1
|
Szmajda-Krygier D, Nocoń Z, Pietrzak J, Krygier A, Balcerczak E. Assessment of Methylation in Selected ADAMTS Family Genes in Non-Small-Cell Lung Cancer. Int J Mol Sci 2025; 26:934. [PMID: 39940703 PMCID: PMC11816904 DOI: 10.3390/ijms26030934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Alterations in the methylation of genetic material can influence carcinogenesis by the downregulation or overexpression of ADAMTS (a disintegrin-like and metalloprotease with thrombospondin motifs) protease genes. Through their proteolytic activity, these enzymes are also capable of promoting angiogenesis. Consequently, ADAMTS proteases can either facilitate or inhibit cancer progression. This study aimed to evaluate the methylation levels of the ADAMTS6, ADAMTS9, and ADAMTS12 genes in non-small-cell lung cancer (NSCLC) using data from bioinformatics databases. The focus was on differences between lung adenocarcinoma (LUAD) and lung squamous-cell carcinoma (LUSC) subtypes and their impact on patient overall survival (OS). ADAMTS6 gene expression is significantly reduced in LUSC, and analysis of ADAMTS9 gene expression showed a significantly reduced gene transcript level in LUAD and LUSC, while both NSCLC subtypes demonstrated ADAMTS12 upregulation. In LUSC, significantly elevated promoter methylation was found in all of the aforementioned genes, while in LUAD, higher promoter methylation was observed only for ADAMTS9 and ADAMTS12. The differential methylation region (DMR) pattern demonstrated by ADAMTS6, ADAMTS9, and ADAMTS12 is a useful tool for distinguishing normal from cancer cells. The areas under the curve (AUCs) ranged from 0.86 to 0.99 for both LUAD and LUSC subtypes. The methylation level of different CpG sites among selected ADAMTS members is related to patient survival, suggesting it may have value as a prognostic marker. The methylation degree of promoter regions in genes encoding ADAMTS family proteins could significantly influence LUSC and LUAD. Increased promoter methylation could also reduce certain gene expression, contributing to cancer progression. The expression levels and specific DMRs of ADAMTS genes may serve as prognostic markers correlating with patient OS. Assessing ADAMTS gene methylation could become a diagnostic tool for differentiating NSCLC subtypes and potentially guide therapeutic strategies. Further research is needed to fully understand the activity and mechanisms of ADAMTS family proteins.
Collapse
Affiliation(s)
- Dagmara Szmajda-Krygier
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| | - Zuzanna Nocoń
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Jacek Pietrzak
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| | - Adrian Krygier
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| | - Ewa Balcerczak
- Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
- BRaIn Laboratories, Medical University of Lodz, Czechoslowacka 4, 92-216 Lodz, Poland
| |
Collapse
|
2
|
Li M, Yuan Z, Tang Z. ADAMTS12, a novel prognostic predictor, promotes cell proliferation, migration, and invasion in head and neck squamous cell carcinoma. Oral Dis 2024; 30:235-246. [PMID: 36222542 DOI: 10.1111/odi.14403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/06/2022] [Accepted: 10/04/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The prognostic significance and potential carcinogenic mechanism of ADAM metallopeptidase with thrombospondin type 1 motif 12 (ADAMTS12) in head and neck squamous cell carcinoma (HNSC) remain unclear. MATERIALS AND METHODS Immunohistochemistry was used to analyze the correlation between ADAMTS12 protein expression and clinicopathological factors in tumor samples from 195 patients with HNSC. Based on clinicopathological data of patients, Cox regression and Kaplan-Meier analysis were used to identify the prognostic significance of the ADAMTS12 expression. The carcinogenicity of the ADAMTS12 in HNSC cells was analyzed by CCK-8 assay, the wound-healing assay, and transwell assays after transfection of ADAMTS12 overexpression or knock-down vector. RESULTS The expression of ADAMTS12 was up-regulated in HNSC compared with normal tissue, related to pathology grade and lymph node metastasis of patients with HNSC, which was an independent prognostic factor. ADAMTS12 overexpression facilitated cell viability, invasion, and migration of HNSC cells, while ADAMTS12 knock-down had inverse results. Moreover, enrichment analysis, ADAMTS12 overexpression assay, and ADAMTS12 knock-down assay confirmed that ADAMTS12 mediated the activation of P13K/Akt pathway in HNSC. CONCLUSIONS Our studies indicated that ADAMTS12 was a novel prognostic biomarker and potentially therapeutic target in HNSC.
