and included 535 patients. The combined HR for the selected studies was 2.250 (95% CI=(1.411, 3.586), P< .001) and no significant heterogeneity was detected between trials (I 2=58.63, P=.089). Furthermore, no severe asymmetry was seen within the funnel plot, indicating a lack of potential publication bias. Conclusion Our findings suggest that TGF-β expression can remarkably predict a worse prognosis in patients with bladder cancer. The results of the present meta-analysis may be validated through further updated reviews and additional relevant investigations in future studies.
Collapse
Affiliation(s)
- Shima Kianmehr
- Department of Clinical Biochemistry, Hamadan University of Medical Sciences School of Medicine, Hamadan, Iran
| | - Mohammad Vahabirad
- Department of Clinical Biochemistry, Hamadan University of Medical Sciences School of Medicine, Hamadan, Iran
| | - Atefeh Seghatoleslam
- Department of Biochemistry, Shiraz University of Medical Sciences School of Medicine, Shiraz, Iran
| | - Erfan Sadeghi
- Department of Biostatistics, Shiraz University of Medical Sciences School of Medicine, Shiraz, Iran
| | - Roozbeh Kiani
- Department of Biochemistry, Shiraz University of Medical Sciences School of Medicine, Shiraz, Iran
| | - Hadi Ghasemi
- Department of Biochemistry, Shiraz University of Medical Sciences School of Medicine, Shiraz, Iran
| |
Collapse
|
6
|
Dastsooz H, Mohammadisoleimani E, Haghi-Aminjan H, Firoozi Z, Mansoori Y. Bioinformatic Approaches for the Identification of Novel Tumor Suppressor Genes and Cancer Pathways in Renal Clear Cell Carcinoma. IRANIAN JOURNAL OF BIOTECHNOLOGY 2024; 22:e3817. [PMID: 40225292 PMCID: PMC11993239 DOI: 10.30498/ijb.2024.421319.3817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 05/15/2024] [Indexed: 04/15/2025]
Abstract
Background Clear cell renal cell carcinoma (ccRCC, KIRC) is the most prevalent subtype of RCC, and even with different available therapies, the average progression-free survival is worse. Therefore, the identification of new molecular targets could be helpful for its therapeutic purposes. Materials and Methods We used the Cancer Genome Atlas to perform bioinformatic analyses for genes with possible tumor suppressor roles in KIRC. Objective This research aims to identify new prognostic biomarkers and potential therapeutic targets for this type of cancer. Results We identified 14 down-regulated genes in KIRC that had not previously been studied or poorly studied, with the majority of them impacted by increased promoter methylation. Eight genes showed shorter overall survival and worse prognosis, indicating their function as tumor suppressors, and six genes revealed good prognosis. From the 8 genes, C7ORF41 and CTXN3 revealed only downregulation in most cancers, proposing them as highly potential tumor suppressors. Among these 8 genes, the function of CTXN3 in cancers is unknown. Moreover, we identified the CWH43 gene as the major signature of KIRC. In addition, we found different genes as signatures of KIRC tumor stages and grades. Conclusions Our results may shed light on identifying KIRC pathogenesis and developing effective therapeutic targets for renal cancers, mainly KIRC.
Collapse
Affiliation(s)
- Hassan Dastsooz
- Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
- Candiolo, C/o IRCCS, IIGM-Italian Institute for Genomic Medicine, Turin, Italy
- Candiolo Cancer (IT), FPO-IRCCS, Candiolo Cancer Institute, Turin, Italy
| | | | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Zahra Firoozi
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| | - Yaser Mansoori
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
7
|
Craig O, Lee S, Pilcher C, Saoud R, Abdirahman S, Salazar C, Williams N, Ascher D, Vary R, Luu J, Cowley K, Ramm S, Li MX, Thio N, Li J, Semple T, Simpson K, Gorringe K, Holien J. A new method for network bioinformatics identifies novel drug targets for mucinous ovarian carcinoma. NAR Genom Bioinform 2024; 6:lqae096. [PMID: 39184376 PMCID: PMC11344246 DOI: 10.1093/nargab/lqae096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/11/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Mucinous ovarian carcinoma (MOC) is a subtype of ovarian cancer that is distinct from all other ovarian cancer subtypes and currently has no targeted therapies. To identify novel therapeutic targets, we developed and applied a new method of differential network analysis comparing MOC to benign mucinous tumours (in the absence of a known normal tissue of origin). This method mapped the protein-protein network in MOC and then utilised structural bioinformatics to prioritise the proteins identified as upregulated in the MOC network for their likelihood of being successfully drugged. Using this protein-protein interaction modelling, we identified the strongest 5 candidates, CDK1, CDC20, PRC1, CCNA2 and TRIP13, as structurally tractable to therapeutic targeting by small molecules. siRNA knockdown of these candidates performed in MOC and control normal fibroblast cell lines identified CDK1, CCNA2, PRC1 and CDC20, as potential drug targets in MOC. Three targets (TRIP13, CDC20, CDK1) were validated using known small molecule inhibitors. Our findings demonstrate the utility of our pipeline for identifying new targets and highlight potential new therapeutic options for MOC patients.
Collapse
Affiliation(s)
- Olivia Craig
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Samuel Lee
- The Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Carlton, VIC 3010, Australia
- St Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Courtney Pilcher
- School of Science, STEM College, RMIT University, Bundoora, VIC 3082, Australia
| | - Rita Saoud
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Suad Abdirahman
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Carolina Salazar
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Nathan Williams
- St Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- School of Science, STEM College, RMIT University, Bundoora, VIC 3082, Australia
| | - David B Ascher
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4067, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Robert Vary
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Jennii Luu
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Karla J Cowley
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Susanne Ramm
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Mark Xiang Li
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Niko Thio
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
| | - Jason Li
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
| | - Tim Semple
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
| | - Kaylene J Simpson
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC 3010, Australia
- The Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia
| | - Kylie L Gorringe
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3052, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Jessica K Holien
- The Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Carlton, VIC 3010, Australia
- St Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- School of Science, STEM College, RMIT University, Bundoora, VIC 3082, Australia
| |
Collapse
|
8
|
Chong ZX, Ho WY, Yeap SK. Tumour-regulatory role of long non-coding RNA HOXA-AS3. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 189:13-25. [PMID: 38593905 DOI: 10.1016/j.pbiomolbio.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/25/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
Dysregulation of long non-coding RNA (lncRNA) HOXA-AS3 has been shown to contribute to the development of multiple cancer types. Several studies have presented the tumour-modulatory role or prognostic significance of this lncRNA in various kinds of cancer. Overall, HOXA-AS3 can act as a competing endogenous RNA (ceRNA) that inhibits the activity of seven microRNAs (miRNAs), including miR-29a-3p, miR-29 b-3p, miR-29c, miR-218-5p, miR-455-5p, miR-1286, and miR-4319. This relieves the downstream messenger RNA (mRNA) targets of these miRNAs from miRNA-mediated translational repression, allowing them to exert their effect in regulating cellular activities. Examples of the pathways regulated by lncRNA HOXA-AS3 and its associated downstream targets include the WNT/β-catenin and epithelial-to-mesenchymal transition (EMT) activities. Besides, HOXA-AS3 can interact with other cellular proteins like homeobox HOXA3 and HOXA6, influencing the oncogenic signaling pathways associated with these proteins. Generally, HOXA-AS3 is overexpressed in most of the discussed human cancers, making this lncRNA a potential candidate to diagnose cancer or predict the clinical outcomes of cancer patients. Hence, targeting HOXA-AS3 could be a new therapeutic approach to slowing cancer progression or as a potential biomarker and therapeutic target. A drawback of using lncRNA HOXA-AS3 as a biomarker or therapeutic target is that most of the studies that have reported the tumour-regulatory roles of lncRNA HOXA-AS3 are single observational, in vitro, or in vivo studies. More in-depth mechanistic and large-scale clinical trials must be conducted to confirm the tumour-modulatory roles of lncRNA HOXA-AS3 further. Besides, no lncRNA HOXA-AS3 inhibitor has been tested preclinically and clinically, and designing such an inhibitor is crucial as it may potentially slow cancer progression.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia.
| |
Collapse
|
9
|
Chen C, Zhang Y, Lin Y, Shen C, Zhang Z, Wu Z, Qie Y, Zhao G, Hu H. The prognostic significance and immune characteristics of bone morphogenetic proteins (BMPs) family: A pan-cancer multi-omics analysis. Technol Health Care 2024; 32:4123-4175. [PMID: 39031404 PMCID: PMC11613112 DOI: 10.3233/thc-232004] [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: 12/21/2023] [Accepted: 05/30/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Bone morphogenetic proteins (BMPs) are a group of cancer-related proteins vital for development and progression of certain cancer types. Nevertheless, function of BMP family in pan-cancer was not detailedly researched. OBJECTIVE Investigating expression pattern and prognostic value of the BMPs family (BMP1-8A and BMP8B) expression across multiple cancer types. METHODS Our research integrated multi-omics data for exploring potential associations between BMPs expression and prognosis, clinicopathological characteristics, copy number or somatic mutations, immune characteristics, tumor microenvironment (TME), tumor mutation burden (TMB), microsatellite instability (MSI), immune checkpoint genes and drug sensitivity in The Cancer Genome Atlas (TCGA) tumors. Furthermore, association of BMPs expression and immunotherapy effectiveness was investigated in some confirmatory cohorts (GSE111636, GSE78220, GSE67501, GSE176307, IMvigor210 and mRNA sequencing data from currently undergoing TRUCE01 clinical research included), and biological function and potential signaling pathways of BMPs in bladder cancer (BCa) was explored via Gene Set Enrichment Analysis (GSEA). Eventually, immune infiltration analysis was done via BMPs expression, copy number or somatic mutations in BCa, as well as validation of the expression levels by reverse transcription-quantitative PCR and western blot, and in vitro functional experiments of BMP8A. RESULTS Discoveries displayed BMPs expression was related to prognosis, clinicopathological characteristics, mutations, TME, TMB, MSI and immune checkpoint genes of TCGA tumors. Anticancer drug sensitivity analysis displayed BMPs were associated with various drug sensitivities. What's more, it was discovered that expression level of certain BMP family members related to objective response to immunotherapy. By GSEA, we discovered multiple immune-associated functions and pathways were enriched. Immune infiltration analysis on BCa also displayed significant associations among BMPs copy number variations, mutation status and infiltration level of diverse immune cells. Furthermore, differential expression validation and in vitro phenotypic experiment indicated that BMP8A significantly promoted BCa cell proliferation, migration and invasion. CONCLUSIONS Current results confirmed significance of both BMPs expression and genomic alteration in the prognosis and treatment of diverse cancer types, and suggested that BMPs may be vital for BCa and can possibly be utilized as biomarkers for immunotherapy.
