1
|
Mótyán JA, Tőzsér J. The human retroviral-like aspartic protease 1 (ASPRV1): From in vitro studies to clinical correlations. J Biol Chem 2024; 300:107634. [PMID: 39098535 PMCID: PMC11402058 DOI: 10.1016/j.jbc.2024.107634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/06/2024] Open
Abstract
The human retroviral-like aspartic protease 1 (ASPRV1) is a retroviral-like protein that was first identified in the skin due to its expression in the stratum granulosum layer of the epidermis. Accordingly, it is also referred to as skin-specific aspartic protease. Similar to the retroviral polyproteins, the full-length ASPRV1 also undergoes self-proteolysis, the processing of the precursor is necessary for the autoactivation of the protease domain. ASPRV1's functions are well-established at the level of the skin: it is part of the epidermal proteolytic network and has a significant contribution to skin moisturization via the limited proteolysis of filaggrin; it is only natural protein substrate identified so far. Filaggrin and ASPRV1 are also specific for mammalians, these proteins provide unique features for the skins of these species, and the importance of filaggrin processing in hydration is proved by the fact that some ASPRV1 mutations are associated with skin diseases such as ichthyosis. ASPRV1 was also found to be expressed in macrophage-like neutrophil cells, indicating that its functions are not limited to the skin. In addition, differential expression of ASPRV1 was detected in many diseases, with yet unknown significance. The currently known enzymatic characteristics-that had been revealed mainly by in vitro studies-and correlations with pathogenic phenotypes imply potentially important functions in multiple cell types, which makes the protein a promising target of functional studies. In this review we describe the currently available knowledge and future perspective in regard to ASPRV1.
Collapse
Affiliation(s)
- János András Mótyán
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - József Tőzsér
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
2
|
Siegel F, Schmidt H, Juneja M, Smith J, Herrmann P, Kobelt D, Sharma K, Fichtner I, Walther W, Dittmar G, Volkmer R, Rathjen FG, Schlag PM, Stein U. GIPC1 regulates MACC1-driven metastasis. Front Oncol 2023; 13:1280977. [PMID: 38144523 PMCID: PMC10748395 DOI: 10.3389/fonc.2023.1280977] [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: 08/21/2023] [Accepted: 11/14/2023] [Indexed: 12/26/2023] Open
Abstract
Background Identification of cancer metastasis-relevant molecular networks is desired to provide the basis for understanding and developing intervention strategies. Here we address the role of GIPC1 in the process of MACC1-driven metastasis. MACC1 is a prognostic indicator for patient metastasis formation and metastasis-free survival. MACC1 controls gene transcription, promotes motility, invasion and proliferation of colon cancer cells in vitro, and causes tumor growth and metastasis in mice. Methods By using yeast-two-hybrid assay, mass spectrometry, co-immunoprecipitation and peptide array we analyzed GIPC1 protein binding partners, by using the MACC1 gene promoter and chromatin immunoprecipitation and electrophoretic mobility shift assay we probed for GIPC1 as transcription factor. We employed GIPC1/MACC1-manipulated cell lines for in vitro and in vivo analyses, and we probed the GIPC1/MACC1 impact using human primary colorectal cancer (CRC) tissue. Results We identified MACC1 and its paralogue SH3BP4 as protein binding partners of the protein GIPC1, and we also demonstrated the binding of GIPC1 as transcription factor to the MACC1 promoter (TSS to -60 bp). GIPC1 knockdown reduced endogenous, but not CMV promoter-driven MACC1 expression, and diminished MACC1-induced cell migration and invasion. GIPC1 suppression reduced tumor growth and metastasis in mice intrasplenically transplanted with MACC1-overexpressing CRC cells. In human primary CRC specimens, GIPC1 correlates with MACC1 expression and is of prognostic value for metastasis formation and metastasis-free survival. Combination of MACC1 and GIPC1 expression improved patient survival prognosis, whereas SH3BP4 expression did not show any prognostic value. Conclusions We identified an important, dual function of GIPC1 - as protein interaction partner and as transcription factor of MACC1 - for tumor progression and cancer metastasis.
Collapse
Affiliation(s)
- Franziska Siegel
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Hannes Schmidt
- Department Developmental Neurobiology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Manisha Juneja
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Janice Smith
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Pia Herrmann
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Dennis Kobelt
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - Kamal Sharma
- Department Developmental Neurobiology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Iduna Fichtner
- Experimental Pharmacology and Oncology, GmbH, Berlin, Germany
| | - Wolfgang Walther
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Gunnar Dittmar
- Department Mass Spectrometry, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Rudolf Volkmer
- Institute for Medicinal Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Fritz G. Rathjen
- Department Developmental Neurobiology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | - Ulrike Stein
- Department Translational Oncology of Solid Tumors, Experimental and Clinical Research Institute, Charité Universitätsmedizin Berlin, and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
3
|
Jarak I, Isabel Santos A, Helena Pinto A, Domingues C, Silva I, Melo R, Veiga F, Figueiras A. Colorectal cancer cell exosome and cytoplasmic membrane for homotypic delivery of therapeutic molecules. Int J Pharm 2023; 646:123456. [PMID: 37778515 DOI: 10.1016/j.ijpharm.2023.123456] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023]
Abstract
Colorectal cancer (CRC) is one of the most common causes of death in the world. The multi-drug resistance, especially in metastatic colorectal cancer, drives the development of new strategies that secure a positive outcome and reduce undesirable side effects. Nanotechnology has made an impact in addressing some pharmacokinetic and safety issues related to administration of free therapeutic agents. However, demands of managing complex biointerfacing require equally complex methods for introducing stimuli-responsive or targeting elements. In order to procure a more efficient solution to the overcoming of biological barriers, the physiological functions of cancer cell plasma and exosomal membranes provided the source of highly functionalized coatings. Biomimetic nanovehicles based on colorectal cancer (CRC) membranes imparted enhanced biological compatibility, immune escape and protection to diverse classes of therapeutic molecules. When loaded with therapeutic load or used as a coating for other therapeutic nanovehicles, they provide highly efficient and selective cell targeting and uptake. This review presents a detailed overview of the recent application of homotypic biomimetic nanovehicles in the management of CRC. We also address some of the current possibilities and challenges associated with the CRC membrane biomimetics.
Collapse
Affiliation(s)
- Ivana Jarak
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Porto, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Ana Isabel Santos
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Ana Helena Pinto
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Cátia Domingues
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Coimbra, Portugal; Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal
| | - Inês Silva
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Raquel Melo
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal
| | - Francisco Veiga
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Coimbra, Portugal
| | - Ana Figueiras
- Univ Coimbra, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, Coimbra, Portugal; Univ Coimbra, REQUIMTE/LAQV, Group of Pharmaceutical Technology, Coimbra, Portugal.
| |
Collapse
|
4
|
Tang J, Huang Q, Li X, Gu S. Comprehensive analysis of the oncogenic and immunological role of SPON2 in human tumors. Medicine (Baltimore) 2023; 102:e35122. [PMID: 37713832 PMCID: PMC10508437 DOI: 10.1097/md.0000000000035122] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/17/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Sapiens spondin-2 (SPON2) is a protein found in the extracellular matrix that plays a role in a number of processes, including immune reactions and cell adhesion, and is closely linked to the emergence of a number of tumor types. However, we know very little about Sapiens spondin-2. Therefore, we performed a systematic pan-carcinogenic analysis to explore the relationship between Sapiens spondin-2 and cancers. MATERIALS AND METHODS By comprehensive use of datasets from TCGA, GEO, GTEx, HPA, CPTAC, GEPIA2, TIMER2, cBioPortal, STRING, we adopted bioinformatics methods to dig up the potential carcinogenesis of SPON2, including dissecting the correlation between SPON2 and gene expression, prognosis, gene mutation, Immunohistochemistry staining, immune cell infiltration, and constructed the interaction network of a total of 54 SPON2-binding proteins as well as explored the enrichment analysis of SPON2-related partners. RESULTS The expression of Sapiens spondin-2 in most tumor tissues was higher than that of normal tissues. In addition, SPON2 showed the early diagnostic value in 33 kinds of tumors and was positively or negatively associated with the prognosis of different tumors. It also validates that SPON2 is the gene associated with the majority of immune-infiltrating cells in pan-cancer. High SPON2 expression is associated with tumor progression related pathways. CONCLUSION We found and validated the potential use of SPON2 in cancer detection for the first time through pan-cancer analysis. The expression levels of SPON2 in various tumors were quite different from those in normal tissues. Furthermore, the performance of SPON2 in tumorigenesis and tumor immunity verified our hypothesis. At the same time, it has high specificity and sensitivity in cancer detection. Therefore, SPON2 can be employed as an auxiliary index for the initial diagnosis of tumors and a prognostic marker for various types of tumors.
Collapse
Affiliation(s)
- Jiali Tang
- College of Environment and Public Health, Xiamen Huaxia University, Xiamen, P.R. China
| | - Qing Huang
- College of Environment and Public Health, Xiamen Huaxia University, Xiamen, P.R. China
| | - Xuanwen Li
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, Japan
| | - Shinong Gu
- College of Environment and Public Health, Xiamen Huaxia University, Xiamen, P.R. China
| |
Collapse
|
5
|
Cheng MZ, Yang BB, Zhan ZT, Lin SM, Fang ZP, Gao Y, Zhou WJ. MACC1 and Gasdermin-E (GSDME) regulate the resistance of colorectal cancer cells to irinotecan. Biochem Biophys Res Commun 2023; 671:236-245. [PMID: 37307707 DOI: 10.1016/j.bbrc.2023.06.002] [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: 04/24/2023] [Accepted: 06/01/2023] [Indexed: 06/14/2023]
Abstract
Metastasis-associated in colon cancer 1 (MACC1) is an oncogene associated with the progression and metastasis of many solid cancer entities. High expression of MACC1 is found in colorectal cancer (CRC) tissues. So far, the role of MACC1 in CRC cell pyroptosis and resistance to irinotecan is unclear. The cleavage of Gasdermin-E (GSDME) is the main executors of activated pyroptosis. We found that GSDME enhanced CRC cell pyroptosis and reduced their resistance to irinotecan, while MACC1 inhibited the cleavage of GSDME and CRC cell pyroptosis, promoted CRC cell proliferation, and enhanced the resistance of CRC cells to irinotecan. Therefore, CRC cells with high MACC1 expression and low GSDME expression had higher resistance to irinotecan, while CRC cells with low MACC1 expression and high GSDME expression had lower resistance to irinotecan. Consistently, by analyzing CRC patients who received FOLFIRI (Fluorouracil + Irinotecan + Leucovorin) in combination with chemotherapy in the GEO database, we found that CRC patients with low MACC1 expression and high GSDME expression had higher survival rate. Our study suggests that the expression of MACC1 and GSDME can be used as detection markers to divide CRC patients into irinotecan resistant and sensitive groups, helping to determine the treatment strategy of patients.
Collapse
Affiliation(s)
- Ming-Zhen Cheng
- State Key Laboratory of Organ Failure Research, Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Department of Gastrointestinal and Hernia Surgery, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, 341000, China
| | - Bo-Bo Yang
- State Key Laboratory of Organ Failure Research, Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Department of Gastrointestinal and Hernia Surgery, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, 341000, China
| | - Ze-Tao Zhan
- State Key Laboratory of Organ Failure Research, Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Department of Gastrointestinal and Hernia Surgery, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, 341000, China
| | - Si-Min Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, China
| | - Zhe-Ping Fang
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, Zhejiang, 317000, China
| | - Yi Gao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial, Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wei-Jie Zhou
- State Key Laboratory of Organ Failure Research, Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial, Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China; Department of Gastrointestinal and Hernia Surgery, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi, 341000, China.
| |
Collapse
|
6
|
Hohmann T, Hohmann U, Dehghani F. MACC1-induced migration in tumors: Current state and perspective. Front Oncol 2023; 13:1165676. [PMID: 37051546 PMCID: PMC10084939 DOI: 10.3389/fonc.2023.1165676] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Malignant tumors are still a global, heavy health burden. Many tumor types cannot be treated curatively, underlining the need for new treatment targets. In recent years, metastasis associated in colon cancer 1 (MACC1) was identified as a promising biomarker and drug target, as it is promoting tumor migration, initiation, proliferation, and others in a multitude of solid cancers. Here, we will summarize the current knowledge about MACC1-induced tumor cell migration with a special focus on the cytoskeletal and adhesive systems. In addition, a brief overview of several in vitro models used for the analysis of cell migration is given. In this context, we will point to issues with the currently most prevalent models used to study MACC1-dependent migration. Lastly, open questions about MACC1-dependent effects on tumor cell migration will be addressed.