Collapse
Affiliation(s)
- Ming Li
- Hunan Key Laboratory of Oral Health Research & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | | | - Zhangui Tang
- Hunan Key Laboratory of Oral Health Research & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
3
|
Meibom D, Wasnaire P, Beyer K, Broehl A, Cancho-Grande Y, Elowe N, Henninger K, Johannes S, Jungmann N, Krainz T, Lindner N, Maassen S, MacDonald B, Menshykau D, Mittendorf J, Sanchez G, Schaefer M, Stefan E, Torge A, Xing Y, Zubov D. BAY-9835: Discovery of the First Orally Bioavailable ADAMTS7 Inhibitor. J Med Chem 2024; 67:2907-2940. [PMID: 38348661 PMCID: PMC10895658 DOI: 10.1021/acs.jmedchem.3c02036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 02/23/2024]
Abstract
The matrix metalloprotease ADAMTS7 has been identified by multiple genome-wide association studies as being involved in the development of coronary artery disease. Subsequent research revealed the proteolytic function of the enzyme to be relevant for atherogenesis and restenosis after vessel injury. Based on a publicly known dual ADAMTS4/ADAMTS5 inhibitor, we have in silico designed an ADAMTS7 inhibitor of the catalytic domain, which served as a starting point for an optimization campaign. Initially our inhibitors suffered from low selectivity vs MMP12. An X-ray cocrystal structure inspired us to exploit amino acid differences in the binding site of MMP12 and ADAMTS7 to improve selectivity. Further optimization composed of employing 5-membered heteroaromatic groups as hydantoin substituents to become more potent on ADAMTS7. Finally, fine-tuning of DMPK properties yielded BAY-9835, the first orally bioavailable ADAMTS7 inhibitor. Further optimization to improve selectivity vs ADAMTS12 seems possible, and a respective starting point could be identified.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Eric Stefan
- Broad
Institute, 02142 Cambridge, United States
| | | | - Yi Xing
- Broad
Institute, 02142 Cambridge, United States
| | | |
Collapse
|
4
|
Mohamedi Y, Fontanil T, Vega JA, Cobo T, Cal S, Obaya ÁJ. Lung Inflammatory Phenotype in Mice Deficient in Fibulin-2 and ADAMTS-12. Int J Mol Sci 2024; 25:2024. [PMID: 38396702 PMCID: PMC10888546 DOI: 10.3390/ijms25042024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Interaction between extracellular matrix (ECM) components plays an important role in the regulation of cellular behavior and hence in tissue function. Consequently, characterization of new interactions within ECM opens the possibility of studying not only the functional but also the pathological consequences derived from those interactions. We have previously described the interaction between fibulin2 and ADAMTS-12 in vitro and the effects of that interaction using cellular models of cancer. Now, we generate a mouse deficient in both ECM components and evaluate functional consequences of their absence using different cancer and inflammation murine models. The main findings indicate that mice deficient in both fibulin2 and ADAMTS12 markedly increase the development of lung tumors following intraperitoneal urethane injections. Moreover, inflammatory phenotype is exacerbated in the lung after LPS treatment as can be inferred from the accumulation of active immune cells in lung parenchyma. Overall, our results suggest that protective effects in cancer or inflammation shown by fibulin2 and ADAMTS12 as interactive partners in vitro are also shown in a more realistic in vivo context.