Collapse
Affiliation(s)
- Changsheng Chen
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Department of Urology, Tianjin Haihe Hospital, Tianjin, China
| | - Yu Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Department of Urology, The Eco-City Hospital of Tianjin Fifth Central Hospital, Tianjin, China
| | - Yuda Lin
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chong Shen
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhe Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhouliang Wu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yunkai Qie
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Gangjian Zhao
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Hailong Hu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
10
|
Vadon-Le Goff S, Tessier A, Napoli M, Dieryckx C, Bauer J, Dussoyer M, Lagoutte P, Peyronnel C, Essayan L, Kleiser S, Tueni N, Bettler E, Mariano N, Errazuriz-Cerda E, Fruchart Gaillard C, Ruggiero F, Becker-Pauly C, Allain JM, Bruckner-Tuderman L, Nyström A, Moali C. Identification of PCPE-2 as the endogenous specific inhibitor of human BMP-1/tolloid-like proteinases. Nat Commun 2023; 14:8020. [PMID: 38049428 PMCID: PMC10696041 DOI: 10.1038/s41467-023-43401-0] [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: 08/24/2022] [Accepted: 11/08/2023] [Indexed: 12/06/2023] Open
Abstract
BMP-1/tolloid-like proteinases (BTPs) are major players in tissue morphogenesis, growth and repair. They act by promoting the deposition of structural extracellular matrix proteins and by controlling the activity of matricellular proteins and TGF-β superfamily growth factors. They have also been implicated in several pathological conditions such as fibrosis, cancer, metabolic disorders and bone diseases. Despite this broad range of pathophysiological functions, the putative existence of a specific endogenous inhibitor capable of controlling their activities could never be confirmed. Here, we show that procollagen C-proteinase enhancer-2 (PCPE-2), a protein previously reported to bind fibrillar collagens and to promote their BTP-dependent maturation, is primarily a potent and specific inhibitor of BTPs which can counteract their proteolytic activities through direct binding. PCPE-2 therefore differs from the cognate PCPE-1 protein and extends the possibilities to fine-tune BTP activities, both in physiological conditions and in therapeutic settings.
Collapse
Affiliation(s)
- Sandrine Vadon-Le Goff
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Agnès Tessier
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, 79104, Freiburg, Germany
- University of Freiburg, Faculty of Biology, 79104, Freiburg, Germany
| | - Manon Napoli
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Cindy Dieryckx
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Julien Bauer
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Mélissa Dussoyer
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Priscillia Lagoutte
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Célian Peyronnel
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Lucie Essayan
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Svenja Kleiser
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, 79104, Freiburg, Germany
- University of Freiburg, Faculty of Biology, 79104, Freiburg, Germany
| | - Nicole Tueni
- Laboratoire de Mécanique des Solides, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91120, Palaiseau, France
- INRIA, 91120, Palaiseau, France
- Institute of Applied Mechanics, Department of Mechanical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Emmanuel Bettler
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Natacha Mariano
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France
| | - Elisabeth Errazuriz-Cerda
- University of Lyon, Centre d'Imagerie Quantitative Lyon-Est (CIQLE), SFR Santé-Lyon Est, 69373, Lyon, France
| | - Carole Fruchart Gaillard
- Université Paris-Saclay, CEA, INRAE, Médicaments et Technologies pour la Santé (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Florence Ruggiero
- ENS Lyon, CNRS UMR 5242, Institut de Génomique Fonctionnelle de Lyon (IGFL), 69007, Lyon, France
| | - Christoph Becker-Pauly
- University of Kiel, Biochemical Institute, Unit for Degradomics of the Protease Web, Kiel, Germany
| | - Jean-Marc Allain
- Laboratoire de Mécanique des Solides, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, 91120, Palaiseau, France
- INRIA, 91120, Palaiseau, France
| | - Leena Bruckner-Tuderman
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, 79104, Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, 79104, Freiburg, Germany
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany
| | - Catherine Moali
- University of Lyon, CNRS UMR5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367, Lyon, France.
| |
Collapse
|
11
|
Gong M, Feng S, Zhou D, Luo J, Lin T, Qiu S, Yuan R, Dong W. Upregulation of BMP1 through ncRNAs correlates with adverse outcomes and immune infiltration in clear cell renal cell carcinoma. Eur J Med Res 2023; 28:440. [PMID: 37848987 PMCID: PMC10580559 DOI: 10.1186/s40001-023-01422-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/01/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) accounts for approximately 2-3% of all adult malignancies. Clear cell renal cell carcinoma (ccRCC), which comprises 70-80% of all RCC cases, is the most common histological subtype. METHODS ccRCC transcriptome data and clinical information were downloaded from the TCGA database. We used the TCGA and GEPIA databases to analyze relative expression of BMP1 in various types of human cancer. GEPIA was used to perform survival analysis for BMP1 in various cancer types. Upstream binding miRNAs of BMP1 were obtained through several important target gene prediction tools. StarBase was used to predict candidate miRNAs that may bind to BMP1 and candidate lncRNAs that may bind to hsa-miR-532-3p. We analyzed the association between expression of BMP1 and immune cell infiltration levels in ccRCC using the TIMER website. The relationship between BMP1 expression levels and immune checkpoint expression levels was also investigated. RESULTS BMP1 was upregulated in GBM, HNSC, KIRC, KIRP and STAD and downregulated in KICH and PRAD. Combined with OS and DFS, BMP1 can be used as a biomarker for poor prognosis among patients with KIRC. Through expression analysis, survival analysis and correlation analysis, LINC00685, SLC16A1-AS1, PVT1, VPS9D1-AS1, SNHG15 and the CCDC18-AS1/hsa-miR-532-3p/BMP1 axis were established as the most potential upstream ncRNA-related pathways of BMP1 in ccRCC. Furthermore, we found that BMP1 levels correlated significantly positively with tumor immune cell infiltration, biomarkers of immune cells, and immune checkpoint expression. CONCLUSION Our results demonstrate that ncRNA-mediated high expression of BMP1 is associated with poor prognosis and tumor immune infiltration in ccRCC.
Collapse
Affiliation(s)
- Mancheng Gong
- Department of Urology, The People's Hospital of Zhongshan, Zhongshan, 528403, Guangdong, China
| | - Shengxing Feng
- Department of Urology, The People's Hospital of Zhongshan, Zhongshan, 528403, Guangdong, China
| | - Dongsheng Zhou
- Department of Urology, The People's Hospital of Zhongshan, Zhongshan, 528403, Guangdong, China
| | - Jinquan Luo
- Department of Urology, The People's Hospital of Zhongshan, Zhongshan, 528403, Guangdong, China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Guangzhou, 510080, Guangdong, China
| | - Shaopeng Qiu
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Runqiang Yuan
- Department of Urology, The People's Hospital of Zhongshan, Zhongshan, 528403, Guangdong, China.
| | - Wenjing Dong
- Department of Oncology, The People's Hospital of Zhongshan, No. 2 Sunwen East Road, Zhongshan, 528403, Guangdong, China.
| |
Collapse
|
12
|
Kumari S, Kumar P. Identification and characterization of putative biomarkers and therapeutic axis in Glioblastoma multiforme microenvironment. Front Cell Dev Biol 2023; 11:1236271. [PMID: 37538397 PMCID: PMC10395518 DOI: 10.3389/fcell.2023.1236271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/23/2023] [Indexed: 08/05/2023] Open
Abstract
Non-cellular secretory components, including chemokines, cytokines, and growth factors in the tumor microenvironment, are often dysregulated, impacting tumorigenesis in Glioblastoma multiforme (GBM) microenvironment, where the prognostic significance of the current treatment remains unsatisfactory. Recent studies have demonstrated the potential of post-translational modifications (PTM) and their respective enzymes, such as acetylation and ubiquitination in GBM etiology through modulating signaling events. However, the relationship between non-cellular secretory components and post-translational modifications will create a research void in GBM therapeutics. Therefore, we aim to bridge the gap between non-cellular secretory components and PTM modifications through machine learning and computational biology approaches. Herein, we highlighted the importance of BMP1, CTSB, LOX, LOXL1, PLOD1, MMP9, SERPINE1, and SERPING1 in GBM etiology. Further, we demonstrated the positive relationship between the E2 conjugating enzymes (Ube2E1, Ube2H, Ube2J2, Ube2C, Ube2J2, and Ube2S), E3 ligases (VHL and GNB2L1) and substrate (HIF1A). Additionally, we reported the novel HAT1-induced acetylation sites of Ube2S (K211) and Ube2H (K8, K52). Structural and functional characterization of Ube2S (8) and Ube2H (1) have identified their association with protein kinases. Lastly, our results found a putative therapeutic axis HAT1-Ube2S(K211)-GNB2L1-HIF1A and potential predictive biomarkers (CTSB, HAT1, Ube2H, VHL, and GNB2L1) that play a critical role in GBM pathogenesis.