Collapse
|
7
|
Wu M, Kong D, Zhang Y. SPON2 promotes the bone metastasis of lung adenocarcinoma via activation of the NF-κB signaling pathway. Bone 2023; 167:116630. [PMID: 36427776 DOI: 10.1016/j.bone.2022.116630] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/11/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Bone metastasis severely affects the life span and quality of life of survivors of lung adenocarcinoma (ADC). There is a pressing need to identify viable biomarkers for this incurable, fatal disease. Spondin-2 (SPON2) has been reported to be involved in metastasis and cancer progression, but its role in bone metastasis in patients with lung ADC has rarely been studied. Here, we showed that the upregulation of SPON2 increased the migration, invasion, and epithelial-to-mesenchymal transition of ADC cells in vitro, whereas silencing SPON2 repressed these processes. Consistently, silencing SPON2 significantly reduced the bone metastasis of ADC cells in vivo. Mechanistically, SPON2 activated the nuclear factor-κB (NF-κB) signaling pathway and increased the expression levels of MMP2 and MMP9. Blocking NF-κB using an inhibitor attenuated the SPON2-induced migration and invasion of ADC cells. In addition, we found that SPON2 expression levels were increased in metastatic bone tissues compared to primary ADC tissues. The upregulation of SPON2 was positively correlated with MMP2 and MMP9 expression levels in metastatic bone tissues. In conclusion, these results illustrate that SPON2 plays a key role in ADC by activating the NF-κB pathway to promote bone metastasis, which suggests that it may be a target for future drug development.
Collapse
Affiliation(s)
- Ming Wu
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Shanghai 200135, China
| | - Dewei Kong
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Shanghai 200135, China
| | - Yan Zhang
- Department of Orthopaedics, Gongli Hospital of Pudong New Area, Shanghai 200135, China.
| |
Collapse
|
8
|
Ershov P, Poyarkov S, Konstantinova Y, Veselovsky E, Makarova A. Transcriptomic Signatures in Colorectal Cancer Progression. Curr Mol Med 2023; 23:239-249. [PMID: 35490318 DOI: 10.2174/1566524022666220427102048] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/05/2021] [Accepted: 03/09/2022] [Indexed: 02/08/2023]
Abstract
AIMS Due to a large number of identified hub-genes encoding key molecular regulators, which are involved in signal transduction and metabolic pathways in cancers, it is relevant to systemize and update these findings. BACKGROUND Colorectal cancer (CRC) is the third leading cause of cancer death in the world, with high metastatic potential. Elucidating the pathogenic mechanisms and selection of novel biomarkers in CRC is of great clinical significance. OBJECTIVE This analytical review aims at the systematization of bioinformatics and experimental identification of hub-genes associated with CRC for a more consolidated understanding of common features in networks and pathways in CRC progression as well as hub-genes selection. RESULTS In total, 301 hub-genes were derived from 40 articles. The "core" consisted of 28 hub-genes (CCNB1, LPAR1, BGN, CXCL3, COL1A2, UBE2C, NMU, COL1A1, CXCL2, CXCL11, CDK1, TOP2A, AURKA, SST, CXCL5, MMP3, CCND1, TIMP1, CXCL8, CXCL1, CXCL12, MYC, CCNA2, GCG, GUCA2A, PAICS, PYY and THBS2) mentioned in not less than three articles and having clinical significance in cancerassociated pathways. Of them, there were two discrete clusters enriched in chemokine signaling and cell cycle regulatory genes. High expression levels of BGN and TIMP1 and low expression levels of CCNB1, CXCL3, CXCL2, CXCL2 and PAICS were associated with unfavorable overall survival of patients with CRC. Differently expressed genes such as LPAR1, SST, CXCL12, GUCA2A, and PYY were shown as down regulated, whereas BGN, CXCL3, UBE2C, NMU, CXCL11, CDK1, TOP2A, AURKA, MMP3, CCND1, CXCL1, MYC, CCNA2, PAICS were up regulated genes in CRC. It was also found that MMP3, THBS2, TIMP1 and CXCL12 genes were associated with metastatic CRC. Network analysis in ONCO.IO showed that upstream master regulators RELA, STAT3, SOX2, FOXM1, SMAD3 and NF-kB were connected with "core" hub-genes. Conclusión: Results obtained are of useful fundamental information on revealing the mechanism of pathogenicity, cellular target selection for optimization of therapeutic interventions, as well as transcriptomics prognostic and predictive biomarkers development.
Collapse
Affiliation(s)
- Pavel Ershov
- Department of Analysis and Forecasting of Medical and Biological Health Risks, Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Stanislav Poyarkov
- Department of Analysis and Forecasting of Medical and Biological Health Risks, Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Yulia Konstantinova
- Oncology Department, Federal Research and Clinical Center of Specialized Kinds of Medical Care and Medical Technology of the Federal Medical Biological Agency, Moscow, Russia
| | - Egor Veselovsky
- Department of Analysis and Forecasting of Medical and Biological Health Risks, Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| | - Anna Makarova
- Department of Analysis and Forecasting of Medical and Biological Health Risks, Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russia
| |
Collapse
|
9
|
Rana I, Kataria S, Tan TL, Hajam EY, Kashyap DK, Saha D, Ajnabi J, Paul S, Jayappa S, Ananthan ASHP, Kumar P, Zaarour RF, Haarshaadri J, Kansagara G, Rizvi A, Zirmire RK, Badarinath K, Khedkar SU, Chandra Y, Samuel R, George R, Danda D, Jacob PM, Dey R, Dhandapany PS, He YW, Varga J, Varghese S, Jamora C. Mindin (SPON2) Is Essential for Cutaneous Fibrogenesis in a Mouse Model of Systemic Sclerosis. J Invest Dermatol 2022; 143:699-710.e10. [PMID: 36528128 DOI: 10.1016/j.jid.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 12/16/2022]
Abstract
Systemic sclerosis is a fibrotic disease that initiates in the skin and progresses to internal organs, leading to a poor prognosis. Unraveling the etiology of a chronic, multifactorial disease such as systemic sclerosis has been aided by various animal models that recapitulate certain aspects of the human pathology. We found that the transcription factor SNAI1 is overexpressed in the epidermis of patients with systemic sclerosis, and a transgenic mouse recapitulating this expression pattern is sufficient to induce many clinical features of the human disease. Using this mouse model as a discovery platform, we have uncovered a critical role for the matricellular protein Mindin (SPON2) in fibrogenesis. Mindin is produced by SNAI1 transgenic skin keratinocytes and aids fibrogenesis by inducing early inflammatory cytokine production and collagen secretion in resident dermal fibroblasts. Given the dispensability of Mindin in normal tissue physiology, targeting this protein holds promise as an effective therapy for fibrosis.
Collapse
Affiliation(s)
- Isha Rana
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India; School of Chemical and Biotechnology, Shanmugha Arts, Science, Technology and Research Academy (SASTRA) Deemed University, Thanjavur, India
| | - Sunny Kataria
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India; National Centre for Biological Sciences, Bangalore, India
| | - Tuan Lin Tan
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, San Diego, California, USA; School of Chemical & Life Sciences, Singapore Polytechnic, Singapore, Singapore
| | - Edries Yousaf Hajam
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India; School of Chemical and Biotechnology, Shanmugha Arts, Science, Technology and Research Academy (SASTRA) Deemed University, Thanjavur, India
| | - Deepak Kumar Kashyap
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India; Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Dyuti Saha
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India; Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Johan Ajnabi
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India
| | - Sayan Paul
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Shashank Jayappa
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Akhil S H P Ananthan
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India
| | - Pankaj Kumar
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India
| | - Rania F Zaarour
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India; Thumbay Research Institute for Precision Medicine (TRIPM), Gulf Medical University, Ajman, United Arab Emirates
| | - J Haarshaadri
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India
| | - Gaurav Kansagara
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India; Animal Care and Resource Centre (ACRC), Bangalore Life Science Cluster, Bangalore, India
| | - Abrar Rizvi
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India
| | - Ravindra K Zirmire
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India; School of Chemical and Biotechnology, Shanmugha Arts, Science, Technology and Research Academy (SASTRA) Deemed University, Thanjavur, India
| | - Krithika Badarinath
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India; National Centre for Biological Sciences, Bangalore, India
| | - Sneha Uday Khedkar
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India
| | - Yogesh Chandra
- Animal Care and Resource Centre (ACRC), Bangalore Life Science Cluster, Bangalore, India
| | - Rekha Samuel
- Centre for Stem Cell Research (CSCR), Christian Medical College Vellore, Vellore, India; Department of Pathology, Manipal - Tata Medical College Jamshedpur, Jamshedpur, India
| | - Renu George
- Department of Dermatology, Venereology and Leprosy, Christian Medical College Vellore, Vellore, India
| | - Debashish Danda
- Department of Clinical Immunology & Rheumatology, Christian Medical College Vellore, Vellore, India
| | | | - Rakesh Dey
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India
| | | | - You-Wen He
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - John Varga
- Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Shyni Varghese
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, San Diego, California, USA; Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; Department of Mechanical Engineering & Materials Science, Duke University, Durham, North Carolina, USA; Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Colin Jamora
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science & Regenerative Medicine, Bangalore, India.
| |
Collapse
|
10
|
Zhang X, Luo Y, Cen Y, Qiu X, Li J, Jie M, Yang S, Qin S. MACC1 promotes pancreatic cancer metastasis by interacting with the EMT regulator SNAI1. Cell Death Dis 2022; 13:923. [PMID: 36333284 PMCID: PMC9636131 DOI: 10.1038/s41419-022-05285-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
Metastasis is the dominant cause of cancer-related mortality. Metastasis-associated with colon cancer protein 1 (MACC1) has been proven to play a critical role in cancer metastasis. However, the prometastatic role of MACC1 in regulating the pancreatic cancer (PC) metastatic phenotype remains elusive. Here, we report that MACC1 is highly expressed in The Cancer Genome Atlas (TCGA) and tissue microarray (TMA) and identified as a good indicator for poor prognosis. Overexpression or knockdown of MACC1 in PC cells correspondingly promoted or inhibited pancreatic cancer cell migration and invasion in a MET proto-oncogene receptor tyrosine kinase (MET)-independent manner. Notably, knockdown of MACC1 in PC cells markedly decreased the liver metastatic lesions in a liver metastasis model. Mechanistically, MACC1 binds to the epithelial-mesenchymal transition (EMT) regulator snail family transcriptional repressor 1 (SNAI1) to drive EMT via upregulating the transcriptional activity of SNAI1, leading to the transactivation of fibronectin 1 (FN1) and the trans-repression of cadherin 1 (CDH1). Collectively, our results unveil a new mechanism by which MACC1 drives pancreatic cancer cell metastasis and suggest that the MACC1-SNAI1 complex-mediated mesenchymal transition may be a therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Xianglian Zhang
- grid.412594.f0000 0004 1757 2961Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Ya Luo
- grid.417298.10000 0004 1762 4928Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Yu Cen
- grid.412594.f0000 0004 1757 2961Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Xin Qiu
- grid.412594.f0000 0004 1757 2961Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Jing Li
- grid.417298.10000 0004 1762 4928Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Mengmeng Jie
- grid.417298.10000 0004 1762 4928Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Shiming Yang
- grid.417298.10000 0004 1762 4928Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Shanyu Qin
- grid.412594.f0000 0004 1757 2961Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| |
Collapse
|
11
|
Kim HJ, Moon SJ, Hong S, Won HH, Kim JH. DBC1 is a key positive regulator of enhancer epigenomic writers KMT2D and p300. Nucleic Acids Res 2022; 50:7873-7888. [PMID: 35801925 PMCID: PMC9371912 DOI: 10.1093/nar/gkac585] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/14/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Histone modification is a key epigenetic mechanism for regulation of chromatin dynamics and gene expression. Deleted in breast cancer 1 (DBC1) has been shown to act as a negative regulator of epigenetic modifiers and as a co-activator for nuclear receptors and other transcription factors. However, little is known about the role of DBC1 in the regulation of histone modifications and chromatin landscapes. Here, we analyzed genome-wide profiles of active enhancer and promoter marks in colorectal cancer cells and report DBC1 as a critical positive regulator of histone epigenetic writers KMT2D (H3K4 methyltransferase) and p300 (histone acetyltransferase). DBC1 is required for establishing the landscape of active enhancers, for genome-wide chromatin binding and enhancer recruitment of KMT2D and p300, and for gene activation involved in colorectal cancer progression. DBC1 interacts directly with KMT2D and p300, and enhances KMT2D-mediated histone H3K4 methylation (H3K4me1/2/3) and p300-mediated H3 acetylation. Importantly, DBC1 contributes to super-enhancer formation and function by facilitating the recruitment of KMT2D and p300 and by enhancing their functional interaction and cooperative cross-talk. Our results highlight the critical role of DBC1 as a key positive regulator of KMT2D and p300, and provide insights into regulatory mechanisms underlying the interplay between the enhancer epigenomic writers in enhancer activation.