Collapse
Affiliation(s)
- Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Spain
| | - José A. Vega
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, 33006 Oviedo, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Providencia—Área Metropolinana, Santiago de Chile 7500912, Chile
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto Asturiano de Odontología (IAO), 33006 Oviedo, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Álvaro J. Obaya
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
| |
Collapse
|
5
|
Jiang Y, Huang J, Huang Z, Li W, Tan R, Li T, Chen Z, Tang X, Zhao Y, Qiu J, Li C, Chen H, Yang Z. ADAMTS12 promotes oxaliplatin chemoresistance and angiogenesis in gastric cancer through VEGF upregulation. Cell Signal 2023; 111:110866. [PMID: 37619822 DOI: 10.1016/j.cellsig.2023.110866] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/06/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND While ADAMTS12 (A disintegrin and metalloproteinase with thrombospondin motifs 12) has been established as an important regulator of gastrointestinal tumor development and angiogenic activity, the precise mechanistic functions of ADAMTS12 have yet to be fully clarified in gastric cancer (GC). Accordingly, this study was developed to explore the molecular functions of ADAMTS12 in GC and to examine its utility as a biomarker associated with chemoresistance and prognostic outcomes in this cancer type. METHODS The ability of ADAMTS12 to modulate the proliferative, migratory, invasive, chemoresistant, and tube formation activity of tumor cells was assessed in vivo and in vitro through gain- and loss-of-function approaches. Correlations between ADAMTS12, CD31, and VEGF expression levels in GC patient tumor tissue samples from individuals that did and did not undergo neoadjuvant chemotherapy (NAC) treatment were analyzed via immunohistochemical staining. RESULTS These analyses revealed the ability of ADAMTS12 to promote in vivo and in vitro cellular proliferative and angiogenic activity, promoting the activation of ERK and the consequent upregulation of VEGF, thereby inducing angiogenesis and decreasing GC cell oxaliplatin sensitivity. A positive correlation between ADAMTS12 levels and both the expression of VEGF as well as the density of microvessels was observed in GC patient tumor tissues. Moreover, those GC patients exhibiting higher intratumoral ADAMTS12 expression exhibited worse responses to NAC treatment and worse overall survival outcomes. CONCLUSIONS These findings suggest that ADAMTS12 can modulate signaling via the MAPK/VEGF axis in GC cells to enhance tumor cell resistance to oxaliplatin treatment under hypoxic and normoxic conditions. Elevated ADAMTS12 levels can additionally predict vascular abnormalities, worse survival outcomes, and chemoresistance in patients with GC.
Collapse
Affiliation(s)
- Yingming Jiang
- Department of General Surgery (Department of Gastrointestinal Endoscopy), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Department of Thyroid Hernia Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, PR China
| | - Jintuan Huang
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Zhenze Huang
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Weiyao Li
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Rongchang Tan
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Tuoyang Li
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Zijian Chen
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Xiaocheng Tang
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Yandong Zhao
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Department of Pathology, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Jun Qiu
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Chujun Li
- Department of General Surgery (Department of Gastrointestinal Endoscopy), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China.
| | - Hao Chen
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China.
| | - Zuli Yang
- Department of General Surgery (Department of Gastric Surgery Section 2), The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China.
| |
Collapse
|
6
|
Zou R, Gu R, Tu X, Chen J, Liu S, Xue X, Li W, Zhang Y. Effects of metalloprotease ADAMTS12 on cervical cancer cell phenotype and its potential mechanism. Discov Oncol 2023; 14:162. [PMID: 37642715 PMCID: PMC10465472 DOI: 10.1007/s12672-023-00776-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
ADAMTS12 is a gene widely expressed in human tissues. We studied the expression level of ADAMTS12 in cervical cancer tissue and its relationship with clinicopathological features. We also explored the function of ADAMTS12 in cervical cancer cells and its underlying mechanisms. We found the higher expression level of ADAMTS12 in cancer tissues, which was associated with the worse overall survival rate. The immunofluorescence assay showed that the cytoplasm of cervical cancer cells is the main expression site of ADAMTS12. Overexpression of ADAMTS12 in HeLa and CaSki cells prominently promoted the cell proliferation, migration and invasion. We found that 2032 genes were correlated with ADAMTS12, which was mainly related to extracellular matrix, TGF-β signaling pathway. The phosphorylation levels of mTOR and 4E-BP1 were upregulated in ADAMTS12-overexpressing cells. Co-Immunoprecipitation combined with protein mass spectrometry showed that TGF-β signaling pathway-related proteins interacting with ADAMTS12 were screened from HeLa cells with ADAMTS12 overexpression. Therefore, we concluded that ADAMTS12 may affect the mTOR signaling pathway through the interacting with TGF-β1, and then affect the biological function of cervical cancer cells.