Collapse
|
13
|
Jena SK, Das S, Chakraborty S, Ain R. Molecular determinants of epithelial mesenchymal transition in mouse placenta and trophoblast stem cell. Sci Rep 2023; 13:10978. [PMID: 37414855 PMCID: PMC10325982 DOI: 10.1038/s41598-023-37977-2] [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: 03/22/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023] Open
Abstract
Trophectoderm cells of the blastocyst are the precursor of the placenta that is comprised of trophoblast, endothelial and smooth muscle cells. Since trophoectoderm cells are epithelial in nature, epithelial mesenchymal transition (EMT) of trophoblast stem (TS) cells might play pivotal role in placental morphogenesis. However, the molecular regulation of EMT during placental development and trophoblast differentiation still remained elusive. In this report, we sought to identify the molecular signature that regulates EMT during placental development and TS cell differentiation in mice. On E7.5 onwards the TS cells, located in the ectoplacental cone (EPC), rapidly divide and differentiate leading to formation of placenta proper. Using a real time PCR based array of functional EMT transcriptome with RNA from mouse implantation sites (IS) on E7.5 and E9.5, it was observed that there was an overall reduction of EMT gene expression in the IS as gestation progressed from E7.5 to E9.5 albeit the levels of EMT gene expression were substantial on both days. Further validation of array results using real time PCR and western blot analysis showed significant decrease in EMT-associated genes that included (a) transcription factors (Snai2, Zeb1, Stat3 and Foxc2), (b) extracellular matrix and cell adhesion related genes (Bmp1, Itga5, Vcan and Col3A1), (c) migration and motility- associated genes (Vim, Msn and FN1) and (d) differentiation and development related genes (Wnt5b, Jag1 and Cleaved Notch-1) on E9.5. To understand whether EMT is an ongoing process during placentation, the EMT-associated signatures genes, prevalent on E 7.5 and 9.5, were analysed on E12.5, E14.5 and E17.5 of mouse placenta. Interestingly, expression of these EMT-signature proteins were significantly higher at E12.5 though substantial expressions was observed in placenta with progression of gestation from mid- to late. To evaluate whether TS cells have the potential to undergo EMT ex vivo, TS cells were subjected to EMT induction, which was confirmed using morphological analysis and marker gene expression. Induction of EMT in TS cells showed similar gene expression profile of placental EMT. These results have broad biological implications, as inadequate mesenchymal transition leading to improper trophoblast-vasculogenic mimicry leads to placental pathophysiology and pregnancy failure.
Collapse
Affiliation(s)
- Shipra Kanti Jena
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West Bengal, 700032, India
- Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, UP, 201002, India
| | - Shreya Das
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West Bengal, 700032, India
| | - Shreeta Chakraborty
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West Bengal, 700032, India
| | - Rupasri Ain
- Division of Cell Biology and Physiology, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Calcutta, West Bengal, 700032, India.
- Academy of Scientific and Innovative Research (AcSIR), Sector 19, Kamla Nehru Nagar, Ghaziabad, UP, 201002, India.
| |
Collapse
|
14
|
Yao J, Liang Z, Duan L, G Y, Liu J, An G. Construction of a novel immune response prediction signature to predict the efficacy of immune checkpoint inhibitors in clear cell renal cell carcinoma patients. Heliyon 2023; 9:e15925. [PMID: 37484396 PMCID: PMC10360603 DOI: 10.1016/j.heliyon.2023.e15925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 07/25/2023] Open
Abstract
Background Immune checkpoint inhibitor (ICI) treatment has enhanced survival outcomes in clear cell renal cell carcinoma (ccRCC) patients. Nevertheless, the effectiveness of immunotherapy in ccRCC patients is restricted and we intended to develop and characterize an immune response prediction signature (IRPS) to forecast the efficacy of immunotherapy. Methods RNA-seq expression profile and clinicopathologic characteristics of 539 kidney cancer and 72 patients with normal specimens, were downloaded from the Cancer Genome Atlas (TCGA) database, while the Gene Expression Omnibus (GEO) database was used as the validation set, which included 24 ccRCC samples. Utilization of the TCGA data and immune genes databases (ImmPort and the InnateDB), we explored through Weighted Gene Co-expression Network Analysis (WGCNA), along with Least Absolute Shrinkage and Selection Operator method (LASSO), and constructed an IRPS for kidney cancer patients. GSEA and CIBERSORT were performed to declare the molecular and immunologic mechanism underlying the predictive value of IRPS. The Human Protein Atlas (HPA) was deployed to verify the protein expressions of IRPS genes. Tumor immune dysfunction and exclusion (TIDE) score and immunophenoscore (IPS) were computed to determine the risk of immune escape and value the discrimination of IRPS. A ccRCC cohort with anti-PD-1 therapy was obtained as an external validation data set to verify the predictive value of IRPS. Results We constructed a 10 gene signature related to the prognosis and immune response of ccRCC patients. Considering the IRPS risk score, patients were split into high and low risk groups. Patients with high risk in the TCGA cohort tended towards advanced tumor stage and grade with poor prognosis (p < 0.001), which was validated in GEO database (p = 0.004). High-risk group tumors were related with lower PD-L1 expression, higher TMB, higher MSIsensor score, lower IPS, higher TIDE score, and enriched Treg cells, which might be the potential mechanism of immune dysfunction and exclusion. Patients in the IRPS low risk group had better PFS (HR:0.73; 95% CI: 0.54-1.0; P = 0.047). Conclusion A novel biomarker of IRPS was constructed to predict the benefit of immunotherapy, which might lead to more individualized prognoses and tailored therapy for kidney cancer patients.
Collapse
Affiliation(s)
- Jiannan Yao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ziwei Liang
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ling Duan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yang G
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jian Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| |
Collapse
|
15
|
Yin G, Tian T, Ji X, Zheng S, Zhu Z, Li Y, Zhang C. Integrated analysis to identify the prognostic and immunotherapeutic roles of coagulation-associated gene signature in clear cell renal cell carcinoma. Front Immunol 2023; 14:1107419. [PMID: 37006234 PMCID: PMC10063824 DOI: 10.3389/fimmu.2023.1107419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
The coagulation system is closely related to the physiological status and immune response of the body. Recent years, studies focusing on the association between coagulation system abnormalities and tumor progression have been widely reported. In clear cell renal cell carcinoma (ccRCC), poor prognosis often occurs in patients with venous tumor thrombosis and coagulation system abnormalities, and there is a lack of research in related fields. Significant differences in coagulation function were also demonstrated in our clinical sample of patients with high ccRCC stage or grade. Therefore, in this study, we analyzed the biological functions of coagulation-related genes (CRGs) in ccRCC patients using single-cell sequencing and TCGA data to establish the 5-CRGs based diagnostic signature and predictive signature for ccRCC. Univariate and multivariate Cox analyses suggested that prognostic signature could be an independent risk factor. Meanwhile, we applied CRGs for consistent clustering of ccRCC patients, and the two classes showed significant survival and genotype differences. The differences in individualized treatment between the two different subtypes were revealed by pathway enrichment analysis and immune cell infiltration analysis. In summary, we present the first systematic analysis of the significance of CRGs in the diagnosis, prognosis, and individualized treatment of ccRCC patients.
Collapse
Affiliation(s)
- Guicao Yin
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Tai Tian
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Xing Ji
- Department of Urology, The Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shengqi Zheng
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Zhenpeng Zhu
- Department of Urology, The Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Zhenpeng Zhu, ; Yifan Li, ; Cuijian Zhang,
| | - Yifan Li
- Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- *Correspondence: Zhenpeng Zhu, ; Yifan Li, ; Cuijian Zhang,
| | - Cuijian Zhang
- Department of Urology, The Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Zhenpeng Zhu, ; Yifan Li, ; Cuijian Zhang,
| |
Collapse
|
16
|
Identification of key biomarkers for STAD using filter feature selection approaches. Sci Rep 2022; 12:19854. [PMID: 36400805 PMCID: PMC9674689 DOI: 10.1038/s41598-022-21760-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/30/2022] [Indexed: 11/19/2022] Open
Abstract
Gastric cancer (GC) is the fifth most common cancer and the third leading cause of cancer death worldwide. Discovery of diagnostic biomarkers prompts the early detection of GC. In this study, we used limma method combined with joint mutual information (JMI), a machine learning algorithm, to identify a signature of 11 genes that performed well in distinguishing tumor and normal samples in a stomach adenocarcinoma cohort. Other two GC datasets were used to validate the classifying performances. Several of the candidate genes were correlated with GC tumor progression and survival. Overall, we highlight the application of feature selection approaches in the analysis of high-dimensional biological data, which will improve study accuracies and reduce workloads for the researchers when identifying potential tumor biomarkers.
Collapse
|
17
|
A SERPINE1-Based Immune Gene Signature Predicts Prognosis and Immunotherapy Response in Gastric Cancer. Pharmaceuticals (Basel) 2022; 15:ph15111401. [PMID: 36422531 PMCID: PMC9692477 DOI: 10.3390/ph15111401] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) therapy has been successfully utilized in the treatment of multiple tumors, but only a fraction of patients with gastric cancer (GC) could greatly benefit from it. A recent study has shown that the tumor microenvironment (TME) can greatly affect the effect of immunotherapy in GC. In this study, we established a novel immune risk signature (IRS) for prognosis and predicting response to ICIs in GC based on the TCGA-STAD dataset. Characterization of the TME was explored and further validated to reveal the underlying survival mechanisms and the potential therapeutic targets of GC. The GC patients were stratified into high- and low-risk groups based on the IRS. Patients in the high-risk group, associated with poorer outcomes, were characterized by significantly higher immune function. Further analysis showed higher T cell immune dysfunction and probability of potential immune escape. In vivo, we detected the expressions of SERPINE1 by the quantitative real-time polymerase chain reaction (qPCR)in tumor tissues and adjacent normal tissues. In vitro, knockdown of SERPINE1 significantly attenuated malignant biological behaviors of tumor cells in GC. Our signature can effectively predict the prognosis and response to immunotherapy in patients with GC.