Collapse
Affiliation(s)
- Hwa Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.,Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Sue Jin Moon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.,Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Sanghoon Hong
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
| | - Hong-Hee Won
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea.,Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.,Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| |
Collapse
|
12
|
Schmid F, Dahlmann M, Röhrich H, Kobelt D, Hoffmann J, Burock S, Walther W, Stein U. Calcium-binding protein S100P is a new target gene of MACC1, drives colorectal cancer metastasis and serves as a prognostic biomarker. Br J Cancer 2022; 127:675-685. [PMID: 35597866 PMCID: PMC9381557 DOI: 10.1038/s41416-022-01833-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 01/05/2023] Open
Abstract
Background The metastasis inducing gene MACC1 is a prognostic and predictive biomarker for metastasis in several cancers. Its mechanism of inducing metastasis includes the transcriptional control of other cancer-related target genes. Here, we investigate the interplay with the metastasis driver S100P in CRC progression. Methods MACC1-dependent S100P expression was analysed by qRT-PCR. The binding of MACC1 to the S100P promoter was determined by ChIP. Alterations in cell proliferation and motility were determined by functional in vitro assays. In vivo metastasis after intrasplenic transplantation was assessed by bioluminescence imaging and evaluation of tumour growth and liver metastasis. The prognostic value of S100P was determined in CRC patients by ROC-based Kaplan–Meier analyses. Results Expression of S100P and MACC1 correlated positively in CRC cells and colorectal tumours. MACC1 was found binding to the S100P promoter and induces its expression. The overexpression of S100P increased proliferation, migration and invasion in vitro and significantly induced liver metastasis in vivo. S100P expression was significantly elevated in metachronously metastasising CRC and was associated with shorter metastasis-free survival. Conclusions We identified S100P as a transcriptional target gene of MACC1. Expression of S100P increases the metastatic potential of CRC cells in vitro and in vivo, and serves as a prognostic biomarker for metastasis-free survival of CRC patients, emphasising novel therapeutic interventions targeting S100P.
Collapse
Affiliation(s)
- Felicitas Schmid
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Experimental and Clinical Research Center of the Charité - Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Mathias Dahlmann
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Experimental and Clinical Research Center of the Charité - Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Hanna Röhrich
- Experimental and Clinical Research Center of the Charité - Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Dennis Kobelt
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Experimental and Clinical Research Center of the Charité - Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Jens Hoffmann
- Experimental Pharmacology and Oncology Berlin-Buch GmbH, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Susen Burock
- Charité Comprehensive Cancer Center, Invalidenstraße 80, 10117, Berlin, Germany
| | - Wolfgang Walther
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Experimental and Clinical Research Center of the Charité - Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Ulrike Stein
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany. .,Experimental and Clinical Research Center of the Charité - Universitätsmedizin Berlin and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany. .,German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
13
|
Bischoff NS, Proquin H, Jetten MJ, Schrooders Y, Jonkhout MCM, Briedé JJ, van Breda SG, Jennen DGJ, Medina-Reyes EI, Delgado-Buenrostro NL, Chirino YI, van Loveren H, de Kok TM. The Effects of the Food Additive Titanium Dioxide (E171) on Tumor Formation and Gene Expression in the Colon of a Transgenic Mouse Model for Colorectal Cancer. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1256. [PMID: 35457963 PMCID: PMC9027218 DOI: 10.3390/nano12081256] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022]
Abstract
Titanium dioxide (TiO2) is present in many different food products as the food additive E171, which is currently scrutinized due to its potential adverse effects, including the stimulation of tumor formation in the gastrointestinal tract. We developed a transgenic mouse model to examine the effects of E171 on colorectal cancer (CRC), using the Cre-LoxP system to create an Apc-gene-knockout model which spontaneously develops colorectal tumors. A pilot study showed that E171 exposed mice developed colorectal adenocarcinomas, which were accompanied by enhanced hyperplasia in epithelial cells, lymphatic nodules at the base of the polyps, and increased tumor size. In the main study, tumor formation was studied following the exposure to 5 mg/kgbw/day of E171 for 9 weeks (Phase I). E171 exposure showed a statistically nonsignificant increase in the number of colorectal tumors in these transgenic mice, as well as a statistically nonsignificant increase in the average number of mice with tumors. Gene expression changes in the colon were analyzed after exposure to 1, 2, and 5 mg/kgbw/day of E171 for 2, 7, 14, and 21 days (Phase II). Whole-genome mRNA analysis revealed the modulation of genes in pathways involved in the regulation of gene expression, cell cycle, post-translational modification, nuclear receptor signaling, and circadian rhythm. The processes associated with these genes might be involved in the enhanced tumor formation and suggest that E171 may contribute to tumor formation and progression by modulation of events related to inflammation, activation of immune responses, cell cycle, and cancer signaling.
Collapse
Affiliation(s)
- Nicolaj S. Bischoff
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Héloïse Proquin
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
- National Institute for Public Health and Environment (RIVM), Bilthoven, 3721 MA De Bilt, The Netherlands
| | - Marlon J. Jetten
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
- Faculty of Health, Medicine and Life Science, Maastricht University Medical Center, 6229 ES Maastricht, The Netherlands
| | - Yannick Schrooders
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
- Laboratory of Biosignaling & Therapeutics, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Marloes C. M. Jonkhout
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
- Laboratory of Biosignaling & Therapeutics, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Jacco J. Briedé
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Simone G. van Breda
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Danyel G. J. Jennen
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Estefany I. Medina-Reyes
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (E.I.M.-R.); (N.L.D.-B.); (Y.I.C.)
| | - Norma L. Delgado-Buenrostro
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (E.I.M.-R.); (N.L.D.-B.); (Y.I.C.)
| | - Yolanda I. Chirino
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (E.I.M.-R.); (N.L.D.-B.); (Y.I.C.)
| | - Henk van Loveren
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| | - Theo M. de Kok
- Department of Toxicogenomics, GROW School for Oncology and Reproduction, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands; (H.P.); (M.J.J.); (Y.S.); (M.C.M.J.); (J.J.B.); (S.G.v.B.); (D.G.J.J.); (H.v.L.); (T.M.d.K.)
| |
Collapse
|
14
|
Inhibition of MACC1-Induced Metastasis in Esophageal and Gastric Adenocarcinomas. Cancers (Basel) 2022; 14:cancers14071773. [PMID: 35406545 PMCID: PMC8997092 DOI: 10.3390/cancers14071773] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Esophageal and Gastric Adenocarcinomas (AGE/S) are characterized by early metastasis and poor survival. MACC1 (Metastasis Associated in Colon Cancer 1) acts in colon cancer as a metastasis inducer and is linked to reduced survival. In this study, we analyzed the prognostic role of MACC1 in a large AGE/S cohort and the potential of MACC1 inhibition in vitro and in vivo. MACC1 is an independent negative prognostic marker in our cohort. In vitro, migration was enhanced by MACC1 in overexpressing cells. This MACC1-related effect could be inhibited by using selumetinib in vitro. In vivo, MACC1 induced faster and larger metastasis development, which could be inhibited by selumetinib. In conclusion, MACC1 is a strong negative prognostic factor in AGE/S and is a potential target for therapy with selumetinib. Abstract Esophageal and Gastric Adenocarcinomas (AGE/S) are characterized by early metastasis and poor survival. MACC1 (Metastasis Associated in Colon Cancer 1) acts in colon cancer as a metastasis inducer and is linked to reduced survival. This project illuminates the role and potential for the inhibition of MACC1 in AGE/S. Using 266 of 360 TMAs and survival data of AGE/S patients, we confirm the value of MACC1 as an independent negative prognostic marker in AGE/S patients. MACC1 gene expression is correlated with survival and morphological characteristics. In vitro analysis of lentivirally MACC1-manipulated subclones of FLO-1 and OE33 showed enhanced migration induced by MACC1 in both cell line models, which could be inhibited by the MEK1 inhibitor selumetinib. In vivo, the efficacy of selumetinib on tumor growths and metastases of MACC1-overexpressing FLO-1 cells xenografted intrasplenically in NOG mice was tested. Mice with high-MACC1-expressing cells developed faster and larger distant metastases. Treatment with selumetinib led to a significant reduction in metastasis exclusively in the MACC1-positive xenografts. MACC1 is an enhancer of tumor aggressiveness and a predictor of poor survival in AGE/S. This effect can be inhibited by selumetinib.
Collapse
|
15
|
Insulin-like Growth Factor Binding Protein-2 (IGFBP2) Is a Key Molecule in the MACC1-Mediated Platelet Communication and Metastasis of Colorectal Cancer Cells. Int J Mol Sci 2021; 22:ijms222212195. [PMID: 34830078 PMCID: PMC8624049 DOI: 10.3390/ijms222212195] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 11/16/2022] Open
Abstract
Tumor cell crosstalk with platelets and, subsequently, their activation are key steps in hematogenous tumor metastasis. MACC1 is an oncogene involved in molecular pathogenesis of colorectal cancer (CRC) and other solid tumor entities, mediating motility and metastasis, making MACC1 an accepted prognostic biomarker. However, the impact of MACC1 on platelet activation has not yet been addressed. Here, we investigated the activation of platelets by human CRC cells upon MACC1 modulation, indicated by platelet aggregation and granule release. These approaches led to the identification of insulin-like growth factor binding protein-2 (IGFBP2) as a functional downstream molecule of MACC1, affecting communication with platelets. This was confirmed by an shRNA-mediated IGFBP2 knockdown, while maintaining MACC1 activity. Although IGFBP2 displayed an attenuated platelet activation potential, obviously by scavenging IGF-I as a platelet costimulatory mediator, the MACC1/IGFBP2 axis did not affect the thrombin formation potential of the cells. Furthermore, the IGFBP2/MACC1-driven cell migration and invasiveness was further accelerated by platelets. The key role of IGFBP2 for the metastatic spread in vivo was confirmed in a xenograft mouse model. Data provide evidence for IGFBP2 as a downstream functional component of MACC1-driven metastasis, linking these two accepted oncogenic biomarkers for the first time in a platelet context.
Collapse
|
16
|
Huang C, Ou R, Chen X, Zhang Y, Li J, Liang Y, Zhu X, Liu L, Li M, Lin D, Qiu J, Liu G, Zhang L, Wu Y, Tang H, Liu Y, Liang L, Ding Y, Liao W. Tumor cell-derived SPON2 promotes M2-polarized tumor-associated macrophage infiltration and cancer progression by activating PYK2 in CRC. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:304. [PMID: 34583750 PMCID: PMC8477524 DOI: 10.1186/s13046-021-02108-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023]
Abstract
Background Tumor-associated macrophages (TAMs) are key regulators of the complex interplay between cancer and the immune microenvironment. Tumor cell-derived spondin 2 (SPON2) is an extracellular matrix glycoprotein that has complicated roles in recruitment of macrophages and neutrophils during inflammation. Overexpression of SPON2 has been shown to promote tumor cell migration in colorectal cancer (CRC). However, the mechanism by which SPON2 regulates the accumulation of TAMs in the tumor microenvironment (TME) of CRC is unknown. Methods Immunohistochemistry was used to examine SPON2 expression in clinical CRC tissues. In vitro migration assays, transendothelial migration assays (iTEM), and cell adhesion assays were used to investigate the effects of SPON2 on monocyte/macrophage migration. Subcutaneous tumor formation and orthotopic implantation assays were performed in C57 BL/6 mice to confirm the effects of SPON2 on TAM infiltration in tumors. Results SPON2 expression is positively correlated with M2-TAM infiltration in clinical CRC tumors and poor prognosis of CRC patients. In addition, SPON2 promotes cytoskeletal remodeling and transendothelial migration of monocytes by activating integrin β1/PYK2 axis. SPON2 may indirectly induce M2-polarization through upregulating cytokines including IL10, CCL2 and CSF1 expression in tumor cells. Blocking M2 polarization and Macrophage depletion inhibited the SPON2-induced tumors growth and invasion. Furthermore, blocking the SPON2/integrin β1/PYK2 axis impairs the transendothelial migration of monocytes and cancer-promoting functions of TAMs in vivo. Conclusions Our findings demonstrate that SPON2-driven M2-TAM infiltration plays an important role during CRC tumor growth and metastasis. SPON2 may be a valuable biomarker guiding the use of macrophage-targeting strategies and a potential therapeutic target in advanced CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02108-0.
Collapse
Affiliation(s)
- Chengmei Huang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Ruizhang Ou
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Xiaoning Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Yaxin Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Jiexi Li
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yihao Liang
- Department of Orthopedist, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Xiaohui Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Lei Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Mingzhou Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Dagui Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Junfeng Qiu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Guanglong Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Lingjie Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Yuanyuan Wu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Huiyi Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yanmin Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China. .,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China.
| | - Wenting Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China. .,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China. .,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
17
|
Kobelt D, Perez-Hernandez D, Fleuter C, Dahlmann M, Zincke F, Smith J, Migotti R, Popp O, Burock S, Walther W, Dittmar G, Mertins P, Stein U. The newly identified MEK1 tyrosine phosphorylation target MACC1 is druggable by approved MEK1 inhibitors to restrict colorectal cancer metastasis. Oncogene 2021; 40:5286-5301. [PMID: 34247190 PMCID: PMC8390371 DOI: 10.1038/s41388-021-01917-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023]
Abstract
Cancer metastasis causes >90% of cancer deaths and remains a major treatment challenge. Here we deciphered the impact of tyrosine phosphorylation of MACC1, a causative driver for cancer metastasis, for cancer cell signaling and novel interventions to restrict cancer metastasis. We identified MACC1 as new MEK1 substrate. MEK1 directly phosphorylates MACC1, leading to accelerated and increased ERK1 activation. Mutating in silico predicted hierarchical MACC1 tyrosine phosphorylation sites abrogates MACC1-induced migration, invasion, and MET expression, a transcriptional MACC1 target. Targeting MEK1 by RNAi or clinically applicable MEK1 inhibitors AZD6244 and GSK1120212 reduces MACC1 tyrosine phosphorylation and restricts MACC1-induced metastasis formation in mice. Although MEK1 levels, contrary to MACC1, are not of prognostic relevance for CRC patients, MEK1 expression was found indispensable for MACC1-induced metastasis. This study identifies MACC1 as new MEK1 substrate for tyrosine phosphorylation decisively impacting cell motility, tumor growth, and metastasis. Thus, MAP kinase signaling is not linear leading to ERK activation, but branches at the level of MEK1. This fundamental finding opens new therapeutic options for targeting the MEK1/MACC1 axis as novel vulnerability in patients at high risk for metastasis. This might be extended from CRC to further solid tumor entities.