Collapse
Affiliation(s)
- Ruanmin Zou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, People's Republic of China.
| | - Ruihong Gu
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, College of Basic Medicine, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xinyu Tu
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, College of Basic Medicine, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jiani Chen
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, College of Basic Medicine, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Songjun Liu
- Department of Gynecology, Tongde Hospital of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xiangyang Xue
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, College of Basic Medicine, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Wensu Li
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, College of Basic Medicine, Wenzhou Medical University, Wenzhou, People's Republic of China.
| | - Yuyang Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, People's Republic of China.
| |
Collapse
|
7
|
Sheng H, Zhang J, Pan C, Wang S, Gu S, Li F, Ma Y, Ma Y. Genome-wide identification of bovine ADAMTS gene family and analysis of its expression profile in the inflammatory process of mammary epithelial cells. Int J Biol Macromol 2023:125304. [PMID: 37315674 DOI: 10.1016/j.ijbiomac.2023.125304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/29/2023] [Accepted: 06/04/2023] [Indexed: 06/16/2023]
Abstract
ADAM metallopeptidase with thrombospondin type 1 motif (ADAMTS) are secreted, multi-domain matrix-related zinc endopeptidases that play a role in organogenesis, assembly and degradation of extracellular matrix (ECM), cancer and inflammation. Genome-wide identification and analysis of the bovine ADAMTS gene family has not yet been carried out. In this study, 19 ADAMTS family genes were identified in Bos taurus by genome-wide bioinformatics analysis, and they were unevenly distributed on 12 chromosomes. Phylogenetic analysis shows that the Bos taurus ADAMTS are divided into eight subfamilies, with highly consistent gene structures and motifs within the same subfamily. Collinearity analysis showed that the Bos taurus ADAMTS gene family is homologous to other bovine subfamily species, and many ADAMTS genes may be derived from tandem replication and segmental replication. In addition, based on the analysis of RNA-seq data, we found the expression pattern of ADAMTS gene in different tissues. Meanwhile, we also analyzed the expression profile of ADAMTS gene in the inflammatory response of bovine mammary epithelial cells (BMECs) stimulated by LPS by qRT-PCR. The results can provide ideas for understanding the evolutionary relationship and expression pattern of ADAMTS gene in Bovidae, and clarify the theoretical basis of the function of ADAMTS in inflammation.
Collapse
Affiliation(s)
- Hui Sheng
- School of Agriculture, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Junxing Zhang
- School of Agriculture, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Cuili Pan
- School of Agriculture, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Shuzhe Wang
- School of Agriculture, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Shuaifeng Gu
- School of Agriculture, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Fen Li
- School of Agriculture, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Yanfen Ma
- School of Agriculture, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Yun Ma
- School of Agriculture, Ningxia University, Yinchuan 750021, China; Key Laboratory of Ruminant Molecular and Cellular Breeding, School of Agriculture, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
8
|
Sharma D, Singh NK. The Biochemistry and Physiology of A Disintegrin and Metalloproteinases (ADAMs and ADAM-TSs) in Human Pathologies. Rev Physiol Biochem Pharmacol 2023; 184:69-120. [PMID: 35061104 DOI: 10.1007/112_2021_67] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metalloproteinases are a group of proteinases that plays a substantial role in extracellular matrix remodeling and its molecular signaling. Among these metalloproteinases, ADAMs (a disintegrin and metalloproteinases) and ADAM-TSs (ADAMs with thrombospondin domains) have emerged as highly efficient contributors mediating proteolytic processing of various signaling molecules. ADAMs are transmembrane metalloenzymes that facilitate the extracellular domain shedding of membrane-anchored proteins, cytokines, growth factors, ligands, and their receptors and therefore modulate their biological functions. ADAM-TSs are secretory, and soluble extracellular proteinases that mediate the cleavage of non-fibrillar extracellular matrix proteins. ADAMs and ADAM-TSs possess pro-domain, metalloproteinase, disintegrin, and cysteine-rich domains in common, but ADAM-TSs have characteristic thrombospondin motifs instead of the transmembrane domain. Most ADAMs and ADAM-TSs are activated by cleavage of pro-domain via pro-protein convertases at their N-terminus, hence directing them to various signaling pathways. In this article, we are discussing not only the structure and regulation of ADAMs and ADAM-TSs, but also the importance of these metalloproteinases in various human pathophysiological conditions like cardiovascular diseases, colorectal cancer, autoinflammatory diseases (sepsis/rheumatoid arthritis), Alzheimer's disease, proliferative retinopathies, and infectious diseases. Therefore, based on the emerging role of ADAMs and ADAM-TSs in various human pathologies, as summarized in this review, these metalloproteases can be considered as critical therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Deepti Sharma
- Department of Ophthalmology, Visual and Anatomical Sciences, Integrative Biosciences Center (IBio), Wayne State University School of Medicine, Detroit, MI, USA
| | - Nikhlesh K Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, Integrative Biosciences Center (IBio), Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