Collapse
|
18
|
An Analysis of BMP1 Associated with m6A Modification and Immune Infiltration in Pancancer. DISEASE MARKERS 2022; 2022:7899961. [PMID: 36267461 PMCID: PMC9578879 DOI: 10.1155/2022/7899961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022]
Abstract
Background. This research explores the underlying link between diagnosis and therapy between bone morphogenetic protein 1 (BMP1) and various cancers. Methods. Three immunotherapeutic cohorts, by the composition of IMvigor210, GSE35640, and GSE78220 were obtained from previously published articles and the Gene Expression Omnibus database. The different expressions of BMP1 in various clinical parameters were conducted, and prognostic analysis was executed utilizing Cox proportional hazard regression and Gene Expression Profiling Interactive Analysis. Moreover, the correlation between BMP1 and tumor microenvironment was analyzed using ESTIMATE and CIBERSORT algorithms. Tumor mutational burden and microsatellite instability were also included. The correlation between m6A modification and the gene expression level was analyzed using Tumor IMmune Estimation Resource, the University of Alabama at Birmingham Cancer data analysis portal. Gene Set Cancer Analysis analyzed the correlation of BMP1 expression level with copy number variations and methylation. Furthermore, the correlation between BMP1 and therapeutic response after antineoplastic drug use was illustrated for further discussion. Results. BMP1 expression had significant differences in 14 cancers. It presented an intimate relationship with immune-relevant biomarkers. A variation analysis indicated that BMP1 had a significant association with immunotherapeutic response. The expression level of BMP1 was closely associated with insulin-like growth factor binding protein 3, an m6A modification relative gene. Except for a few cancer types, methylation negatively correlated with BMP1, and copy number variations positively correlated with BMP1. Notably, low BMP1 expression was connected with immunotherapeutic response in the cohorts, and its expression was related to increased sectional sensitivity of drugs. Conclusion. BMP1 may serve as a potential biomarker for prognostic prediction and immunologic infiltration in diversified cancers, providing a new thought approach for oncotherapy.
Collapse
|
19
|
Dong S, Zhang S, Zhao P, Lin G, Ma X, Xu J, Zhang H, Hu J, Zou C. A combined analysis of bulk and single-cell sequencing data reveals that depleted extracellular matrix and enhanced immune processes co-contribute to fluorouracil beneficial responses in gastric cancer. Front Immunol 2022; 13:999551. [PMID: 36189263 PMCID: PMC9520597 DOI: 10.3389/fimmu.2022.999551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Fluorouracil, also known as 5-FU, is one of the most commonly used chemotherapy drugs in the treatment of advanced gastric cancer (GC). Whereas, the presence of innate or acquired resistance largely limits its survival benefit in GC patients. Although accumulated studies have demonstrated the involvement of tumor microenvironments (TMEs) in chemo-resistance induction, so far little is known about the relevance of GC TMEs in 5-FU resistance. To this end, in this study, we investigated the relationship between TME features and 5-FU responses in GC patients using a combined analysis involving both bulk sequencing data from the TCGA database and single-cell RNA sequencing data from the GEO database. We found that depleted extracellular matrix (ECM) components such as capillary/stroma cells and enhanced immune processes such as increased number of M1 polarized macrophages/Memory T cells/Natural Killer T cells/B cells and decreased number of regulatory T cells are two important features relating to 5-FU beneficial responses in GC patients, especially in diffuse-type patients. We further validated these two features in the tumor tissues of 5-FU-benefit GC patients using immunofluorescence staining experiments. Based on this finding, we also established a Pro (63 genes) and Con (199 genes) gene cohort that could predict 5-FU responses in GC with an AUC (area under curve) score of 0.90 in diffuse-type GC patients, and further proved the partial applicability of this gene panel pan-cancer-wide. Moreover, we identified possible communications mediated by heparanase and galectin-1 which could regulate ECM remodeling and tumor immune microenvironment (TIME) reshaping. Altogether, these findings deciphered the relationship between GC TMEs and 5-FU resistance for the first time, as well as provided potential therapeutic targets and predicting rationale to overcome this chemo-resistance, which could shed some light on developing novel precision treatment strategies in clinical practice.
Collapse
Affiliation(s)
- Shaowei Dong
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | - Siyu Zhang
- School of Medicine, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Pan Zhao
- School of Medicine, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Guanchuan Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Xiaoshi Ma
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Jing Xu
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Hao Zhang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, China
| | - Jiliang Hu
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- Guangdong Engineering Technological Research Center for Nervous Anatomy and Related Clinical Applications, Shenzhen, China
| | - Chang Zou
- The Second Clinical Medical College, The First Affiliated Hospital of Southern University of Science and Technology, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| |
Collapse
|
20
|
Abstract
Organ-specific metastasis to secondary organs is dependent on the formation of a supportive pre-metastatic niche. This tissue-specific microenvironmental response is thought to be mediated by mutational and epigenetic changes to primary tumour cells resulting in altered cross-talk between cell types. This response is augmented through the release of tumour and stromal signalling mediators including cytokines, chemokines, exosomes and growth factors. Although researchers have elucidated some of the cancer-promoting features that are bespoke to organotropic metastasis to the lungs, it remains unclear if these are organ-specific or generic between organs. Understanding the mechanisms that mediate the metastasis-promoting synergy between the host microenvironment, immunity, and pulmonary structures may elucidate predictive, prognostic and therapeutic markers that could be targeted to reduce the metastatic burden of disease. Herein, we give an updated summary of the known cellular and molecular mechanisms that contribute to the formation of the lung pre-metastatic niche and tissue-specific metastasis.
Collapse
Affiliation(s)
- Oliver Cucanic
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Rae H Farnsworth
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Steven A Stacker
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Australia
| |
Collapse
|
21
|
Hua S, Xie Z, Zhang Y, Wu L, Shi F, Wang X, Xia S, Dong S, Jiang J. Identification and validation of an immune-related gene prognostic signature for clear cell renal carcinoma. Front Immunol 2022; 13:869297. [PMID: 35936012 PMCID: PMC9352939 DOI: 10.3389/fimmu.2022.869297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/27/2022] [Indexed: 12/03/2022] Open
Abstract
Clear Cell Renal Carcinoma (ccRCC) accounts for nearly 80% of renal carcinoma cases, and immunotherapy plays an important role in ccRCC therapy. However, the responses to immunotherapy and overall survival for ccRCC patients are still hard to predict. Here, we constructed an immune-related predictive signature using 19 genes based on TCGA datasets. We also analyzed its relationships between disease prognosis, infiltrating immune cells, immune subtypes, mutation load, immune dysfunction, immune escape, etc. We found that our signature can distinguish immune characteristics and predict immunotherapeutic response for ccRCC patients with better prognostic prediction value than other immune scores. The expression levels of prognostic genes were determined by RT-qPCR assay. This signature may help to predict overall survival and guide the treatment for patients with ccRCC.
Collapse
Affiliation(s)
- Shan Hua
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiwen Xie
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongqing Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wu
- Department of Urology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Fei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingjie Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujie Xia
- Department of Urology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Shengli Dong
- Nursing Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Shengli Dong, ; Juntao Jiang,
| | - Juntao Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Shengli Dong, ; Juntao Jiang,
| |
Collapse
|
22
|
Abstract
Decades of research have concluded that disruptions to Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) have profound effects on cancer progression. However, as our understanding of the tumor stroma has evolved, we can appreciate that disruptions to tumor suppressors such as PTEN should not be studied solely in an epithelial context. Inactivation of PTEN in the stroma is associated with worse outcomes in human cancers, therefore, it is important to understand activities regulated downstream of PTEN in stromal compartments. Studies reviewed herein provide evidence for important mechanistic targets downstream of PTEN signaling in cancer-associated fibroblasts (CAFs), a major component of the tumor stroma. We also discuss the potential clinical implications for these findings.
Collapse
Affiliation(s)
- Julia E Lefler
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Cara Seward
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Michael C Ostrowski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, United States; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
23
|
A Novel Prognostic Ferroptosis-Related Long Noncoding RNA Signature in Clear Cell Renal Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:6304824. [PMID: 35242188 PMCID: PMC8888116 DOI: 10.1155/2022/6304824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/18/2022] [Indexed: 12/25/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common primary malignancy of renal cancer in adults. Ferroptosis is critically associated with the prognosis of ccRCC. However, knowledge of long noncoding RNA- (lncRNA-) related ferroptosis that affects the prognosis of ccRCC is still insufficient. Using the LASSO regression, we created a risk model based on differentially expressed ferroptosis-related lncRNAs (FRLRS) in ccRCC. The analysis of Kaplan-Meier for survival, area under the curve (AUC) for diagnosis, nomogram for predicting overall survival, and gene expression for immune checkpoints were performed based on the screened independent prognostic factors. Nine lncRNAs were found to be associated with ccRCC prognosis. Furthermore, the prognostic AUC of the FRLRS signature was 0.78, demonstrating its usefulness in predicting ccRCC prognosis. The lncRNA risk model outperformed the standard clinical variables in predicting ccRCC prognosis. Finally, The Cancer Genome Atlas revealed that T cell functions, such as cytolytic activity, human leukocyte antigen activity, inflammation regulation, and type II interferon response coordination, are significantly different between two different risk levels of ccRCC. Immune checkpoints were also expressed differently in programmed cell death 1 receptor, inducible T cell costimulator, cytotoxic T-lymphocyte antigen-4, and leukocyte-associated immunoglobulin-like receptor 1. The nine FRLRS signature models may affect the prognosis of ccRCC.
Collapse
|
24
|
Xie Z, Wu L, Hua S, Zhang Y, Shi F, Chen M, Zhao S, Liu Z, Liu M, Jiang J. External Validation of the Prognostic Value of an Immune-Associated Gene Panel for Clear Cell Renal Cell Carcinomas. Front Cell Dev Biol 2022; 9:794840. [PMID: 35004689 PMCID: PMC8733896 DOI: 10.3389/fcell.2021.794840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/07/2021] [Indexed: 12/25/2022] Open
Abstract
Clear cell renal cell carcinomas (ccRCCs) are highly immune infiltrates, and many of them respond to immunotherapy with checkpoint inhibitors including anti-PD-L1 or anti-PD1 agents. However, the effect of immune genes on clinical outcomes in ccRCCs has not been fully studied. Here, we show in this study that an immune-associated gene panel has a prognostic value for clear cell renal cell carcinomas. We performed single-sample gene set enrichment analysis (ssGSEA) and cell type identification by estimating subsets of RNA transcripts (CIBERSORT) algorithms on patient-matched normal renal and RCC tissues to characterize two immunophenotypes and immunological characteristic subpopulations. Furthermore, LASSO Cox regression was applied to develop a novel prognosis-associated model for ccRCC patients based on an immune-gene panel. The results were verified by the Gene Expression Omnibus (GEO) dataset and coordinated with the clinicopathological characteristics of ccRCCs, along with genomic signatures. Finally, based on the above perspectives, we generated a nomogram with a high prognostic efficiency for ccRCC patients. Overall, this study offers a unique perspective that can contribute to improving the accuracy of prognosis prediction and treatment with immunotherapy.