Collapse
Affiliation(s)
- Dennis Kobelt
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Daniel Perez-Hernandez
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Proteome and Genome Research Laboratory, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Claudia Fleuter
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mathias Dahlmann
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Fabian Zincke
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Janice Smith
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Rebekka Migotti
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Oliver Popp
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Susen Burock
- Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Walther
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Gunnar Dittmar
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Proteome and Genome Research Laboratory, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Philipp Mertins
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrike Stein
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
18
|
Wang P, Yao L, Luo M, Zhou W, Jin X, Xu Z, Yan S, Li Y, Xu C, Cheng R, Huang Y, Lin X, Ma K, Cao H, Liu H, Xue G, Han F, Nie H, Jiang Q. Single-cell transcriptome and TCR profiling reveal activated and expanded T cell populations in Parkinson's disease. Cell Discov 2021; 7:52. [PMID: 34282123 PMCID: PMC8289849 DOI: 10.1038/s41421-021-00280-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023] Open
Abstract
Given the chronic inflammatory nature of Parkinson's disease (PD), T cell immunity may be important for disease onset. Here, we performed single-cell transcriptome and TCR sequencing, and conducted integrative analyses to decode composition, function and lineage relationship of T cells in the blood and cerebrospinal fluid of PD. Combined expression and TCR-based lineage tracking, we discovered a large population of CD8+ T cells showing continuous progression from central memory to terminal effector T cells in PD patients. Additionally, we identified a group of cytotoxic CD4+ T cells (CD4 CTLs) remarkably expanded in PD patients, which derived from Th1 cells by TCR-based fate decision. Finally, we screened putative TCR-antigen pairs that existed in both blood and cerebrospinal fluid of PD patients to provide potential evidence for peripheral T cells to participate in neuronal degeneration. Our study provides valuable insights and rich resources for understanding the adaptive immune response in PD.
Collapse
Affiliation(s)
- Pingping Wang
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Lifen Yao
- grid.412596.d0000 0004 1797 9737Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang China
| | - Meng Luo
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Wenyang Zhou
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Xiyun Jin
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Zhaochun Xu
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Shi Yan
- grid.412596.d0000 0004 1797 9737Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang China
| | - Yiqun Li
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Chang Xu
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Rui Cheng
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Yan Huang
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Xiaoyu Lin
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Kexin Ma
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Huimin Cao
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Hongxin Liu
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Guangfu Xue
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Fang Han
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Huan Nie
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Qinghua Jiang
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Big Data (Harbin Institute of Technology), Ministry of Education, Harbin, Heilongjiang China
| |
Collapse
|
19
|
Jiang H, Guo W, Huang K, Jiang H, Zhang R, Hu H, Lin X, Wang S. Screening of radiotracer for diagnosis of colorectal cancer liver metastasis based on MACC1-SPON2. Abdom Radiol (NY) 2021; 46:3227-3237. [PMID: 33712897 PMCID: PMC8215036 DOI: 10.1007/s00261-021-03015-w] [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: 08/04/2020] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 12/09/2022]
Abstract
Background Metastasis-associated in colon cancer 1 (MACC1) and Spondin2 (SPON2) are newly discovered oncogenes, but little is known about their role in colorectal cancer(CRC) liver metastases. PET has become an important molecular imaging technology due to its high sensitivity and quantifiability. In particular, its targeted, specific molecular probes can detect biological behaviors. This study was designed to evaluate the different biological properties of 18F-FDG, 18F-FLT, and 18F-FMISO PET. The value of the CRC liver metastasis model explores the correlation and potential mechanisms of three tracers uptakes with tumor-related biological characteristics. Methods Human CRC cell lines(LoVo and HCT8), were cultured for in vitro radionuclide uptake experiments to compare the molecular imaging features of colorectal cancer cells with different metastatic potentials. Two kinds of cells were injected into the spleen of nude mice to establish a liver metastasis model. After the tumor formation, three kinds of tracer PET images were performed to evaluate the characteristics of live PET imaging of high and low liver metastasis colorectal cancer models. The expression levels of MACC1 and SPON2 in tissues were detected by immunohistochemistry and Western blot. Correlation between tracer uptake and expression of MACC1 and SPON2 in liver metastases was assessed by linear regression analysis. Results The uptake rate of in vitro three tracers uptake experiments was LoVo > HCT8. Micro-PET scan showed no significant difference between the 18F-FDG SUV values of the two cells (P > 0.05); there was significant difference between the 18F-FLT and 18F-FMISO SUV values (P < 0.05). All in vivo FLT and FMISO SUV values were significantly higher in LoVo tumors than in HCT8 tumors. The results of Western blot and immunohistochemistry showed that the expression levels of MACC1 and SPON2 in LoVo liver metastasis were higher than those in HCT8 (P < 0.05). The 18F-FLT SUVmax ratio was significantly correlated with the expression of MACC1 and SPON2 in hepatic metastases (r = 0.737, P = 0.0026; r = 0.842, P = 0.0002). The 18F-FMISO SUVmax ratio was only significantly correlated with the expression of MACC1 in hepatic metastasis (r = 0.770, P = 0.0013). Conclusions Early screening with 18F-FLT and 18F-FMISO tracers has important clinical value for the efficient diagnosis and treatment of colorectal cancer liver metastases.
Collapse
|
20
|
Zhou P, Xiang CX, Wei JF. The clinical significance of spondin 2 eccentric expression in peripheral blood mononuclear cells in bronchial asthma. J Clin Lab Anal 2021; 35:e23764. [PMID: 33998076 PMCID: PMC8183911 DOI: 10.1002/jcla.23764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background Bronchial asthma (BA) was a heterogeneous disease characterized by chronic airway inflammation. Spondin 2 (SPON2) was reported to be implicated in the integrin pathway, protein metabolism, and drug‐induced lupus erythematosus. The purpose of this study was to evaluate the significance of SPON2 in BA diagnosis and treatment. Methods Peripheral blood samples were obtained from 137 BA pediatric patients (61 mild‐to‐moderate BA and 76 severe BA) and 59 healthy children. Subject's information, clinical indexes, pulmonary ventilation functions were recorded in the two groups. Peripheral blood mononuclear cells (PBMCs) were isolated from patients’ samples. qRT‐PCR and ELISA assays were employed to examine the levels of SPON2 and inflammatory cytokines, respectively. Pearson's correlation analysis confirmed the association between SPON2 and inflammatory cytokines. Receiver operating characteristic (ROC) analysis was used to evaluate the potentials of SPON2 in terms of BA detection and discriminating against the severity of BA. Results Bioinformatics analysis showed that SPON2, OLFM4, XIST, and TSIX were significantly upregulated, while KDM5D and RPS4Y1 were reduced in BA. GO analysis verified that these six genes were mainly involved in neutrophil degranulation, neutrophil activation involved in immune response, neutrophil activation, and neutrophil‐mediated immunity. After isolating PBMCs, we found that SPON2 was remarkably increased in BA pediatric group compared with healthy children, and the relative levels of SPON2 were related to the severity of BA. The receiver operating characteristic (ROC) analysis revealed the high potentials of SPON2 in BA diagnosis (AUC was 0.8080) and severity distinctions (AUCs were 0.7341 and 0.8541, respectively). Also, we found that there were significant differences in fractional exhaled nitric oxide (FeNO), forced expiratory volume in 1 s (FEV1)%, FEV1/ forced vital capacity (FVC)%, immunoglobulin E (IgE), serum eosinophils, and serum neutrophils between mild‐to‐moderate BA group and severe BA group. Finally, SPON2 was negatively correlated with IL‐12 while positively associated with IL‐4, IL‐13, and IL‐17A. Conclusions SPON2 was a viable biomarker for diagnosing and degree of severity in BA, providing more insight into exploring BA and treatment's pathogenesis.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Pediatric, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shengzhou, China
| | - Cai-Xia Xiang
- Department of Pediatric, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University Shengzhou Branch), Shengzhou, China
| | - Jin-Feng Wei
- Department of Respiratory, Hangzhou Children's Hospital, Hangzhou, China
| |
Collapse
|
21
|
Zhang W, Yang H, Wang Z, Wu Y, Wang J, Duan G, Guo Q, Zhang Y. miR-320a/SP1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer. Cancer Cell Int 2021; 21:175. [PMID: 33731131 PMCID: PMC7972346 DOI: 10.1186/s12935-021-01874-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Transcription factors (TFs) may be engaged in reciprocal regulatory circuits with certain miRNAs to maintain cellular homeostasis. Disequilibrium of the reciprocities by certain tumor-related stimuli may give rise to deregulation of downstream cellular signaling pathways, thus promoting malignant tumor phenotypes. Specificity Protein 1 (SP1) is the most representative member of the tumor-related transcription factors. Previous studies disclosed that SP1 can transcriptionally regulate miRNAs and coding genes to facilitate tumor progression. In our study, we used bioinformatic analysis to predict several SP1-binding sites within the miR-320a promoter and found that SP1 is a predicted target gene of miR-320a. Therefore, we hypothesize a reciprocal regulatory link between SP1 and miR-320a that participates in colorectal cancer (CRC) development METHODS: We performed bioinformatic analysis, quantitative polymerase chain reaction (qPCR), immunoblotting, dual-luciferase reporter assays, and a series of in vitro and in vivo functional assays to describe a novel SP1/miR-320a reciprocal interaction in CRC RESULTS: First, we found that miR-320a was significantly downregulated in CRC tissues and cell lines. Consistent with findings in other cancers, miR-320a exhibited inhibitory effects on cell growth and invasion of CRC in vitro and in vivo. Moreover, we identified SP1 as a target gene of miR-320a, and ectopic SP1 expression partly abolished miR-320a-induced inhibitory effects. Conversely, we confirmed that SP1 interacts with the miR-320a promoter, leading to depression of miR-320a. This illustrates a double-negative feedback loop between miR-320a and SP1. Additionally, based on the fact that SP1 promotes MACC1 transcription, we determined via immunoblotting that the oncogenic MACC1/MET signaling pathway was inactivated in the context of miR-320a-induced SP1 downregulation CONCLUSION: Taken together, our study is the first to describe a miR-320a/SP1 negative reciprocal interaction, which contributes to cell growth and invasion in CRC through modulation of the MACC1/MET signaling pathway.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Medical Oncology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China.,Medical School, Kunming University of Science and Technology, Kunming, 650000, China
| | - Haitao Yang
- Medical School, Kunming University of Science and Technology, Kunming, 650000, China
| | - Zhongqiu Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yanting Wu
- Medical School, Kunming University of Science and Technology, Kunming, 650000, China
| | - Jingzhai Wang
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China.,Yunnan Provincial Clinical Medicine Center of Gastrointestinal Endoscopy, Kunming, 650000, China
| | - Guihua Duan
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China.,Yunnan Provincial Clinical Medicine Center of Gastrointestinal Endoscopy, Kunming, 650000, China
| | - Qiang Guo
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China. .,Yunnan Provincial Clinical Medicine Center of Gastrointestinal Endoscopy, Kunming, 650000, China.
| | - Yu Zhang
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, China. .,Yunnan Provincial Clinical Medicine Center of Gastrointestinal Endoscopy, Kunming, 650000, China.
| |
Collapse
|
22
|
Xiang Y, Liang B, Jiang Y, Sun F, Zhao Y, Wu Q, Hu X, Liu Y, Huang Q, Liao W, Yao Z, Li S, Shi M. MET transcriptional regulator/serine peptidase inhibitor kunitz type 1 panel operating through HGF/c-MET axis as a prognostic signature in pan-cancer. Cancer Med 2021; 10:2442-2460. [PMID: 33751856 PMCID: PMC7982633 DOI: 10.1002/cam4.3834] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 01/24/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022] Open
Abstract
Dysregulations in transcription factors (TFs) and their genetic products play important roles in tumorigenesis, tumor progression and metastasis. However, prognostic value of the transcriptional regulatory networks in different cancers has not been investigated in depth. The purpose of our study was to identify and validate a potential predictive signature that combines TFs and their regulatory products in eight solid tumors. We used bioinformatics analysis to identify MET Transcriptional Regulator (MACC1) and Serine Peptidase Inhibitor Kunitz Type 1 (SPINT1) as candidate TFs with the respective downstream regulatory proteins for patient prognosis in pan‐cancer. Subsequent molecular analysis of clinical gastric cancer tissue samples further verified the negative correlation between MACC1 and SPINT1. Further, we showed that mechanistically, MACC1/SPINT1 mediated the pro‐HGF proteolysis and c‐Met phosphorylation in HGF/c‐MET signaling pathway. Kaplan‐Meier plots and receiver operating characteristics analysis revealed that the two‐gene signature combining MACC1 with SPINT1 was effective in predicting survival in all eight cancer cohorts tested. In conclusion, our study clarified the regulatory relationship between MACC1 and SPINT1 in the context of the HGF/c‐MET signaling pathway and determined MACC1/SPINT1 panel as a valuable signature for the prediction of prognosis in patients for multiple solid cancer types.