9
|
Song C, Chen J, Zhang C, Dong D. An Integrated Pan-Cancer Analysis of ADAMTS12 and Its Potential Implications in Pancreatic Adenocarcinoma. Front Oncol 2022; 12:849717. [PMID: 35280819 PMCID: PMC8904364 DOI: 10.3389/fonc.2022.849717] [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: 01/06/2022] [Accepted: 01/31/2022] [Indexed: 11/23/2022] Open
Abstract
Background A Disintegrin and Metallopeptidase with Thrombospondin Type 1 Motif 12 (ADAMTS12), a member of the ADAMTS family of multidomain extracellular protease enzymes, is involved in the progression of many tumors. However, a pan-cancer analysis of this gene has not yet been performed. Its role in pancreatic adenocarcinoma (PAAD) also remains unclear. Methods The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression data (GTEx) databases were used to analyze ADAMTS12 expression in pan-cancer. We assessed the expression, clinical characteristics, prognostic significance, copy number alteration, methylation, and mutation of ADAMTS12 and its correlation with the tumor immune microenvironment. qRT-PCR and immunohistochemistry assays were also performed to validate the expression of ADAMTS12 in PAAD. Results Through bioinformatics analysis and preliminary experimental verification, ADAMTS12 was found to be substantially overexpressed in PAAD. High expression level of ADAMTS12 was correlated with worse survival rates in patients with PAAD and high infiltration levels of tumor-associated macrophages, cancer-associated fibroblasts, immune checkpoint proteins, and immunosuppressive genes. Conclusion Our findings suggest ADAMTS12 as a potential prognostic biomarker in PAAD. Elevated ADAMTS12 expression may also indicate an immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Caiyun Song
- Department of Gastroenterology, Wenzhou People's Hospital, Wenzhou, China
| | - Jionghuang Chen
- Zhejiang Engineering Research Center of Cognitive Healthcare, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chaolei Zhang
- Zhejiang Engineering Research Center of Cognitive Healthcare, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dapeng Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Abstract
A Disintegrin and Metalloproteinase with Thrombospondin motifs (ADAMTS) are major mediators in extracellular matrix (ECM) turnover and have gained increasing interest over the last years as major players in ECM remodeling during tissue homeostasis and the development of diseases. Although, ADAMTSs are recognized in playing important roles during tissue remodeling, and loss of function in various member of the ADAMTS family could be associated with the development of numerous diseases, limited knowledge is available about their specific substrates and mechanism of action. In this chapter, we will review current knowledge about ADAMTSs and their use as disease biomarkers.
Collapse
Affiliation(s)
- Rahel Schnellmann
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
11
|
Liu L, Xu K, Zhou Y. Development of a novel embryonic germline gene-related prognostic model of lung adenocarcinoma. PeerJ 2021; 9:e12257. [PMID: 34721973 PMCID: PMC8542372 DOI: 10.7717/peerj.12257] [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: 06/04/2021] [Accepted: 09/15/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Emerging evidence implicates the correlation of embryonic germline genes with the tumor progress and patient's outcome. However, the prognostic value of these genes in lung adenocarcinoma (LUAD) has not been fully studied. Here we systematically evaluated this issue, and constructed a novel signature and a nomogram associated with embryonic germline genes for predicting the outcomes of lung adenocarcinoma. METHODS The LUAD cohorts retrieved from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database were used as training set and testing set, respectively. The embryonic germline genes were downloaded from the website https://venn.lodder.dev. Then, the differentially expressed embryonic germline genes (DEGGs) between the tumor and normal samples were identified by limma package. The functional enrichment and pathway analyses were also performed by clusterProfiler package. The prognostic model was constructed by the least absolute shrinkage and selection operator (LASSO)-Cox regression method. Survival and Receiver Operating Characteristic (ROC) analyses were performed to validate the model using training set and four testing GEO datasets. Finally, a prognostic nomogram based on the signature genes was constructed using multivariate regression method. RESULTS Among the identified 269 DEGGs, 249 were up-regulated and 20 were down-regulated. GO and KEGG analyses revealed that these DEGGs were mainly enriched in the process of cell proliferation and DNA damage repair. Then, 103 DEGGs with prognostic value were identified by univariate Cox regression and further filtered by LASSO method. The resulting sixteen DEGGs were included in step multivariate Cox regression and an eleven embryonic germline gene related signature (EGRS) was constructed. The model could robustly stratify the LUAD patients into high-risk and low-risk groups in both training and testing sets, and low-risk patients had much better outcomes. The multi-ROC analysis also showed that the EGRS model had the best predictive efficacy compared with other common clinicopathological factors. The EGRS model also showed robust predictive ability in four independent external datasets, and the area under curve (AUC) was 0.726 (GSE30219), 0.764 (GSE50081), 0.657 (GSE37745) and 0.668 (GSE72094). More importantly, the expression level of some genes in EGRS has a significant correlation with the progression of LUAD clinicopathology, suggesting these genes might play an important role in the progression of LUAD. Finally, based on EGRS genes, we built and calibrated a nomogram for conveniently evaluating patients' outcomes.