Collapse
Affiliation(s)
- Zhiwen Xie
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wu
- Department of Urology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Shan Hua
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongqing Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Chen
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng Zhao
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihong Liu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Liu
- Department of Urology, The Fifth People's Hospital of Zunyi, Guizhou, China
| | - Juntao Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Urology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| |
Collapse
|
25
|
Zhong T, Jiang Z, Wang X, Wang H, Song M, Chen W, Yang S. Key genes associated with prognosis and metastasis of clear cell renal cell carcinoma. PeerJ 2022; 10:e12493. [PMID: 35036081 PMCID: PMC8740509 DOI: 10.7717/peerj.12493] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/25/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a tumor that frequently shows the hematogenous pathway and tends to be resistant to radiotherapy and chemotherapy. However, the exact mechanism of ccRCC metastasis remains unknown. METHODS Differentially expressed genes (DEGs) of three gene expression profiles (GSE85258, GSE105288 and GSE22541) downloaded from the Gene Expression Omnibus (GEO) database were analyzed by GEO2R analysis, and co-expressed DEGs among the datasets were identified using a Venn drawing tool. The co-expressed DEGs were investigated using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, and hub genes were determined based on the protein-protein interaction network established by STRING. After survival analysis performed on UALCAN website, possible key genes were selected and verified in ccRCC cell lines and ccRCC tissues (n = 44). Statistical analysis was conducted using GraphPad Prism (Version 8.1.1). RESULTS A total of 104 co-expressed DEGs were identified in the three datasets. Pathway analysis revealed that these genes were enriched in the extracellular matrix (ECM)-receptor interaction, protein digestion and absorption and focal adhesion. Survival analysis on 17 hub genes revealed that four key genes with a significant impact on survival: procollagen C-endopeptidase enhancer (PCOLCE), prolyl 4-hydroxylase subunit beta (P4HB), collagen type VI alpha 2 (COL6A2) and collagen type VI alpha 3 (COL6A3). Patients with higher expression of these key genes had worse survival than those with lower expression. In vitro experiments revealed that the mRNA expression levels of PCOLCE, P4HB and COL6A2 were three times higher and that of COL6A3 mRNA was 16 times higher in the metastatic ccRCC cell line Caki-1 than the corresponding primary cell line Caki-2. Immunohistochemistry revealed higher expression of the proteins encoded by these four genes in metastatic ccRCC compared with tumors from the corresponding primary sites, with statistical significance. CONCLUSION PCOLCE, P4HB, COL6A2 and COL6A3 are upregulated in metastatic ccRCC and might be related to poor prognosis and distant metastases.
Collapse
Affiliation(s)
- Tingting Zhong
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zeying Jiang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiangdong Wang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Honglei Wang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Meiyi Song
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenfang Chen
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shicong Yang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
26
|
Liu Y, Zhang Y, Tan Z, Wang J, Hu Y, Sun J, Bao M, Huang P, Ge M, Chai YJ, Zheng C. Lysyl oxidase promotes anaplastic thyroid carcinoma cell proliferation and metastasis mediated via BMP1. Gland Surg 2022; 11:245-257. [PMID: 35242686 PMCID: PMC8825512 DOI: 10.21037/gs-21-908] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/14/2022] [Indexed: 12/01/2023]
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) is an extremely aggressive solid tumor with no effective treatment at present. Because of the rapid growth and aggressiveness, nearly all patients die within six months after developing ATC. Hence, more research regarding novel therapeutic targets for ATC is urgently needed. METHODS Single-cell RNA sequencing data and microarray data of ATC were retrieved from the Gene Expression Omnibus (GEO) database. Cell clustering was performed using the Seurat package. Then, differential expression and functional enrichment analyses were performed. Gene set enrichment analysis (GSEA) was further used to investigate the functional enrichment of lysyl oxidase (LOX) and bone morphogenetic protein-1 (BMP1). The expression levels of LOX and BMP1 were measured using quantitative real-time PCR and Western blot. LOX and BMP1 were knocked down using si-RNAs. Cell proliferation was evaluated by the CCK-8 and clone formation assays. Cell migration and invasion were assessed by the wound healing assay and Transwell assay, respectively. RESULTS LOX was upregulated at the single-cell level, as well as in ATC tissues and cell lines. LOX knockdown significantly inhibited ATC cell proliferation. Furthermore, the migration and invasion of ATC cells were remarkably inhibited after LOX inhibition. In addition, BMP1 regulated LOX expression in 8505C cells, while BMP1 overexpression restored the LOX activity blocked by the LOX inhibitor BAPN. BMP1 could also induce the cell proliferation and metastasis of ATC. CONCLUSIONS LOX/BMP1 mediates the malignant progression of ATC, highlighting the potential application of LOX/BMP1 in the treatment of ATC. This study provides new insights for efficient therapeutic agents based on the LOX/BMP1 axis.
Collapse
Affiliation(s)
- Yujia Liu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital; Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yiwen Zhang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital; Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Zhuo Tan
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
- Department of Head and Neck & Thyroid Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jiafeng Wang
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
- Department of Head and Neck & Thyroid Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ying Hu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital; Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jiao Sun
- Department of Pharmacy, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Meihua Bao
- Academician Workstation, School of Stomatology, Changsha Medical University, Changsha, China
| | - Ping Huang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People’s Hospital; Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Minghua Ge
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
- Department of Head and Neck & Thyroid Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Young Jun Chai
- Department of Surgery, Seoul National University College of Medicine, Seoul Metropolitan Government, Seoul National University Boramae Medical Center, Transdisciplinary Department of Medicine & Advanced Technology, Seoul National University Hospital, Seoul, Korea
| | - Chuanming Zheng
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
- Department of Head and Neck & Thyroid Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
27
|
Tong J, Meng X, Lv Q, Yuan H, Li W, Xiao W, Zhang X. The Downregulation of Prognosis- and Immune Infiltration-Related Gene CYFIP2 Serves as a Novel Target in ccRCC. Int J Gen Med 2021; 14:6587-6599. [PMID: 34703279 PMCID: PMC8523908 DOI: 10.2147/ijgm.s335713] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/06/2021] [Indexed: 01/18/2023] Open
Abstract
Background Increasing evidence indicated that the aberrant expression of the cytoplasmic FMR1-interacting protein (CYFIP) family might possess critical role and potential functions in cancer. But the role of CYFIP2 in clear cell renal cell carcinoma (ccRCC) is still uncharacteristic. Methods We investigated the Cancer Genome Atlas Kidney Clear Cell Carcinoma (TCGA-KIRC) database for the expression profile, clinicopathological variables, clinical prognosis information, and promoter methylation levels of CYFIPs in ccRCC. The aberrant CYFIP2 protein expression was validated by the Human Protein Atlas (HPA) and Clinical Proteomic Tumor Analysis Consortium (CPTAC). Quantitative real-time polymerase chain reaction (qRT-PCR) was used to uncover CYFIP2 mRNA levels in 28 pairs of ccRCC cancer tissues. Kaplan–Meier analysis, univariate and multivariate Cox proportional hazard regression were performed to assess CYFIPs’ prognosis value. Gene set enrichment analysis (GSEA) was used to determined hallmark functions, gene ontology of CYFIP2. TIMER database was utilized to assess the correlation with immune infiltration in ccRCC. Results Results showed CYFIP2 was downregulated in ccRCC, relative to paired normal tissues in TCGA-KIRC database and 28 pairs of clinical samples (P < 0.0001). Similarly, a decreased CYFIP2 protein expression was confirmed by ccRCC tissues. The results showed CYFIP2 was negatively regulated by promoter DNA methylation. Survival analysis results showed CYFIP2 could be an independent biomarker for ccRCC and its reduction predicted a poor overall survival (OS) and disease-free survival (DFS). GSEA showed CYFIP2 was involved in metabolic pathways and epithelial–mesenchymal transition (EMT). Immune infiltration analysis revealed that a list of immune markers was significantly correlated with CYFIP2 expression especially with CD4+ cells and CD8+ cells in ccRCC. Conclusion These results show that CYFIP2 was downregulated in ccRCC patients and predicted an unfavorable prognosis. CYFIP2 might be a potential novel prognostic molecule, and related to immune infiltration, the metabolism, as well as EMT process in ccRCC. CYFIP2 could act as tumor suppressor gene in ccRCC and positive modulation of CYFIP2 might lead to development of a novel strategy for ccRCC treatment.