Collapse
Affiliation(s)
- Yi Xiang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bishan Liang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Jiang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fei Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yang Zhao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qijing Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xingbin Hu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yajing Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiong Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiqi Yao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaowei Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
23
|
Zhang M, Yang J, Jiang H, Jiang H, Wang Z. Correlation between glucose metabolism parameters derived from FDG and tumor TNM stages and metastasis-associated proteins in colorectal carcinoma patients. BMC Cancer 2021; 21:258. [PMID: 33750337 PMCID: PMC7941722 DOI: 10.1186/s12885-021-07944-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 02/21/2021] [Indexed: 01/04/2023] Open
Abstract
Background The aim of this study was to investigate the relationship between multiple metabolism parameters derived from FDG and tumor TNM stages as well as tumor metastasis-associated protein of GLUT-1 and MACC1 in colorectal carcinoma (CRC). Methods Thirty-eight patients (24 males and 14 females) with primary CRC confirmed by elective surgery pathological, who also accepted 18F-FDG PET/CT scans during 2017 to 2019 were included in this study. The tumor classification of T, N and M is explained by the 7th American Joint Committee on Cancer (AJCC). 18F-FDG parameters of SUVmax, SUVmean, TLG and MTV were measured by drawing a region of interest on the primary lesions. The expression of GLUT-1 and MACC1 was quantified by immunohistochemical, and the correlation between metabolism parameters and tumor biomarkers were analyzed. Results According to our analysis, the 18F-FDG parameters of SUVmean was significantly correlated with tumor M status (P = 0.000) of primary CRC. The primary tumor lesion with higher SUVmax, TLG and MTV values prone to a high-T status (P = 0.002, 0.002 and 0.001, respectively). The high expression of GLUT-1/MACC1 weas more frequently involved with T3–4 stage and was poorly differentiated in CRC patients. Multivariate analysis found that the expression of GLUT-1 protein was correlated with SUVmax and MTV (R2 = 0.42, P = 0.013 and 0.004, respectively), moreover, the expression of MACC1 protein was correlated with TLG (R2 = 0.372, P = 0.000). Conclusion Glucose metabolism parameters derived from FDG provides a noninvasive assessment of M status and T status in CRC patients. The expression of GLUT-1 and MACC1 was associated with 18F-FDG uptake in CRC patients.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, No.95 Yongan Road, Xicheng District, Beijing, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, No.95 Yongan Road, Xicheng District, Beijing, China
| | - Hao Jiang
- Department of Radiology, Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, Heilongjiang Province, China
| | - Huijie Jiang
- Department of Radiology, Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, Heilongjiang Province, China.
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, China.
| |
Collapse
|
24
|
Pucci M, Gomes Ferreira I, Malagolini N, Ferracin M, Dall’Olio F. The Sd a Synthase B4GALNT2 Reduces Malignancy and Stemness in Colon Cancer Cell Lines Independently of Sialyl Lewis X Inhibition. Int J Mol Sci 2020; 21:ijms21186558. [PMID: 32911675 PMCID: PMC7555213 DOI: 10.3390/ijms21186558] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background: The Sda antigen and its biosynthetic enzyme B4GALNT2 are highly expressed in healthy colon but undergo a variable down-regulation in colon cancer. The biosynthesis of the malignancy-associated sialyl Lewis x (sLex) antigen in normal and cancerous colon is mediated by fucosyltransferase 6 (FUT6) and is mutually exclusive from that of Sda. It is thought that the reduced malignancy associated with high B4GALNT2 was due to sLex inhibition. Methods: We transfected the cell lines SW480 and SW620, derived respectively from a primary tumor and a metastasis of the same patient, with the cDNAs of FUT6 or B4GALNT2, generating cell variants expressing either the sLex or the Sda antigens. Transfectants were analyzed for growth in poor adherence, wound healing, stemness and gene expression profile. Results: B4GALNT2/Sda expression down-regulated all malignancy-associated phenotypes in SW620 but only those associated with stemness in SW480. FUT6/sLex enhanced some malignancy-associated phenotypes in SW620, but had little effect in SW480. The impact on the transcriptome was stronger for FUT6 than for B4GALNT2 and only partially overlapping between SW480 and SW620. Conclusions: B4GALNT2/Sda inhibits the stemness-associated malignant phenotype, independently of sLex inhibition. The impact of glycosyltransferases on the phenotype and the transcriptome is highly cell-line specific.
Collapse
|
25
|
Overexpression of Spondin-2 Is Associated with Recurrence-Free Survival in Patients with Localized Clear Cell Renal Cell Carcinoma. DISEASE MARKERS 2020; 2020:5074239. [PMID: 32952742 PMCID: PMC7487092 DOI: 10.1155/2020/5074239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/08/2020] [Accepted: 08/21/2020] [Indexed: 02/05/2023]
Abstract
Background The spondin-2 (SPON2) gene is overexpressed in multiple malignant tumors and may promote tumor aggressiveness. However, its expression profile and functional roles in clear cell renal cell carcinoma (ccRCC) are still unclear. Methods SPON2 expression in ccRCC was evaluated using expression data from TCGA and GEO databases, then confirmed by local patient population (94 patients). The clinical significance of SPON2 expression was evaluated. Downregulation of SPON2 was performed using small-interfering RNA (siRNA). The effects of SPON2 silencing on cell proliferation, apoptosis, invasion, and migration in vitro were investigated. Results SPON2 was overexpressed in the majority of the ccRCC at both mRNA and protein levels. SPON2 expression was significantly correlated with stage, grade, and recurrence (all P < 0.05) in patients with localized ccRCC. The receiver operating characteristic (ROC) curve showed that SPON2 expression could serve as a predictor of recurrence. SPON2 expression was significantly associated with recurrence-free survival (RFS) in patients with localized ccRCC. Knocking down SPON2 resulted in suppressed cell invasion and migration in vitro. Conclusion SPON2 expression might function as a prognostic biomarker in patients with localized ccRCC.
Collapse
|
26
|
Screening and identification of potential prognostic biomarkers in bladder urothelial carcinoma: Evidence from bioinformatics analysis. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
27
|
Li N, Liu S, Zhang Y, Yu L, Hu Y, Wu T, Fang M, Xu Y. Transcriptional Activation of Matricellular Protein Spondin2 (SPON2) by BRG1 in Vascular Endothelial Cells Promotes Macrophage Chemotaxis. Front Cell Dev Biol 2020; 8:794. [PMID: 32974343 PMCID: PMC7461951 DOI: 10.3389/fcell.2020.00794] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
The matricellular protein SPON2 plays diverse roles in the development of cardiovascular diseases. SPON2 is expressed in endothelial cells, but its transcription regulation in the context of atherogenesis remains incompletely appreciated. Here we report that SPON2 expression was up-regulated by pro-atherogenic stimuli (oxLDL and TNF-α) in vascular endothelia cells. In addition, endothelial SPON2 was elevated in Apoe–/– mice fed on a Western diet compared to the control mice. Induction of SPON2 in endothelial cells by pro-atherogenic stimuli was mediated by BRG1, a chromatin remodeling protein, both in vitro and in vivo. Further analysis revealed that BRG1 interacted with the sequence-specific transcription factor Egr-1 to activate SPON2 transcription. BRG1 contributed to SPON2 trans-activation by modulating chromatin structure surrounding the SPON2 promoter. Functionally, activation of SPON2 transcription by the Egr-1/BRG1 complex provided chemoattractive cues for macrophage trafficking. SPON2 depletion abrogated the ability of BRG1 or Egr-1 to stimulate endothelial derived chemoattractive cue for macrophage migration. On the contrary, recombinant SPON2 rescued endothelial chemo-attractability in the absence of BRG1 or Egr-1. In conclusion, our data have identified a novel transcriptional cascade in endothelial cells that may potentially promote macrophage recruitment and vascular inflammation leading to atherogenesis.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Shuai Liu
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research and Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou, China.,Department of Cardiology, Kaifeng People's Hospital, Kaifeng, China
| | - Yuanyuan Zhang
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Liming Yu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yanjiang Hu
- Department of Cardiothoracic Surgery, Liyang People's Hospital, Liyang, China
| | - Teng Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Mingming Fang
- Department of Clinical Medicine and Laboratory Center for Experimental Medicine, Jiangsu Health Vocational Institute, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
28
|
Cheng XS, Huo YN, Fan YY, Xiao CX, Ouyang XM, Liang LY, Lin Y, Wu JF, Ren JL, Guleng B. Mindin serves as a tumour suppressor gene during colon cancer progression through MAPK/ERK signalling pathway in mice. J Cell Mol Med 2020; 24:8391-8404. [PMID: 32614521 PMCID: PMC7412704 DOI: 10.1111/jcmm.15332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 01/14/2020] [Accepted: 02/06/2020] [Indexed: 12/13/2022] Open
Abstract
Mindin is important in broad spectrum of immune responses. On the other hand, we previously reported that mindin attenuated human colon cancer development by blocking angiogenesis through Egr-1-mediated regulation. However, the mice original mindin directly suppressed the syngenic colorectal cancer (CRC) growth in our recent study and we aimed to further define the role of mindin during CRC development in mice. We established the mouse syngeneic CRC CMT93 and CT26 WT cell lines with stable mindin knock-down or overexpression. These cells were also subcutaneously injected into C57BL/6 and BALB/c mice as well as established a colitis-associated colorectal cancer (CAC) mouse model treated with lentiviral-based overexpression and knocked-down of mindin. Furthermore, we generated mindin knockout mice using a CRISPR-Cas9 system with CAC model. Our data showed that overexpression of mindin suppressed cell proliferation in both of CMT93 and CT26 WT colon cancer cell lines, while the silencing of mindin promoted in vitro cell proliferation via the ERK and c-Fos pathways and cell cycle control. Moreover, the overexpression of mindin significantly suppressed in vivo tumour growth in both the subcutaneous transplantation and the AOM/DSS-induced CAC models. Consistently, the silencing of mindin reversed these in vivo observations. Expectedly, the tumour growth was promoted in the CAC model on mindin-deficient mice. Thus, mindin plays a direct tumour suppressive function during colon cancer progression and suggesting that mindin might be exploited as a therapeutic target for CRC.
Collapse
Affiliation(s)
- Xiao-Shen Cheng
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Ya-Ni Huo
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Yan-Yun Fan
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Chuan-Xing Xiao
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Xiao-Mei Ouyang
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Lai-Ying Liang
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Ying Lin
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Jian-Feng Wu
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Jian-Lin Ren
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Bayasi Guleng
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China.,School of Medicine, Cancer Research Center & Institute of Microbial Ecology, Xiamen University, Xiamen, China.,State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, China
| |
Collapse
|
29
|
Kang HG, Kim WJ, Noh MG, Chun KH, Kim SJ. SPON2 Is Upregulated through Notch Signaling Pathway and Promotes Tumor Progression in Gastric Cancer. Cancers (Basel) 2020; 12:cancers12061439. [PMID: 32492954 PMCID: PMC7352369 DOI: 10.3390/cancers12061439] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 05/30/2020] [Indexed: 02/06/2023] Open
Abstract
Spondin-2 (SPON2) is involved in cancer progression and metastasis of many tumors; however, its role and underlying mechanism in gastric cancer are still obscure. In this study, we investigated the role of SPON2 and related signaling pathway in gastric cancer progression and metastasis. SPON2 expression levels were found to be upregulated in gastric cancer cell lines and patient tissues compared to normal gastric epithelial cells and normal controls. Furthermore, SPON2 silencing was observed to decrease cell proliferation and motility and reduce tumor growth in xenograft mice. Conversely, SPON2 overexpression was found to increase cell proliferation and motility. Subsequently, we focused on regulatory mechanism of SPON2 in gastric cancer. cDNA microarray and in vitro study showed that Notch signaling is significantly correlated to SPON2 expression. Therefore, we confirmed how Notch signaling pathway regulate SPON2 expression using Notch signaling-related transcription factor interaction and reporter gene assay. Additionally, activation of Notch signaling was observed to increase cell proliferation, migration, and invasion through SPON2 expression. Our study demonstrated that Notch signaling-mediated SPON2 upregulation is associated with aggressive progression of gastric cancer. In conclusion, we suggest upregulated SPON2 via Notch signaling as a potential target gene to inhibit gastric cancer progression.
Collapse
Affiliation(s)
- Hyeon-Gu Kang
- Department of Biomedical Science, Department of Life Science & BK21-Plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea; (H.-G.K.); (W.-J.K.)
| | - Won-Jin Kim
- Department of Biomedical Science, Department of Life Science & BK21-Plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea; (H.-G.K.); (W.-J.K.)
| | - Myung-Giun Noh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-gu, Gwangju 61005, Korea;
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
- Correspondence: (K.-H.C.); (S.-J.K.); Tel.: +82-2-2228-1699 (K.-H.C.); +82-62-230-6664 (S.-J.K.); Fax: +82-2-312-5041 (K.-H.C.); +82-62-234-4326 (S.-J.K.)
| | - Seok-Jun Kim
- Department of Biomedical Science, Department of Life Science & BK21-Plus Research Team for Bioactive Control Technology, College of Natural Sciences, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea; (H.-G.K.); (W.-J.K.)