Collapse
Affiliation(s)
- Linjun Liu
- Department of Biotechnology, College of Life Science & Chemistry, Beijing University of Technology, Chaoyang, Beijing, China
| | - Ke Xu
- NHC Key Laboratory of Biosafety, China CDC, National Institute for Viral Disease Control and Prevention, Beijing, China
| | - Yubai Zhou
- Department of Biotechnology, College of Life Science & Chemistry, Beijing University of Technology, Chaoyang, Beijing, China
| |
Collapse
|
12
|
Dynein Heavy Chain 64C Differentially Regulates Cell Survival and Proliferation of Wingless-Producing Cells in Drosophila melanogaster. J Dev Biol 2021; 9:jdb9040043. [PMID: 34698231 PMCID: PMC8544498 DOI: 10.3390/jdb9040043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022] Open
Abstract
Dynein is a multi-subunit motor protein that moves toward the minus-end of microtubules, and plays important roles in fly development. We identified Dhc64Cm115, a new mutant allele of the fly Dynein heavy chain 64C (Dhc64C) gene whose heterozygotes survive against lethality induced by overexpression of Sol narae (Sona). Sona is a secreted metalloprotease that positively regulates Wingless (Wg) signaling, and promotes cell survival and proliferation. Knockdown of Dhc64C in fly wings induced extensive cell death accompanied by widespread and disorganized expression of Wg. The disrupted pattern of the Wg protein was due to cell death of the Wg-producing cells at the DV midline and overproliferation of the Wg-producing cells at the hinge in disorganized ways. Coexpression of Dhc64C RNAi and p35 resulted in no cell death and normal pattern of Wg, demonstrating that cell death is responsible for all phenotypes induced by Dhc64C RNAi expression. The effect of Dhc64C on Wg-producing cells was unique among components of Dynein and other microtubule motors. We propose that Dhc64C differentially regulates survival of Wg-producing cells, which is essential for maintaining normal expression pattern of Wg for wing development.
Collapse
|
13
|
Rose KWJ, Taye N, Karoulias SZ, Hubmacher D. Regulation of ADAMTS Proteases. Front Mol Biosci 2021; 8:701959. [PMID: 34268335 PMCID: PMC8275829 DOI: 10.3389/fmolb.2021.701959] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/16/2021] [Indexed: 01/01/2023] Open
Abstract
A disintegrin and metalloprotease with thrombospondin type I motifs (ADAMTS) proteases are secreted metalloproteinases that play key roles in the formation, homeostasis and remodeling of the extracellular matrix (ECM). The substrate spectrum of ADAMTS proteases can range from individual ECM proteins to entire families of ECM proteins, such as the hyalectans. ADAMTS-mediated substrate cleavage is required for the formation, remodeling and physiological adaptation of the ECM to the needs of individual tissues and organ systems. However, ADAMTS proteases can also be involved in the destruction of tissues, resulting in pathologies such as arthritis. Specifically, ADAMTS4 and ADAMTS5 contribute to irreparable cartilage erosion by degrading aggrecan, which is a major constituent of cartilage. Arthritic joint damage is a major contributor to musculoskeletal morbidity and the most frequent clinical indication for total joint arthroplasty. Due to the high sequence homology of ADAMTS proteases in their catalytically active site, it remains a formidable challenge to design ADAMTS isotype-specific inhibitors that selectively inhibit ADAMTS proteases responsible for tissue destruction without affecting the beneficial functions of other ADAMTS proteases. In vivo, proteolytic activity of ADAMTS proteases is regulated on the transcriptional and posttranslational level. Here, we review the current knowledge of mechanisms that regulate ADAMTS protease activity in tissues including factors that induce ADAMTS gene expression, consequences of posttranslational modifications such as furin processing, the role of endogenous inhibitors and pharmacological approaches to limit ADAMTS protease activity in tissues, which almost exclusively focus on inhibiting the aggrecanase activity of ADAMTS4 and ADAMTS5.