Collapse
Affiliation(s)
- Junwei Tong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xiangui Meng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Qingyang Lv
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Hongwei Yuan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Weiquan Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| |
Collapse
|
28
|
Deng L, Wang P, Qu Z, Liu N. The Construction and Analysis of ceRNA Network and Immune Infiltration in Kidney Renal Clear Cell Carcinoma. Front Genet 2021; 12:667610. [PMID: 34567057 PMCID: PMC8455958 DOI: 10.3389/fgene.2021.667610] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Kidney renal clear cell carcinoma (KIRC) has the highest invasion, mortality and metastasis of the renal cell carcinomas and seriously affects patient’s quality of life. However, the composition of the immune microenvironment and regulatory mechanisms at transcriptomic level such as ceRNA of KIRC are still unclear. Methods: We constructed a ceRNA network associated with KIRC by analyzing the long non-coding RNA (lncRNA), miRNA and mRNA expression data of 506 tumor tissue samples and 71 normal adjacent tissue samples downloaded from The Cancer Genome Atlas (TCGA) database. In addition, we estimated the proportion of 22 immune cell types in these samples through “The Cell Type Identification by Estimating Relative Subsets of RNA Transcripts.” Based on the ceRNA network and immune cells screened by univariate Cox analysis and Lasso regression, two nomograms were constructed to predict the prognosis of patients with KIRC. Receiver operating characteristic curves (ROC) and calibration curves were employed to assess the discrimination and accuracy of the nomograms. Consequently, co-expression analysis was carried out to explore the relationship between each prognostic gene in a Cox proportional hazards regression model of ceRNA and each survival-related immune cell in a Cox proportional hazards regression model of immune cell types to reveal the potential regulatory mechanism. Results: We established a ceRNA network consisting of 12 lncRNAs, 25 miRNAs and 136 mRNAs. Two nomograms containing seven prognostic genes and two immune cells, respectively, were successfully constructed. Both ROC [area under curves (AUCs) of 1, 3, and 5-year survival in the nomogram based on ceRNA network: 0.779, 0.747, and 0.772; AUCs of 1, 3, and 5-year survivals in nomogram based on immune cells: 0.603, 0.642, and 0.607] and calibration curves indicated good accuracy and clinical application value of both models. Through co-correlation analysis between ceRNA and immune cells, we found both LINC00894 and KIAA1324 were positively correlated with follicular helper T (Tfh) cells and negatively correlated with resting mast cells. Conclusion: Based on the ceRNA network and tumor-infiltrating immune cells, we constructed two nomograms to predict the survival of KIRC patients and demonstrated their value in improving the personalized management of KIRC.
Collapse
Affiliation(s)
- Lugang Deng
- School of Public Health, Guangzhou Medical University, Guangzhou, China.,South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Peixi Wang
- Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China
| | - Zhi Qu
- Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China
| | - Nan Liu
- South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China.,Institute of Chronic Disease Risks Assessment, School of Nursing and Health, Henan University, Kaifeng, China
| |
Collapse
|
29
|
Meng X, Yuan H, Li W, Xiong Z, Dong W, Xiao W, Zhang X. Solute carrier family 16 member 5 downregulation and its methylation might serve as a prognostic indicator of prostate cancer. IUBMB Life 2021; 73:1363-1377. [PMID: 34549875 DOI: 10.1002/iub.2560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/04/2021] [Accepted: 09/13/2021] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa), characterized by high invasion, metastasis, and recurrence, is the most prevalent malignant tumor in men worldwide. A clear understanding of the underlying molecular mechanisms and their role during PCa tumorigenesis can help develop prognostic and targeted therapies. We analyzed datasets from public databases, including the Cancer Genome Atlas (TCGA) and Oncomine and Gene Expression Profiling Interactive Analysis for differential expression of solute carrier family 16 member 5 (SLC16A5). We further investigated its relationship with clinical stage, pathological grade, and prognosis of PCa. The promoter methylation level of SLC16A5 in PCa was also investigated by UALCAN. We also utilized datasets from UCSC Xena to explore the prognostic role of SLC16A5 methylation levels and CpG site. Correlations between SLC16A5 and immune infiltration were discovered through TIMER. We observed significantly lower levels of SLC16A5 mRNA in PCa relative to normal tissues across six datasets from Oncomine database (p < .001) and 498 cases from TCGA database (p < .0001). SLC16A5 is strongly negatively regulated by its DNA methylation, with a Spearman of -0.81 and Pearson of -0.80 (p < .001). The aberrant SLC16A5 expression resulted in a significant relationship with clinical stage, pathological grade, and lower SLC16A5 mRNA expression, and its hypermethylation was related to a poorer PCa prognosis. SLC16A5 acted as an important factor for PCa diagnosis, with an AUC of 0.9038 (95% CI: 0.8597-0.9479; p < .0001). Besides, the aberrant SLC16A5 expression revealed close correlations with multiple immune cells. Overall, these results indicate that decreased SLC16A5 expression might be a potential biomarker for determining prognosis and immune infiltration in PCa. The positive SLC16A5 modulation might be a promising therapeutic target for PCa.
Collapse
Affiliation(s)
- Xiangui Meng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongwei Yuan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiquan Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyong Xiong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Dong
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Meng X, Yuan H, Li W, Xiao W, Zhang X. Biomarker Screening and Prognostic Significance Analysis for Renal Cell Carcinoma. Int J Gen Med 2021; 14:5255-5267. [PMID: 34522125 PMCID: PMC8434883 DOI: 10.2147/ijgm.s325347] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/11/2021] [Indexed: 12/17/2022] Open
Abstract
Background Studies report that conventional treatment of clear cell renal cell carcinoma (ccRCC) is effective, but several advanced patients present with poor prognosis. The current study explored potential new tumor markers and therapeutic targets in advanced ccRCC. Methods Biomarker gene expression of ccRCC was retrieved from GEO database and the Cancer Genome Atlas Kidney Clear Cell Carcinoma (TCGA-KIRC) database. Gene ontology (GO) analysis and protein–protein interaction (PPI) networks of biomarker genes were constructed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) tool. Kaplan–Meier analysis and receiver operating characteristic curve (ROC) analysis were performed to explore the prognostic and diagnostic roles of these genes. Gene set enrichment analysis (GSEA) analysis was used to determine hallmark functions of the biomarker genes. qRT-PCR was used to verify the reliability of the analysis results in tumor tissues. Results A total of 21 upregulated genes were identified between advanced ccRCC and early ccRCC (grade III+IV vs grade I+II). Gene ontology analysis showed that the 21 upregulated genes were mainly implicated in biological processes including metabolic and lipid transport. The findings showed that 7 out of the 21 genes were significantly upregulated in 72-paired samples retrieved from the TCGA-KIRC. High expression of 5 genes indicated a poor prognosis of overall survival and disease-free survival in KIRC. Three genes effectively distinguished renal cancer tissue and adjacent renal tissues in a total of 533 ccRCC samples. GSEA showed that the 3 biomarkers were significantly enriched in epithelial–mesenchymal transition, G2M checkpoint, and angiogenesis. The results of qRT-PCR showed that STEAP3, IBSP, and AQP9 had a significant identification effect in ccRCC. Conclusion The findings showed that 3 biomarkers were significantly upregulated in advanced ccRCC and could be used for diagnosis, prediction, and potential novel therapeutic targets for progression of ccRCC.
Collapse
Affiliation(s)
- Xiangui Meng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Hongwei Yuan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Weiquan Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Wen Xiao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, People's Republic of China.,Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| |
Collapse
|
31
|
Longmate WM, Miskin RP, Van De Water L, DiPersio CM. Epidermal Integrin α3β1 Regulates Tumor-Derived Proteases BMP-1, Matrix Metalloprotease-9, and Matrix Metalloprotease-3. JID INNOVATIONS : SKIN SCIENCE FROM MOLECULES TO POPULATION HEALTH 2021; 1:100017. [PMID: 34909716 PMCID: PMC8659409 DOI: 10.1016/j.xjidi.2021.100017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/05/2021] [Accepted: 04/14/2021] [Indexed: 10/28/2022]
Abstract
As the major cell surface receptors for the extracellular matrix, integrins regulate adhesion and migration and have been shown to drive tumor growth and progression. Previous studies showed that mice lacking integrin α3β1 in the epidermis fail to form skin tumors during two-step chemical tumorigenesis, indicating a protumorigenic role for α3β1. Furthermore, genetic ablation of α3β1 in established skin tumors caused their rapid regression, indicating an essential role in the maintenance of tumor growth. In this study, analysis of immortalized keratinocyte lines and their conditioned media support a role for α3β1 in regulating the expression of several extracellular proteases of the keratinocyte secretome, namely BMP-1, matrix metalloprotease (MMP)-9, and MMP-3. Moreover, immunofluorescence revealed reduced levels of each protease in α3β1-deficient tumors, and RNA in situ hybridization showed that their expression was correspondingly reduced in α3β1-deficient tumor cells in vivo. Bioinformatic analysis confirmed that the expression of BMP1, MMP9, and MMP3 genes correlate with the expression of ITGA3 (gene encoding the integrin α3 subunit) in human squamous cell carcinoma and that high ITGA3 and MMP3 associate with poor survival outcome in these patients. Overall, our findings identify α3β1 as a regulator of several proteases within the secretome of epidermal tumors and as a potential therapeutic target.
Collapse
Key Words
- CM, conditioned medium
- ECM, extracellular matrix
- IMK, immortalized mouse keratinocyte
- ISH, in situ hybridization
- KC, keratinocyte
- MK, mouse keratinocyte
- MMP, matrix metalloprotease
- SCC, squamous cell carcinoma
- TME, tumor microenvironment
- TMK, transformed mouse keratinocyte
Collapse
Affiliation(s)
| | - Rakshitha Pandulal Miskin
- The Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - Livingston Van De Water
- Department of Surgery, Albany Medical College, Albany, New York, USA,The Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - C. Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, New York, USA,Department of Molecular and Cellular Physiology (MCP), Albany Medical College, Albany, New York, USA,Correspondence: C. Michael DiPersio, Department of Surgery, Albany Medical College, Mail Code 8, Room MR-421, 47 New Scotland Avenue, Albany, New York 12208-3479, USA.
| |
Collapse
|
32
|
Tian C, Huang Y, Clauser KR, Rickelt S, Lau AN, Carr SA, Vander Heiden MG, Hynes RO. Suppression of pancreatic ductal adenocarcinoma growth and metastasis by fibrillar collagens produced selectively by tumor cells. Nat Commun 2021; 12:2328. [PMID: 33879793 PMCID: PMC8058088 DOI: 10.1038/s41467-021-22490-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 03/16/2021] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a collagen-rich dense extracellular matrix (ECM) that promotes malignancy of cancer cells and presents a barrier for drug delivery. Data analysis of our published mass spectrometry (MS)-based studies on enriched ECM from samples of progressive PDAC stages reveal that the C-terminal prodomains of fibrillar collagens are partially uncleaved in PDAC ECM, suggesting reduced procollagen C-proteinase activity. We further show that the enzyme responsible for procollagen C-proteinase activity, bone morphogenetic protein1 (BMP1), selectively suppresses tumor growth and metastasis in cells expressing high levels of COL1A1. Although BMP1, as a secreted proteinase, promotes fibrillar collagen deposition from both cancer cells and stromal cells, only cancer-cell-derived procollagen cleavage and deposition suppresses tumor malignancy. These studies reveal a role for cancer-cell-derived fibrillar collagen in selectively restraining tumor growth and suggest stratification of patients based on their tumor epithelial collagen I expression when considering treatments related to perturbation of fibrillar collagens.