- Correspondence: (K.-H.C.); (S.-J.K.); Tel.: +82-2-2228-1699 (K.-H.C.); +82-62-230-6664 (S.-J.K.); Fax: +82-2-312-5041 (K.-H.C.); +82-62-234-4326 (S.-J.K.)
| |
Collapse
|
30
|
Andreuzzi E, Capuano A, Poletto E, Pivetta E, Fejza A, Favero A, Doliana R, Cannizzaro R, Spessotto P, Mongiat M. Role of Extracellular Matrix in Gastrointestinal Cancer-Associated Angiogenesis. Int J Mol Sci 2020; 21:E3686. [PMID: 32456248 PMCID: PMC7279269 DOI: 10.3390/ijms21103686] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal tumors are responsible for more cancer-related fatalities than any other type of tumors, and colorectal and gastric malignancies account for a large part of these diseases. Thus, there is an urgent need to develop new therapeutic approaches to improve the patients' outcome and the tumor microenvironment is a promising arena for the development of such treatments. In fact, the nature of the microenvironment in the different gastrointestinal tracts may significantly influence not only tumor development but also the therapy response. In particular, an important microenvironmental component and a potential therapeutic target is the vasculature. In this context, the extracellular matrix is a key component exerting an active effect in all the hallmarks of cancer, including angiogenesis. Here, we summarized the current knowledge on the role of extracellular matrix in affecting endothelial cell function and intratumoral vascularization in the context of colorectal and gastric cancer. The extracellular matrix acts both directly on endothelial cells and indirectly through its remodeling and the consequent release of growth factors. We envision that a deeper understanding of the role of extracellular matrix and of its remodeling during cancer progression is of chief importance for the development of new, more efficacious, targeted therapies.
Collapse
Affiliation(s)
- Eva Andreuzzi
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Alessandra Capuano
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Evelina Poletto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Eliana Pivetta
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Albina Fejza
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Andrea Favero
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Roberto Doliana
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Renato Cannizzaro
- Department of Clinical Oncology, Experimental Gastrointestinal Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| | - Paola Spessotto
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| | - Maurizio Mongiat
- Department of Research and Diagnosis, Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (E.A.); (A.C.); (E.P.); (E.P.); (A.F.); (A.F.); (R.D.); (P.S.)
| |
Collapse
|
31
|
Dahlmann M, Werner R, Kortüm B, Kobelt D, Walther W, Stein U. Restoring Treatment Response in Colorectal Cancer Cells by Targeting MACC1-Dependent ABCB1 Expression in Combination Therapy. Front Oncol 2020; 10:599. [PMID: 32391276 PMCID: PMC7190815 DOI: 10.3389/fonc.2020.00599] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/01/2020] [Indexed: 12/29/2022] Open
Abstract
Treatment failure of solid cancers, represented by the development of drug resistance in the primary tumor or later outgrowth of drug resistant metastases, is the major cause of death for cancer patients. It represents an urgent clinical need for predictive biomarkers which indicate the success or failure of standard treatment regimens. Besides treatment prediction, interfering with cellular processes associated with drug resistance might improve treatment response by applying combination therapies. Metastasis-associated in colon cancer (MACC) 1 was identified in our group as a prognostic biomarker in human colorectal cancer, and has been established as key player, prognostic, and predictive biomarker for tumor progression and metastasis in a variety of solid cancers. Besides increased cell proliferation and motility, subsequently contributing to growth and metastatic spread of the primary tumor, MACC1 has also been shown to dysregulate apoptosis and is contributing to treatment resistance. Here we report the MACC1 dependent treatment resistance of colorectal cancer (CRC) cells to standard therapeutics like doxorubicin by upregulating ATP-binding cassette subfamily B member 1 (ABCB1) protein. Overexpression of MACC1 in CRC cells increased both its presence on the ABCB1 promoter and its transcriptional activity, resulting in elevated ABCB1 expression and thus treatment resistance to standard therapeutics. In contrast, depleting MACC1 increased intracellular drug concentrations, leading to better treatment response. We already identified the first MACC1 transcriptional inhibitors, such as lovastatin, by high-throughput screening of clinically approved small molecule drugs. These compounds inhibited cell motility in vitro but also restricted metastasis development in xenograft mouse models by reducing MACC1 expression. Here we report, that treating high MACC1 expressing CRC cells with a combination of statins and standard therapeutics increased the rate of cytotoxicity and resulted in higher treatment response.
Collapse
Affiliation(s)
- Mathias Dahlmann
- Experimental and Clinical Research Center, Charité University Medicine and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Rebecca Werner
- Experimental and Clinical Research Center, Charité University Medicine and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Benedikt Kortüm
- Experimental and Clinical Research Center, Charité University Medicine and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité University Medicine and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité University Medicine and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité University Medicine and the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
32
|
Examination of the expression levels of MACC1, Filamin A and FBXW7 genes in colorectal cancer patients. North Clin Istanb 2020; 7:1-5. [PMID: 32232196 PMCID: PMC7103745 DOI: 10.14744/nci.2019.26780] [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: 11/21/2018] [Accepted: 02/19/2019] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE: Colorectal cancer (CRC) is the third most common type of cancer observed in cancer-related mortality because it has a high metastasis ratio. This study aims to investigate the expression levels of several genes, including metastasis-related colon cancer 1 (MACC1), Filamin A (FLNA), F-box/WD repeat-containing protein 7 (FBXW7), which has an important role in cell signaling, migration and adhesion through the remodeling of the cell skeleton. METHODS: In this study, 21 patients with a precise diagnosis of CRC and 21 controls were included. Gene expressions were examined using the RT-PCR technique. To define the relationship of the genes with metastasis, blood samples were collected from all patients with colon/rectal cancer diagnosis without metastasis at six months before and after the medication with Xelox. RESULTS: Our findings showed that no significant difference was observed in the pre-treatment values compared to the control group, whereas FLNA (p=0.001) expression was observed to be significantly increased following treatment with Xelox. CONCLUSION: To our knowledge, our study is the first study to investigate the effects of Xelox treatment on the expression levels of MACC1, FBXW7 and FLNA genes in non-metastatic colorectal cancer patients in Turkey.
Collapse
|
33
|
Lu H, Feng Y, Hu Y, Guo Y, Liu Y, Mao Q, Xue W. Spondin 2 promotes the proliferation, migration and invasion of gastric cancer cells. J Cell Mol Med 2019; 24:98-113. [PMID: 31691494 PMCID: PMC6933360 DOI: 10.1111/jcmm.14618] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/17/2019] [Accepted: 07/28/2019] [Indexed: 12/24/2022] Open
Abstract
Spondin 2 (SPON2), a member of the Mindin F‐Spondin family, identifies pathogens, activates congenital immunity and promotes the growth and adhesion of neurons as well as binding to their receptors, but its role in promoting or inhibiting tumour metastasis is controversial. Here, we investigated its expression levels and mechanism of action in gastric cancer (GC). Western blotting and GC tissue arrays were used to determine the expression levels of SPON2. ELISAs were performed to measure the serum levels of SPON2 in patients with GC. Two GC cell lines expressing low levels of SPON2 were used to analyse the effects of regulating SPON2 expression on proliferation, migration, invasion, the cell cycle and apoptosis. The results revealed that SPON2 was highly expressed in GC tissues from patients with relapse or metastasis. The levels of SPON2 in sera of patients with GC were significantly higher compared with those of healthy individuals and patients with atrophic gastritis. Knockdown of SPON2 expression significantly inhibited the proliferation, migration and invasion of GC cells in vitro and in vivo. Down‐regulation of SPON2 arrested the cell cycle in G1/S, accelerated apoptosis through the mitochondrial pathway and inhibited the epithelial‐mesenchymal transition by blocking activation of the ERK1/2 pathway. In summary, this study suggests that SPON2 acts as an oncogene in the development of GC and may serve as a marker for the diagnosing GC as well as a new therapeutic target for GC.
Collapse
Affiliation(s)
- Haoming Lu
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China.,Research Center of Clinical Medicine, Nantong University Affiliated Hospital, Nantong, China
| | - Ying Feng
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China
| | - Yilin Hu
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China
| | - Yibing Guo
- Research Center of Clinical Medicine, Nantong University Affiliated Hospital, Nantong, China
| | - Yifei Liu
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, China
| | - Qinsheng Mao
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China
| | - Wanjiang Xue
- Department of Gastrointestinal Surgery, Nantong University Affiliated Hospital, Nantong, China.,Research Center of Clinical Medicine, Nantong University Affiliated Hospital, Nantong, China
| |
Collapse
|
34
|
Sousa AR, Oliveira AV, Oliveira MJ, Sarmento B. Nanotechnology-based siRNA delivery strategies for metastatic colorectal cancer therapy. Int J Pharm 2019; 568:118530. [DOI: 10.1016/j.ijpharm.2019.118530] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 12/17/2022]
|
35
|
Decoding and targeting the molecular basis of MACC1-driven metastatic spread: Lessons from big data mining and clinical-experimental approaches. Semin Cancer Biol 2019; 60:365-379. [PMID: 31430556 DOI: 10.1016/j.semcancer.2019.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Metastasis remains the key issue impacting cancer patient survival and failure or success of cancer therapies. Metastatic spread is a complex process including dissemination of single cells or collective cell migration, penetration of the blood or lymphatic vessels and seeding at a distant organ site. Hundreds of genes involved in metastasis have been identified in studies across numerous cancer types. Here, we analyzed how the metastasis-associated gene MACC1 cooperates with other genes in metastatic spread and how these coactions could be exploited by combination therapies: We performed (i) a MACC1 correlation analysis across 33 cancer types in the mRNA expression data of TCGA and (ii) a comprehensive literature search on reported MACC1 combinations and regulation mechanisms. The key genes MET, HGF and MMP7 reported together with MACC1 showed significant positive correlations with MACC1 in more than half of the cancer types included in the big data analysis. However, ten other genes also reported together with MACC1 in the literature showed significant positive correlations with MACC1 in only a minority of 5 to 15 cancer types. To uncover transcriptional regulation mechanisms that are activated simultaneously with MACC1, we isolated pan-cancer consensus lists of 1306 positively and 590 negatively MACC1-correlating genes from the TCGA data and analyzed each of these lists for sharing transcription factor binding motifs in the promotor region. In these lists, binding sites for the transcription factors TELF1, ETS2, ETV4, TEAD1, FOXO4, NFE2L1, ELK1, SP1 and NFE2L2 were significantly enriched, but none of them except SP1 was reported in combination with MACC1 in the literature. Thus, while some of the results of the big data analysis were in line with the reported experimental results, hypotheses on new genes involved in MACC1-driven metastasis formation could be generated and warrant experimental validation. Furthermore, the results of the big data analysis can help to prioritize cancer types for experimental studies and testing of combination therapies.
Collapse
|
36
|
Radhakrishnan H, Walther W, Zincke F, Kobelt D, Imbastari F, Erdem M, Kortüm B, Dahlmann M, Stein U. MACC1-the first decade of a key metastasis molecule from gene discovery to clinical translation. Cancer Metastasis Rev 2019; 37:805-820. [PMID: 30607625 DOI: 10.1007/s10555-018-9771-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Deciphering the paths to metastasis and identifying key molecules driving this process is one important issue for understanding and treatment of cancer. Such a key driver molecule is Metastasis Associated in Colon Cancer 1 (MACC1). A decade long research on this evolutionarily conserved molecule with features of a transcription factor as well as an adapter protein for versatile protein-protein interactions has shown that it has manifold properties driving tumors to their metastatic stage. MACC1 transcriptionally regulates genes involved in epithelial-mesenchymal transition (EMT), including those which are able to directly induce metastasis like c-MET, impacts tumor cell migration and invasion, and induces metastasis in solid cancers. MACC1 has proven as a valuable biomarker for prognosis of metastasis formation linked to patient survival and gives promise to also act as a predictive marker for individualized therapies in a broad variety of cancers. This review discusses the many features of MACC1 in the context of the hallmarks of cancer and the potential of this molecule as biomarker and novel therapeutic target for restriction and prevention of metastasis.