Collapse
Affiliation(s)
| | | | | | - Dirk Hubmacher
- Orthopaedic Research Laboratories, Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
14
|
Daniunaite K, Bakavicius A, Zukauskaite K, Rauluseviciute I, Lazutka JR, Ulys A, Jankevicius F, Jarmalaite S. Promoter Methylation of PRKCB, ADAMTS12, and NAALAD2 Is Specific to Prostate Cancer and Predicts Biochemical Disease Recurrence. Int J Mol Sci 2021; 22:ijms22116091. [PMID: 34198725 PMCID: PMC8201120 DOI: 10.3390/ijms22116091] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 01/19/2023] Open
Abstract
The molecular diversity of prostate cancer (PCa) has been demonstrated by recent genome-wide studies, proposing a significant number of different molecular markers. However, only a few of them have been transferred into clinical practice so far. The present study aimed to identify and validate novel DNA methylation biomarkers for PCa diagnosis and prognosis. Microarray-based methylome data of well-characterized cancerous and noncancerous prostate tissue (NPT) pairs was used for the initial screening. Ten protein-coding genes were selected for validation in a set of 151 PCa, 51 NPT, as well as 17 benign prostatic hyperplasia samples. The Prostate Cancer Dataset (PRAD) of The Cancer Genome Atlas (TCGA) was utilized for independent validation of our findings. Methylation frequencies of ADAMTS12, CCDC181, FILIP1L, NAALAD2, PRKCB, and ZMIZ1 were up to 91% in our study. PCa specific methylation of ADAMTS12, CCDC181, NAALAD2, and PRKCB was demonstrated by qualitative and quantitative means (all p < 0.05). In agreement with PRAD, promoter methylation of these four genes was associated with the transcript down-regulation in the Lithuanian cohort (all p < 0.05). Methylation of ADAMTS12, NAALAD2, and PRKCB was independently predictive for biochemical disease recurrence, while NAALAD2 and PRKCB increased the prognostic power of multivariate models (all p < 0.01). The present study identified methylation of ADAMTS12, NAALAD2, and PRKCB as novel diagnostic and prognostic PCa biomarkers that might guide treatment decisions in clinical practice.
Collapse
Affiliation(s)
- Kristina Daniunaite
- Life Sciences Center, Institute of Biosciences, Vilnius University, 10257 Vilnius, Lithuania; (K.D.); (I.R.); (J.R.L.)
| | - Arnas Bakavicius
- National Cancer Institute, 08660 Vilnius, Lithuania; (A.B.); (K.Z.); (A.U.); (F.J.)
- Centre of Urology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Kristina Zukauskaite
- National Cancer Institute, 08660 Vilnius, Lithuania; (A.B.); (K.Z.); (A.U.); (F.J.)
| | - Ieva Rauluseviciute
- Life Sciences Center, Institute of Biosciences, Vilnius University, 10257 Vilnius, Lithuania; (K.D.); (I.R.); (J.R.L.)
| | - Juozas Rimantas Lazutka
- Life Sciences Center, Institute of Biosciences, Vilnius University, 10257 Vilnius, Lithuania; (K.D.); (I.R.); (J.R.L.)
| | - Albertas Ulys
- National Cancer Institute, 08660 Vilnius, Lithuania; (A.B.); (K.Z.); (A.U.); (F.J.)
| | - Feliksas Jankevicius
- National Cancer Institute, 08660 Vilnius, Lithuania; (A.B.); (K.Z.); (A.U.); (F.J.)
- Centre of Urology, Vilnius University Hospital Santaros Klinikos, 08661 Vilnius, Lithuania
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Sonata Jarmalaite
- Life Sciences Center, Institute of Biosciences, Vilnius University, 10257 Vilnius, Lithuania; (K.D.); (I.R.); (J.R.L.)
- National Cancer Institute, 08660 Vilnius, Lithuania; (A.B.); (K.Z.); (A.U.); (F.J.)