Collapse
MESH Headings
- Animals
- Bone Morphogenetic Protein 1/metabolism
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/secondary
- Cell Line, Tumor
- Collagen Type I/chemistry
- Collagen Type I/genetics
- Collagen Type I/metabolism
- Collagen Type I, alpha 1 Chain
- Disease Progression
- Extracellular Matrix/metabolism
- Extracellular Matrix Proteins/metabolism
- Fibrillar Collagens/chemistry
- Fibrillar Collagens/genetics
- Fibrillar Collagens/metabolism
- Humans
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Mutagenesis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Procollagen/chemistry
- Procollagen/genetics
- Procollagen/metabolism
- Protein Domains
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
Collapse
Affiliation(s)
- Chenxi Tian
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ying Huang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Steffen Rickelt
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Allison N Lau
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Richard O Hynes
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
33
|
Fei Z, Xie R, Chen Z, Xie J, Gu Y, Zhou Y, Xu T. Establishment of a Novel Risk Score System of Immune Genes Associated With Prognosis in Esophageal Carcinoma. Front Oncol 2021; 11:625271. [PMID: 33859939 PMCID: PMC8042266 DOI: 10.3389/fonc.2021.625271] [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: 11/02/2020] [Accepted: 03/15/2021] [Indexed: 01/19/2023] Open
Abstract
Background Few studies have addressed the role of immune-related genes in the survival and prognosis of different esophageal cancer (EC) sub-types. We established two new prognostic model indexes by bioinformatics analysis to select patients with esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC) who may benefit from immunotherapy. Methods Based on TCGA and ImmPort data sets, we screened immune genes differentially expressed between tumor and normal tissues in ESCC and EAC and analyzed the relationship between these genes and patient survival outcomes. We established the risk score models of immune-related genes in ESCC and EAC by multivariate COX regression analysis. Results We identified 12 and 11 immune-related differentially expressed genes associated with the clinical prognosis of ESCC and EAC respectively, based on which two prognostic risk score models of the two EC sub-types were constructed. It was found that the survival probability of patients with high scores was significantly lower than that of patients with low scores (p < 0.001). BMP1, EGFR, S100A12, HLA-B, TNFSF18, IL1B, MAPT and OXTR were significantly related to sex, TNM stage or survival outcomes of ESCC or EAC patients (p < 0.05). In addition, the risk score of ESCC was significantly correlated with the level of B cell infiltration in immune cells (p < 0.05). Conclusions The prognosis-related immune gene model indexes described herein prove to be useful prognostic biomarkers of the two EC sub-types in that they may provide a reference direction for looking for the beneficiaries of immunotherapy for EC patients.
Collapse
Affiliation(s)
- Zhenghua Fei
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rongrong Xie
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhi Chen
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junhui Xie
- Department of Head and Neck Surgery, Tumor Hospital of Ganzhou, Ganzhou, China
| | - Yuyang Gu
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yue Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tongpeng Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
34
|
Petersen EV, Chudakova DA, Skorova EY, Anikin V, Reshetov IV, Mynbaev OA. The Extracellular Matrix-Derived Biomarkers for Diagnosis, Prognosis, and Personalized Therapy of Malignant Tumors. Front Oncol 2020; 10:575569. [PMID: 33425730 PMCID: PMC7793707 DOI: 10.3389/fonc.2020.575569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/10/2020] [Indexed: 01/18/2023] Open
Abstract
The tumor biomarkers already have proven clinical value and have become an integral part in cancer management and modern translational oncology. The tumor tissue microenvironment (TME), which includes extracellular matrix (ECM), signaling molecules, immune and stromal cells, and adjacent non-tumorous tissue, contributes to cancer pathogenesis. Thus, TME-derived biomarkers have many clinical applications. This review is predominately based on the most recent publications (manuscripts published in a last 5 years, or seminal publications published earlier) and fills a gap in the current literature on the cancer biomarkers derived from the TME, with particular attention given to the ECM and products of its processing and degradation, ECM-associated extracellular vesicles (EVs), biomechanical characteristics of ECM, and ECM-derived biomarkers predicting response to the immunotherapy. We discuss the clinical utility of the TME-incorporating three-dimensional in vitro and ex vivo cell culture models for personalized therapy. We conclude that ECM is a critical driver of malignancies and ECM-derived biomarkers should be included in diagnostics and prognostics panels of markers in the clinic.
Collapse
Affiliation(s)
- Elena V. Petersen
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Daria A. Chudakova
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Ekaterina Yu. Skorova
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Vladimir Anikin
- Harefield Hospital, The Royal Brompton and Harefield Hospitals NHS Foundation Trust, Harefield, United Kingdom
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Igor V. Reshetov
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Ospan A. Mynbaev
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
35
|
Anastasi C, Rousselle P, Talantikite M, Tessier A, Cluzel C, Bachmann A, Mariano N, Dussoyer M, Alcaraz LB, Fortin L, Aubert A, Delolme F, El Kholti N, Armengaud J, Fournié P, Auxenfans C, Valcourt U, Goff SVL, Moali C. BMP-1 disrupts cell adhesion and enhances TGF-β activation through cleavage of the matricellular protein thrombospondin-1. Sci Signal 2020; 13:13/639/eaba3880. [PMID: 32636307 DOI: 10.1126/scisignal.aba3880] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bone morphogenetic protein 1 (BMP-1) is an important metalloproteinase that synchronizes growth factor activation with extracellular matrix assembly during morphogenesis and tissue repair. The mechanisms by which BMP-1 exerts these effects are highly context dependent. Because BMP-1 overexpression induces marked phenotypic changes in two human cell lines (HT1080 and 293-EBNA cells), we investigated how BMP-1 simultaneously affects cell-matrix interactions and growth factor activity in these cells. Increasing BMP-1 led to a loss of cell adhesion that depended on the matricellular glycoprotein thrombospondin-1 (TSP-1). BMP-1 cleaved TSP-1 between the VWFC/procollagen-like domain and the type 1 repeats that mediate several key TSP-1 functions. This cleavage induced the release of TSP-1 C-terminal domains from the extracellular matrix and abolished its previously described multisite cooperative interactions with heparan sulfate proteoglycans and CD36 on HT1080 cells. In addition, BMP-1-dependent proteolysis potentiated the TSP-1-mediated activation of latent transforming growth factor-β (TGF-β), leading to increased signaling through the canonical SMAD pathway. In primary human corneal stromal cells (keratocytes), endogenous BMP-1 cleaved TSP-1, and the addition of exogenous BMP-1 enhanced cleavage, but this had no substantial effect on cell adhesion. Instead, processed TSP-1 promoted the differentiation of keratocytes into myofibroblasts and stimulated production of the myofibroblast marker α-SMA, consistent with the presence of processed TSP-1 in human corneal scars. Our results indicate that BMP-1 can both trigger the disruption of cell adhesion and stimulate TGF-β signaling in TSP-1-rich microenvironments, which has important potential consequences for wound healing and tumor progression.
Collapse
Affiliation(s)
- Cyril Anastasi
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Patricia Rousselle
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Maya Talantikite
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Agnès Tessier
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Caroline Cluzel
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Alice Bachmann
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Natacha Mariano
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Mélissa Dussoyer
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Lindsay B Alcaraz
- University of Lyon, Centre Léon Bérard, INSERM U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), F-69373 Lyon, France
| | - Laëtitia Fortin
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Alexandre Aubert
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Frédéric Delolme
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France.,University of Lyon, ENS de Lyon, INSERM US8, CNRS UMS3444, SFR Biosciences, F-69366 Lyon, France
| | - Naïma El Kholti
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Jean Armengaud
- CEA Marcoule, Innovative Technologies for Detection and Diagnostics Laboratory (DRF/Joliot/DMTS/SPI/Li2D), F-30200 Bagnols-sur-Cèze, France
| | - Pierre Fournié
- Purpan University Hospital, Ophthalmology Department, F-31059 Toulouse, France.,University of Toulouse, CNRS UMR 5165, INSERM U1056, Epithelial Differentiation and Rheumatoid Autoimmunity Unit (UDEAR), F-31059 Toulouse, France
| | - Céline Auxenfans
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France.,Hospices Civils de Lyon, Tissue and Cell Bank, F-69437 Lyon, France
| | - Ulrich Valcourt
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France.,University of Lyon, Centre Léon Bérard, INSERM U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), F-69373 Lyon, France
| | - Sandrine Vadon-Le Goff
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France
| | - Catherine Moali
- University of Lyon, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), F-69367 Lyon, France.
| |
Collapse
|
36
|
Chauhan SJ, Thyagarajan A, Sahu RP. Functional Significance of Mirna-149 in Lung Cancer: Can it be Utilized as a Potential Biomarker or a Therapeutic Target? AUSTIN JOURNAL OF MEDICAL ONCOLOGY 2020; 7:1048. [PMID: 38628497 PMCID: PMC11019914 DOI: 10.26420/austinjmedoncol.2020.1048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Accumulating evidence has documented the significance of miR-149 as a promising tumor-suppressive non-coding RNA that play critical roles in regulating genes involved in cancer growth and metastasis. Notably, the ability of miR-149 to be utilized as a potential biomarker in the diagnosis/prognosis or a therapeutic target has also been explored using various cellular and preclinical models, as well as in clinical settings of lung cancer. While the applicability of miR-149 in assessing tumor progression has been suggested, its potential in predicting treatment outcomes is needed to be verified in diverse settings of lung cancer patients. The current review presents an overview of the functional significance of miR-149 with ongoing challenges in non-small cell lung cancer.