Collapse
Affiliation(s)
- Harikrishnan Radhakrishnan
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Fabian Zincke
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Francesca Imbastari
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Müge Erdem
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Benedikt Kortüm
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Mathias Dahlmann
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany. .,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
37
|
Zhou B, Yan Y, Wang Y, You S, Freeman MR, Yang W. Quantitative proteomic analysis of prostate tissue specimens identifies deregulated protein complexes in primary prostate cancer. Clin Proteomics 2019; 16:15. [PMID: 31011308 PMCID: PMC6461817 DOI: 10.1186/s12014-019-9236-2] [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: 09/10/2018] [Accepted: 04/09/2019] [Indexed: 12/18/2022] Open
Abstract
Background Prostate cancer (PCa) is the most frequently diagnosed non-skin cancer and a leading cause of mortality among males in developed countries. However, our understanding of the global changes of protein complexes within PCa tissue specimens remains very limited, although it has been well recognized that protein complexes carry out essentially all major processes in living organisms and that their deregulation drives the pathogenesis and progression of various diseases. Methods By coupling tandem mass tagging-synchronous precursor selection-mass spectrometry/mass spectrometry/mass spectrometry with differential expression and co-regulation analyses, the present study compared the differences between protein complexes in normal prostate, low-grade PCa, and high-grade PCa tissue specimens. Results Globally, a large downregulated putative protein–protein interaction (PPI) network was detected in both low-grade and high-grade PCa, yet a large upregulated putative PPI network was only detected in high-grade but not low-grade PCa, compared with normal controls. To identify specific protein complexes that are deregulated in PCa, quantified proteins were mapped to protein complexes in CORUM (v3.0), a high-quality collection of 4274 experimentally verified mammalian protein complexes. Differential expression and gene ontology (GO) enrichment analyses suggested that 13 integrin complexes involved in cell adhesion were significantly downregulated in both low- and high-grade PCa compared with normal prostate, and that four Prothymosin alpha (ProTα) complexes were significantly upregulated in high-grade PCa compared with normal prostate. Moreover, differential co-regulation and GO enrichment analyses indicated that the assembly levels of six protein complexes involved in RNA splicing were significantly increased in low-grade PCa, and those of four subcomplexes of mitochondrial complex I were significantly increased in high-grade PCa, compared with normal prostate. Conclusions In summary, to the best of our knowledge, the study represents the first large-scale and quantitative, albeit indirect, comparison of individual protein complexes in human PCa tissue specimens. It may serve as a useful resource for better understanding the deregulation of protein complexes in primary PCa. Electronic supplementary material The online version of this article (10.1186/s12014-019-9236-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bo Zhou
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Rm. 4009, Davis Research Bldg 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Yiwu Yan
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Rm. 4009, Davis Research Bldg 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Yang Wang
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Rm. 4009, Davis Research Bldg 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Sungyong You
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Rm. 4009, Davis Research Bldg 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Michael R Freeman
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Rm. 4009, Davis Research Bldg 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Wei Yang
- Division of Cancer Biology and Therapeutics, Departments of Surgery and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Rm. 4009, Davis Research Bldg 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| |
Collapse
|
38
|
Ni H, Ni T, Feng J, Bian T, Liu Y, Zhang J. Spondin-2 is a novel diagnostic biomarker for laryngeal squamous cell carcinoma. Pathol Res Pract 2018; 215:286-291. [PMID: 30527359 DOI: 10.1016/j.prp.2018.11.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/08/2018] [Accepted: 11/23/2018] [Indexed: 12/19/2022]
Abstract
Spondin-2, belongs to the SOX (SRY-related HMG box) gene family, plays a vital role in the development of malignancy, however, the role of Spondin-2 in laryngeal squamous cell carcinoma (LSCC) remains unknown. The aim of this study is to investigate the prognostic significance of and probable mechanism of Spondin-2 in LSCC. qRT-PCR, western blotting assays and IHC analysis demonstrated that Spondin-2 was significantly increased in LSCC tissues compared with adjacent non-tumorous tissues. In addition, high levels of Spondin-2 was associated with clinical stage, lymph node metastasis and pathology grade of LSCC patients (P <0.05). Kaplan-Meier analysis showed that patients with high expression of Spondin-2 had a lower overall survival rate (P<0.05) than that with low expression of Spondin-2. Moreover, spondin-2 silencing inhibited the proliferation of LSCC cells through inhibiting the activation of PI3K/AKT signaling. In conclusion, spondin-2 might be a novel therapeutic target and prognostic biomarker for LSCC patients.
Collapse
Affiliation(s)
- Haosheng Ni
- Department of Otorhinolaryngology, Affiliated Hospital of Nantong University, No. 20 Xi Si Road, Nantong, 226001, China
| | - Tingting Ni
- Department of Oncology, Nantong Tumor Hospital, No. 30 Tong Yang North Road, Nantong 226001, China
| | - Jia Feng
- Department of Pathology, Affiliated Hospital of Nantong University, No. 20 Xi Si Road, Nantong, 226001, China
| | - Tingting Bian
- Department of Pathology, Affiliated Hospital of Nantong University, No. 20 Xi Si Road, Nantong, 226001, China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, No. 20 Xi Si Road, Nantong, 226001, China.
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, No. 20 Xi Si Road, Nantong, 226001, China.
| |
Collapse
|
39
|
Feng Y, Hu Y, Mao Q, Guo Y, Liu Y, Xue W, Cheng S. Upregulation of Spondin-2 protein expression correlates with poor prognosis in hepatocellular carcinoma. J Int Med Res 2018; 47:569-579. [PMID: 30318967 PMCID: PMC6381490 DOI: 10.1177/0300060518803232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE The aim of this study was to measure the extracellular matrix protein Spondin-2 (SPON2) in hepatocellular carcinoma (HCC) tissues and to determine its potential value as a prognostic indicator by assessing its correlation with clinicopathological variables and survival. METHODS SPON2 mRNA expression was assessed in 20 matched pairs of HCC and non-cancerous liver tissues by quantitative reverse transcription-polymerase chain reaction analysis. SPON2 protein expression was determined in 107 matched pairs of HCC and normal liver tissue by immunohistochemical staining of tissue microarrays. RESULTS Analysis of patient tissues and Oncomine datasets showed that SPON2 mRNA and SPON2 protein expression were both significantly upregulated in HCC tissues, compared with non-cancerous liver tissue; moreover, both correlated significantly with tumor size. Kaplan-Meier analysis revealed that HCC patients who showed high levels of cytoplasmic SPON2 protein had poorer survival following curative resection, compared with HCC patients who exhibited low protein expression levels. Multivariate Cox regression analysis showed that tumor thrombus and SPON2 protein expression both independently correlated with reduced survival in HCC patients. CONCLUSION Upregulated expression of SPON2 protein in tumor tissue could be an effective prognostic indicator for patients with HCC.
Collapse
Affiliation(s)
- Ying Feng
- 1 The Third Affiliated Hospital of Soochow University, Changzhou, China.,2 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yilin Hu
- 2 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Qinsheng Mao
- 2 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yibing Guo
- 3 Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yifei Liu
- 4 Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wanjiang Xue
- 2 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, China.,3 Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Shuqun Cheng
- 5 Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
40
|
Kim HJ, Moon SJ, Kim SH, Heo K, Kim JH. DBC1 regulates Wnt/β-catenin-mediated expression of MACC1, a key regulator of cancer progression, in colon cancer. Cell Death Dis 2018; 9:831. [PMID: 30082743 PMCID: PMC6079074 DOI: 10.1038/s41419-018-0899-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/22/2022]
Abstract
Metastasis-associated in colon cancer 1 (MACC1) has been reported to be overexpressed in multiple cancers and promote proliferation, metastasis, cancer stem cell-like properties, and drug resistance of cancer cells. Despite its significance and the considerable knowledge accumulated on the function of MACC1 in various types of human malignancies, regulatory mechanisms underlying MACC1 expression remain unclear. Here we report that MACC1 is a direct target of Wnt/β-catenin signaling pathway in colon cancer cells and that DBC1 functions as a coactivator for Wnt-mediated MACC1 expression by promoting the activity of a LEF1/β-catenin-dependent enhancer located in intron 1 of MACC1 gene. DBC1 is required for LEF1/β-catenin complex formation on the MACC1 enhancer and for long-distance enhancer-promoter interaction of the MACC1 locus. MACC1 expression was increased in colonosphere cells compared to adherent colon cancer cells, and DBC1 overexpression further increased MACC1 expression in colonospheres and promoted sphere-forming abilities of colon cancer cells and drug resistance of colonospheres. Importantly, expressions of MACC1 and DBC1 are positively correlated with each other, upregulated in high-risk groups of colorectal cancer patients, and associated with poor survival. Our results establish MACC1 as a transcriptional target of Wnt/β-catenin signaling and suggest that DBC1 plays a key role in colorectal cancer progression through Wnt/β-catenin-MACC1 signaling axis.
Collapse
Affiliation(s)
- Hwa Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Korea.,Department of Biomedical Sciences, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, Korea
| | - Sue Jin Moon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Korea.,Department of Biomedical Sciences, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, Korea
| | - Seok-Hyung Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Korea.,Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea
| | - Kyu Heo
- Department of Clinical Research, Dongnam Institute of Radiological and Medical Sciences, Busan, 46033, Korea
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, Korea. .,Department of Biomedical Sciences, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, 06351, Korea.
| |
Collapse
|
41
|
Zhao Y, Dai C, Wang M, Kang H, Lin S, Yang P, Liu X, Liu K, Xu P, Zheng Y, Li S, Dai Z. Clinicopathological and prognostic significance of metastasis-associated in colon cancer-1 (MACC1) overexpression in colorectal cancer: a meta-analysis. Oncotarget 2018; 7:62966-62975. [PMID: 27542234 PMCID: PMC5325340 DOI: 10.18632/oncotarget.11287] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/19/2016] [Indexed: 01/11/2023] Open
Abstract
Metastasis-associated in colon cancer-1 (MACC1) has been reported to be overexpressed in diverse human malignancies, and the increasing amount of evidences suggest that its overexpression is associated with the development and progression of many human tumors. However, the prognostic and clinicopathological value of MACC1 in colorectal cancer remains inconclusive. Therefore, we conducted this meta-analysis to investigate the effect of MACC1 overexpression on clinicopathological features and survival outcomes in colorectal cancer. PubMed, CNKI, and Wanfang databases were searched for relevant articles published update to December 2015. Correlation of MACC1 expression level with overall survival (OS), disease-free survival (DFS), and clinicopathological features were analyzed. In this meta-analysis, fifteen studies with a total of 2,161 colorectal cancer patients were included. Our results showed that MACC1 overexpression was significantly associated with poorer OS and DFS. Moreover, MACC1 overexpression was significantly associated with gender, localization, TNM stage, T stage, and N stage. Together, our meta-analysis showed that MACC1 overexpression was significantly associated with poor survival rates, regional invasion and lymph-node metastasis. MACC1 expression level can serve as a novel prognostic factor in colorectal cancer patients.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Cong Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Wang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huafeng Kang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuai Lin
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pengtao Yang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinghan Liu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kang Liu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Peng Xu
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Zheng
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shanli Li
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhijun Dai
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
42
|
Zhang YL, Li Q, Yang XM, Fang F, Li J, Wang YH, Yang Q, Zhu L, Nie HZ, Zhang XL, Feng MX, Jiang SH, Tian GA, Hu LP, Lee HY, Lee SJ, Xia Q, Zhang ZG. SPON2 Promotes M1-like Macrophage Recruitment and Inhibits Hepatocellular Carcinoma Metastasis by Distinct Integrin-Rho GTPase-Hippo Pathways. Cancer Res 2018; 78:2305-2317. [PMID: 29440144 DOI: 10.1158/0008-5472.can-17-2867] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/27/2017] [Accepted: 02/09/2018] [Indexed: 11/16/2022]
Abstract
Tumor-associated macrophages (TAM) represent key regulators of the complex interplay between cancer and the immune microenvironment. Matricellular protein SPON2 is essential for recruiting lymphocytes and initiating immune responses. Recent studies have shown that SPON2 has complicated roles in cell migration and tumor progression. Here we report that, in the tumor microenvironment of hepatocellular carcinoma (HCC), SPON2 not only promotes infiltration of M1-like macrophages but also inhibits tumor metastasis. SPON2-α4β1 integrin signaling activated RhoA and Rac1, increased F-actin reorganization, and promoted M1-like macrophage recruitment. F-Actin accumulation also activated the Hippo pathway by suppressing LATS1 phosphorylation, promoting YAP nuclear translocation, and initiating downstream gene expression. However, SPON2-α5β1 integrin signaling inactivated RhoA and prevented F-actin assembly, thereby inhibiting HCC cell migration; the Hippo pathway was not noticeably involved in SPON2-mediated HCC cell migration. In HCC patients, SPON2 levels correlated positively with prognosis. Overall, our findings provide evidence that SPON2 is a critical factor in mediating the immune response against tumor cell growth and migration in HCC.Significance: Matricellular protein SPON2 acts as an HCC suppressor and utilizes distinct signaling events to perform dual functions in HCC microenvironment.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/9/2305/F1.large.jpg Cancer Res; 78(9); 2305-17. ©2018 AACR.
Collapse
Affiliation(s)
- Yan-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xiao-Mei Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Fang Fang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Ya-Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Lei Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Hui-Zhen Nie
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Ming-Xuan Feng
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Guang-Ang Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Ho-Young Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Su-Jae Lee
- Department of Life Science, Research Institute for Nature Sciences, Hanyang University, Seoul, Republic of Korea
| | - Qiang Xia
- Department of Liver Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China.
| |
Collapse
|
43
|
MACC1 is post-transcriptionally regulated by miR-218 in colorectal cancer. Oncotarget 2018; 7:53443-53458. [PMID: 27462788 PMCID: PMC5288198 DOI: 10.18632/oncotarget.10803] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023] Open
Abstract
Metastasis is a multistep molecular network process, which is lethal for more than 90% of the cancer patients. Understanding the regulatory functions of metastasis-inducing molecules is in high demand for improved therapeutic cancer approaches. Thus, we studied the post-transcriptional regulation of the crucial carcinogenic and metastasis-mediating molecule metastasis associated in colon cancer 1 (MACC1). In silico analysis revealed MACC1 as a potential target of miR-218, a tumor suppressor miRNA. Expression of these two molecules inversely correlated in colorectal cancer (CRC) cell lines. In a cohort of CRC patient tissues (n = 59), miR-218 is significantly downregulated and MACC1 is upregulated compared with normal mucosa. Luciferase reporter assays with a construct of the MACC1-3′-UTR harboring either the wild type or the mutated miR-218 seed sequence confirmed the specificity of the targeting. miR-218 inhibited significantly MACC1 protein expression, and consistently, MACC1-mediated migration, invasion and colony formation in CRC cells. Anti-miR-218 enhanced the MACC1-mediated migration, invasion and colony formation. Similar findings were observed in the gastric cancer cell line MKN-45. Further, we performed methylation-specific PCR of the SLIT2 and SLIT3 promoter, where miR-218 is encoded in intronic regions. The SLIT2 and SLIT3 promoters are hypermethylated in CRC cell lines. miR-218 and SLIT2 expressions correlated positively. Methyltransferase inhibitor 5-Azacytidine induced miR-218 expression and inhibited the expression of its target MACC1. We also determined that MACC1 has alternative polyadenylation (APA) sites, which results in different lengths of 3′-UTR variants in a CRC cell line. Taken together, miR-218 is post-transcriptionally inhibiting the MACC1 expression and its metastasis-inducing abilities.
Collapse
|
44
|
Patil VS, Madrigal A, Schmiedel BJ, Clarke J, O'Rourke P, de Silva AD, Harris E, Peters B, Seumois G, Weiskopf D, Sette A, Vijayanand P. Precursors of human CD4 + cytotoxic T lymphocytes identified by single-cell transcriptome analysis. Sci Immunol 2018; 3:eaan8664. [PMID: 29352091 PMCID: PMC5931334 DOI: 10.1126/sciimmunol.aan8664] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/27/2017] [Accepted: 11/30/2017] [Indexed: 01/03/2023]
Abstract
CD4+ cytotoxic T lymphocytes (CD4-CTLs) have been reported to play a protective role in several viral infections. However, little is known in humans about the biology of CD4-CTL generation, their functional properties, and heterogeneity, especially in relation to other well-described CD4+ memory T cell subsets. We performed single-cell RNA sequencing in more than 9000 cells to unravel CD4-CTL heterogeneity, transcriptional profile, and clonality in humans. Single-cell differential gene expression analysis revealed a spectrum of known transcripts, including several linked to cytotoxic and costimulatory function that are expressed at higher levels in the TEMRA (effector memory T cells expressing CD45RA) subset, which is highly enriched for CD4-CTLs, compared with CD4+ T cells in the central memory (TCM) and effector memory (TEM) subsets. Simultaneous T cell antigen receptor (TCR) analysis in single cells and bulk subsets revealed that CD4-TEMRA cells show marked clonal expansion compared with TCM and TEM cells and that most of CD4-TEMRA were dengue virus (DENV)-specific in donors with previous DENV infection. The profile of CD4-TEMRA was highly heterogeneous across donors, with four distinct clusters identified by the single-cell analysis. We identified distinct clusters of CD4-CTL effector and precursor cells in the TEMRA subset; the precursor cells shared TCR clonotypes with CD4-CTL effectors and were distinguished by high expression of the interleukin-7 receptor. Our identification of a CD4-CTL precursor population may allow further investigation of how CD4-CTLs arise in humans and, thus, could provide insights into the mechanisms that may be used to generate durable and effective CD4-CTL immunity.
Collapse
Affiliation(s)
- Veena S Patil
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Ariel Madrigal
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Benjamin J Schmiedel
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - James Clarke
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Patrick O'Rourke
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Aruna D de Silva
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
- Genetech Research Institute, Colombo, Sri Lanka
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
- Department of Medicine, University of California San Diego, 9500 Gilman Drive #0656, La Jolla, CA 92093, USA
| | - Gregory Seumois
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
- Department of Medicine, University of California San Diego, 9500 Gilman Drive #0656, La Jolla, CA 92093, USA
| | - Pandurangan Vijayanand
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.
- Department of Medicine, University of California San Diego, 9500 Gilman Drive #0656, La Jolla, CA 92093, USA
- Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine University of Southampton, Southampton, UK
| |
Collapse
|
45
|
Wang LF, Liu YS, Yang B, Li P, Cheng XS, Xiao CX, Liu JJ, Li S, Ren JL, Guleng B. The extracellular matrix protein mindin attenuates colon cancer progression by blocking angiogenesis via Egr-1-mediated regulation. Oncogene 2017; 37:601-615. [PMID: 28991232 DOI: 10.1038/onc.2017.359] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 08/15/2017] [Accepted: 08/19/2017] [Indexed: 12/13/2022]
Abstract
Mindin, a secreted, highly conserved extracellular matrix (ECM) protein, exerts a broad spectrum of effects on the innate immune system. However, its function in colorectal cancer (CRC) progression is not well established, and its upstream regulation mechanisms remain unclear. Contrary to previous reports, this study used two different enzyme-linked immunosorbent assay (ELISA) kits to show that the serum level of mindin was significantly decreased in CRC patients and that this decreased level is more significantly associated with the early stages of the disease. To explore the regulation of mindin, we used a bioinformatics approach to predict potential transcription factors and determined that early growth response factor (Egr)-1 directly regulates mindin expression at the transcriptional level using dual luciferase, chromatin immunoprecipitation (ChIP) DNA and electrophoretic mobility shift assay (EMSA) methods. Egr-1 regulates mindin mRNA and protein expression in CRC cells, and the protein expression of both Egr-1 and mindin was significantly decreased in tumor lesions of patients compared with adjacent control tissues. Mindin is essential for Egr-1-mediated inhibition of endothelial cell tube formation, and mindin inhibits endotheliocyte proliferation, migration and angiogenic sprouts in vitro. Overexpression of mindin suppressed xenograft tumor growth by blocking angiogenesis instead of directly suppressing CRC cell proliferation. Mechanically, mindin inhibits the hypoxia-induced HIF-1a and VEGFA protein expression in CRC cells and the phosphorylation of VEGFR-2 in endothelial cells. The results suggest that the serum level of mindin can be used as a novel biomarker for early detection of CRC and that the Egr-1/mindin axis is a potential therapeutic target for the inhibition of angiogenesis in CRC development.
Collapse
Affiliation(s)
- L-F Wang
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - Y-S Liu
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - B Yang
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - P Li
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China.,Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - X-S Cheng
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - C-X Xiao
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - J-J Liu
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - S Li
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, China
| | - J-L Ren
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - B Guleng
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China.,Faculty of Clinical Medicine, Medical College of Xiamen University, Xiamen, China.,State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, China
| |
Collapse
|
46
|
Rohr UP, Herrmann P, Ilm K, Zhang H, Lohmann S, Reiser A, Muranyi A, Smith J, Burock S, Osterland M, Leith K, Singh S, Brunhoeber P, Bowermaster R, Tie J, Christie M, Wong HL, Waring P, Shanmugam K, Gibbs P, Stein U. Prognostic value of MACC1 and proficient mismatch repair status for recurrence risk prediction in stage II colon cancer patients: the BIOGRID studies. Ann Oncol 2017; 28:1869-1875. [DOI: 10.1093/annonc/mdx207] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
47
|
Yuan X, Bian T, Liu J, Ke H, Feng J, Zhang Q, Qian L, Li X, Liu Y, Zhang J. Spondin2 is a new prognostic biomarker for lung adenocarcinoma. Oncotarget 2017; 8:59324-59332. [PMID: 28938639 PMCID: PMC5601735 DOI: 10.18632/oncotarget.19577] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 06/27/2017] [Indexed: 12/16/2022] Open
Abstract
Spondin 2 (SPON2) is a member of the F-spondin superfamily of genes that encode an extracellular matrix protein. SPON2 has been identified by mRNA differential display screening of cancerous and noncancerous lung cell lines in vitro [1], however, its role in pulmonary adenocarcinoma (ADC) patients remains unclear. In our study, we evaluated whether SPON2 can be used as a biomarker for the diagnosis of pulmonary ADC and any association between SPON2 protein levels and clinicopathological characteristics. Firstly, the mRNA levels of SPON2 in pulmonary ADCs and normal adjacent tissue samples were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR) (n = 60) assay and the expression of SPON2 protein were detected by tissue microarray immunohistochemistry analysis (TMA-IHC) (n = 280). Overexpression of SPON2 protein in cancerous tissues was associated with the clinical characteristics of ADC patients and their overall survival. Levels of SPON2 mRNA and protein were significantly expressed higher in ADC tissues than in adjacent normal tissues. Finally, through univariate and multivariate regression analysis, we found that overexpression of SPON2 protein levels correlates with differentiation, positive lymph nodes metastasis, higher serum carcinoembryonic antigen (CEA) level and poor overall survival. Overexpression of SPON2 protein is an independent prognostic biomarker in ADC patients. Our data revealed that SPON2 played an oncogene role in ADC development and progression. Inhibiting SPON2 might represent a new strategy for pulmonary ADC.
Collapse
Affiliation(s)
- Xiaopeng Yuan
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China.,Department of Radiation Oncology, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Tingting Bian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Jian Liu
- Department of Chemotherapy, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Honggang Ke
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Jia Feng
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Qing Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Li Qian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Xiaoli Li
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| |
Collapse
|
48
|
Fan JY, Zhang Y, Guo Q. MACC1 regulatory network in tumor metastasis. Shijie Huaren Xiaohua Zazhi 2017; 25:989-995. [DOI: 10.11569/wcjd.v25.i11.989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The MACC1 gene was firstly identified in colorectal cancer. Recently, abnormal upregulation of MACC1 has been detected in multiple tumors. The expression of MACC1 is shown to be positively associated with tumor metastasis, but negatively with prognosis of patients, and it represents a potential therapeutic target for anti-tumor strategies. MACC1 has increasingly emerged as a key regulator in metastatic processes, and it has been identified to be able to maintain multiple tumor-associated signaling pathways, transactivate oncogenic genes, and regulate epithelial-mesenchymal transition and tumor vascularization. On the other hand, MACC1 is regulated and influenced by non-coding RNAs and SNPs. The present review will summarize the recent progress in understanding the role of the MACC1 regulatory network in tumor metastasis.
Collapse
|
49
|
Pachmayr E, Treese C, Stein U. Underlying Mechanisms for Distant Metastasis - Molecular Biology. Visc Med 2017; 33:11-20. [PMID: 28785563 DOI: 10.1159/000454696] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The formation of distant metastases constitutes a complex process with a variety of different genes and pathways involved. To improve patient survival, it is necessary to understand the underlying mechanisms of metastasis to allow for targeted intervention. METHODS This review provides an overview of the general concepts of metastasis, focusing on the most important genes and pathways involved and on interventional strategies. RESULTS Cancer cells undergo different steps to form metastasis: most prominently, local invasion, intravasation, survival in the circulation, arrest at a distant organ site and extravasation, micrometastasis formation, and metastatic colonization. In order to pass these steps, different molecular pathways are of major importance: EGF/RAS/RAF/MEK/ERK, PI3K/Akt/mTOR, HGF/Met, Wnt/β-catenin, and VEGF signaling. The HGF/Met regulator MACC1 and the Wnt signaling target S100A4 have been shown to play a major role in the metastatic process. Each gene and pathway provides an opportunity for therapeutic intervention. CONCLUSION Since metastasis represents a highly limiting factor in cancer therapy causing 90% of cancer deaths, it is imperative to reveal the underlying mechanisms. This is fundamental for uncovering prognostic markers and new targeted therapy options.
Collapse
Affiliation(s)
- Eva Pachmayr
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Christoph Treese
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases, Rheumatology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
50
|
Mei Y, Yang JP, Qian CN. For robust big data analyses: a collection of 150 important pro-metastatic genes. CHINESE JOURNAL OF CANCER 2017; 36:16. [PMID: 28109319 PMCID: PMC5251273 DOI: 10.1186/s40880-016-0178-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/03/2016] [Indexed: 02/08/2023]
Abstract
Metastasis is the greatest contributor to cancer-related death. In the era of precision medicine, it is essential to predict and to prevent the spread of cancer cells to significantly improve patient survival. Thanks to the application of a variety of high-throughput technologies, accumulating big data enables researchers and clinicians to identify aggressive tumors as well as patients with a high risk of cancer metastasis. However, there have been few large-scale gene collection studies to enable metastasis-related analyses. In the last several years, emerging efforts have identified pro-metastatic genes in a variety of cancers, providing us the ability to generate a pro-metastatic gene cluster for big data analyses. We carefully selected 285 genes with in vivo evidence of promoting metastasis reported in the literature. These genes have been investigated in different tumor types. We used two datasets downloaded from The Cancer Genome Atlas database, specifically, datasets of clear cell renal cell carcinoma and hepatocellular carcinoma, for validation tests, and excluded any genes for which elevated expression level correlated with longer overall survival in any of the datasets. Ultimately, 150 pro-metastatic genes remained in our analyses. We believe this collection of pro-metastatic genes will be helpful for big data analyses, and eventually will accelerate anti-metastasis research and clinical intervention.
Collapse
Affiliation(s)
- Yan Mei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Jun-Ping Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China. .,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
| |
Collapse
|