Collapse
Affiliation(s)
- S J Chauhan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, USA
| | - A Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, USA
| | - R P Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, USA
| |
Collapse
|
37
|
Han X, Hu F, Chen F, Wang W. The inhibition of bone morphogenetic protein 1 attenuates endometriosis lesions in vivo and in vitro. Arch Gynecol Obstet 2020; 302:415-422. [PMID: 32524385 DOI: 10.1007/s00404-020-05612-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 05/19/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE To investigate the potential role of Bone morphogenetic protein 1 (BMP1) in endometriosis lesions. METHODS Endometriosis model in mice was established. The expression of BMP1-3 expression in mice of endometriosis lesions was evaluated. The effect of the treatment with anti-BMP1 antibodies on the expression of MMP2, MMP9, TGF-β, IL-17, IL-1β, Col1a1 and Col1a2 levels in mice was evaluated. In endometriosis cell model, the expression of IL-17, IL-1β, MMP2 and MMP9 levels and MIF, YWHAZ, β-catenin and CAP39 mRNA levels was also detected. RESULTS The expression of BMP1-3 expression was upregulated in mice of endometriosis lesions (p < 0.01). Treatment with anti-BMP1 antibodies dose-dependently reduced MMP2, MMP9, TGF-β, IL-17, IL-1β, Col1a1 and Col1a2 levels in mice (p < 0.01). Treatment with anti-BMP1 antibodies suppressed TGF-β/PI3K/Akt signaling pathway. In vitro cell, si-BMP1 suppressed TGF-β/PI3K/Akt signaling pathway. CONCLUSION The data support the hypothesis that the inhibition of BMP1 is involved in the pathogenesis of endometriosis lesions.
Collapse
Affiliation(s)
- Xinyue Han
- Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, China
| | - Feifei Hu
- Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, China
| | - Fang Chen
- Department of Obstetrics and Gynecology, Baotou City Central Hospital, Baotou, China
| | - Wei Wang
- Department of Neurology, The University of Hong Kong-Shenzhen Hospital, 1 Haiyuan 1st Road, Futian District, Shenzhen City, 518000, China.
| |
Collapse
|
38
|
Peng Q, Zhou Y, Jin L, Cao C, Gao C, Zhou J, Yang D, Zhu J. Development and validation of an integrative methylation signature and nomogram for predicting survival in clear cell renal cell carcinoma. Transl Androl Urol 2020; 9:1082-1098. [PMID: 32676392 PMCID: PMC7354314 DOI: 10.21037/tau-19-853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Growing evidence has shown that genetic or epigenetic alterations are highly involved in the initiation and progression of renal cell carcinoma (RCC). This study aimed to find prognostic methylation markers in clear cell RCC (ccRCC). Methods In this study, we developed and confirmed an integrated and comprehensive methylation signature by integrating DNA methylation, gene expression, and The Cancer Genome Atlas (TCGA) survival data. First, the methylation signature was found and checked based on data analysis of published datasets. Then, independent predictive factors were selected using the Cox proportional model and incorporated into the nomogram. Finally, the predictive nomogram was derived and validated using a concordance index and calibration plots. Results A series of differentially expressed and methylated genes were identified. After intersection analysis, Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, protein-protein interaction (PPI) analysis, and correlation analysis, FCGR1A, F2, and NOD2 were established as a predictive signature. According to the Kaplan-Meier survival analysis, the risk score system based on the predictive signature was able to stratify the patients into high- and low-risk groups with significantly different overall survival. The receiver operating characteristic (ROC) analysis further showed that the predictive signature yielded high sensitivity and specificity in predicting the prognosis outcome of ccRCC patients. Moreover, univariate and multivariate Cox regression analysis confirmed that the three-gene methylation signature was an independent prognostic factor in ccRCC. Finally, a nomogram comprising the predictive signature and several independent variables were constructed and proved to effectively predict ccRCC patient survival. Conclusions The three-gene methylation signature was revealed to be a potential novel and independent adverse predictor of prognosis for ccRCC patients and may serve as a promising marker for treatment management and survival outcome improvement. However, substantial validation experiments are required to characterize the molecular background of the predictive signature.
Collapse
Affiliation(s)
- Qiliang Peng
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China
| | - Yibin Zhou
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Lu Jin
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Cao
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Gao
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianfang Zhou
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dongrong Yang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Zhu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
39
|
Ye T, Wan X, Li J, Feng J, Guo J, Li G, Liu J. The Clinical Significance of PPEF1 as a Promising Biomarker and Its Potential Mechanism in Breast Cancer. Onco Targets Ther 2020; 13:199-214. [PMID: 32021267 PMCID: PMC6955604 DOI: 10.2147/ott.s229432] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Background Breast cancer (BC) is the leading cause of malignancy death in females worldwide. While intense efforts have been made to elucidate the pathogeny, the molecular mechanism of BC remains elusive. Thus, this study aimed to investigate the role of PPEF1 in the progression of BC and further explore the better clinical significance. Methods The diagnostic and prognostic values of elevated PPEF1 expression in BC were unveiled via public databases analysis. In addition, Gene Ontology (GO), Gene Set Enrichment Analysis (GSEA) and Protein–protein interaction (PPI) analysis were performed to explore the potential functions and molecular mechanisms of PPEF1 in BC progression. Experimentally, transwell and CCK-8 assays were carried out to estimate the effects of PPEF1 on the BC metastasis. Meanwhile, the differential expressions of PPEF1 in paraffin-embedded tissues and serum samples were, respectively, analyzed by Immunohistochemical (IHC) analysis and enzyme-linked immunosorbent assay (ELISA) kit. Results The transcriptional levels of PPEF1 were higher in BC than in normal breast tissues or adjacent normal tissues. Moreover, survival analysis revealed that higher PPEF1 expression was negatively associated with overall survival (OS), all events-free (AE-free) and metastatic recurrence-free (MR-free) survival, and further was an independent risk factor of unfavorable prognosis in BC patients. Additionally, the present study provided the first evidence that PPEF1 participated in multiple biological processes and underly signaling pathways involving in tumorigenesis and development of BC. Furthermore, PPEF1 promotes the BC progression and can be used as a noninvasive diagnostic marker. Noteworthy, the combined determination of serum PPEF1 and traditional tumor markers can enhance diagnostic accuracy thus is of vital importance in the early diagnosis of BC. Conclusion PPEF1 exerted a tumorigenic role and involved in molecular mechanism of tumorigenesis in BC which served as a promising biomarker for prognosis and diagnosis.
Collapse
Affiliation(s)
- Ting Ye
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Xue Wan
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Jingyuan Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Jia Feng
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Jinglan Guo
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Guangrong Li
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan 646000, People's Republic of China
| |
Collapse
|
40
|
Targeting the lysyl oxidases in tumour desmoplasia. Biochem Soc Trans 2019; 47:1661-1678. [DOI: 10.1042/bst20190098] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 02/08/2023]
Abstract
The extracellular matrix (ECM) is a fundamental component of tissue microenvironments and its dysregulation has been implicated in a number of diseases, in particular cancer. Tumour desmoplasia (fibrosis) accompanies the progression of many solid cancers, and is also often induced as a result of many frontline chemotherapies. This has recently led to an increased interest in targeting the underlying processes. The major structural components of the ECM contributing to desmoplasia are the fibrillar collagens, whose key assembly mechanism is the enzymatic stabilisation of procollagen monomers by the lysyl oxidases. The lysyl oxidase family of copper-dependent amine oxidase enzymes are required for covalent cross-linking of collagen (as well as elastin) molecules into the mature ECM. This key step in the assembly of collagens is of particular interest in the cancer field since it is essential to the tumour desmoplastic response. LOX family members are dysregulated in many cancers and consequently the development of small molecule inhibitors targeting their enzymatic activity has been initiated by many groups. Development of specific small molecule inhibitors however has been hindered by the lack of crystal structures of the active sites, and therefore alternate indirect approaches to target LOX have also been explored. In this review, we introduce the importance of, and assembly steps of the ECM in the tumour desmoplastic response focussing on the role of the lysyl oxidases. We also discuss recent progress in targeting this family of enzymes as a potential therapeutic approach.
Collapse
|
41
|
Extracellular Matrix Alterations in Metastatic Processes. Int J Mol Sci 2019; 20:ijms20194947. [PMID: 31591367 PMCID: PMC6802000 DOI: 10.3390/ijms20194947] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 09/26/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a complex network of extracellular-secreted macromolecules, such as collagen, enzymes and glycoproteins, whose main functions deal with structural scaffolding and biochemical support of cells and tissues. ECM homeostasis is essential for organ development and functioning under physiological conditions, while its sustained modification or dysregulation can result in pathological conditions. During cancer progression, epithelial tumor cells may undergo epithelial-to-mesenchymal transition (EMT), a morphological and functional remodeling, that deeply alters tumor cell features, leading to loss of epithelial markers (i.e., E-cadherin), changes in cell polarity and intercellular junctions and increase of mesenchymal markers (i.e., N-cadherin, fibronectin and vimentin). This process enhances cancer cell detachment from the original tumor mass and invasiveness, which are necessary for metastasis onset, thus allowing cancer cells to enter the bloodstream or lymphatic flow and colonize distant sites. The mechanisms that lead to development of metastases in specific sites are still largely obscure but modifications occurring in target tissue ECM are being intensively studied. Matrix metalloproteases and several adhesion receptors, among which integrins play a key role, are involved in metastasis-linked ECM modifications. In addition, cells involved in the metastatic niche formation, like cancer associated fibroblasts (CAF) and tumor associated macrophages (TAM), have been found to play crucial roles in ECM alterations aimed at promoting cancer cells adhesion and growth. In this review we focus on molecular mechanisms of ECM modifications occurring during cancer progression and metastatic dissemination to distant sites, with special attention to lung, liver and bone. Moreover, the functional role of cells forming the tumor niche will also be reviewed in light of the most recent findings.
Collapse
|