- Correspondence: ; Tel.: +370-5-2190901; Fax: +370-5-2720164
| |
Collapse
|
15
|
Mohamedi Y, Fontanil T, Cal S, Cobo T, Obaya ÁJ. ADAMTS-12: Functions and Challenges for a Complex Metalloprotease. Front Mol Biosci 2021; 8:686763. [PMID: 33996918 PMCID: PMC8119882 DOI: 10.3389/fmolb.2021.686763] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Nineteen members of the ADAMTS family of secreted zinc metalloproteinases are present in the human degradome. A wide range of different functions are being attributed to these enzymes and the number of their known substrates is considerably increasing in recent years. ADAMTSs can participate in processes such as fertility, inflammation, arthritis, neuronal and behavioral disorders, as well as cancer. Since its first annotation in 2001, ADAMTS-12 has been described to participate in different processes displayed by members of this family of proteinases. In this sense, ADAMTS-12 performs essential roles in modulation and recovery from inflammatory processes such as colitis, endotoxic sepsis and pancreatitis. ADAMTS-12 has also been involved in cancer development acting either as a tumor suppressor or as a pro-tumoral agent. Furthermore, participation of ADAMTS-12 in arthritis or in neuronal disorders has also been suggested through degradation of components of the extracellular matrix. In addition, ADAMTS-12 proteinase activity can also be modified by interaction with other proteins and thus, can be an alternative way of modulating ADAMTS-12 functions. In this review we revised the most relevant findings about ADAMTS-12 function on the 20th anniversary of its identification.
Collapse
Affiliation(s)
- Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain
| | - Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain.,Departamento de Investigación, Instituto Ordóñez, Oviedo, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, Oviedo, Spain.,Instituto Asturiano de Odontología, Oviedo, Spain
| | - Álvaro J Obaya
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain.,Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
16
|
Identification of crucial long non-coding RNAs and mRNAs along with related regulatory networks through microarray analysis in esophageal carcinoma. Funct Integr Genomics 2021; 21:377-391. [PMID: 33864185 DOI: 10.1007/s10142-021-00784-x] [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: 04/27/2020] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 02/01/2023]
Abstract
Esophageal carcinoma (EC) is a tremendous threat to human health and life worldwide. Long non-coding RNAs (lncRNAs) have been identified as crucial players in carcinomas including EC. An in-depth understanding on regulatory networks of lncRNAs contributes to the better management of EC. In this text, 2052 lncRNAs and 3240 mRNAs were found to be differentially expressed in 5 EC tumor tissues versus adjacent normal tissues by microarray analysis. Moreover, 297 carcinoma-related genes were screened out according to pathway and disease annotation analyses. In addition, 410 potential lncRNA-mRNA cis-regulation pairs and 395 lncRNA-mRNA trans-regulation pairs were screened out. Among these genes, 14 trans-regulated and 19 cis-regulated genes were found to be related with carcinomas. Additionally, 42 possible lncRNA-mRNA trans-regulation pairs and 26 cis-regulation pairs were found to be related with carcinomas. Also, 4 differentially expressed transcription factors in EC and lncRNAs possibly regulated by these transcription factors were screened out. Moreover, plenty of common upregulated or downregulated lncRNAs and mRNAs in EC were identified by comparative analysis for our microarray outcomes and previous high-throughput data. Furthermore, we demonstrated that ENST00000437781.1 knockdown inhibited cell proliferation and facilitated cell apoptosis by downregulating SIX homeobox 4 (SIX4) and ENST00000524987.1 knockdown had no influence on anoctamin 1 calcium activated chloride channel (ANO1) expression in EC cells. In conclusion, we identified some crucial lncRNAs and genes along with potential regulatory networks of lncRNAs/genes, deepening our understanding on pathogenesis of EC.
Collapse
|
17
|
Rabadán R, Mohamedi Y, Rubin U, Chu T, Alghalith AN, Elliott O, Arnés L, Cal S, Obaya ÁJ, Levine AJ, Cámara PG. Identification of relevant genetic alterations in cancer using topological data analysis. Nat Commun 2020; 11:3808. [PMID: 32732999 PMCID: PMC7393176 DOI: 10.1038/s41467-020-17659-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 07/09/2020] [Indexed: 01/05/2023] Open
Abstract
Large-scale cancer genomic studies enable the systematic identification of mutations that lead to the genesis and progression of tumors, uncovering the underlying molecular mechanisms and potential therapies. While some such mutations are recurrently found in many tumors, many others exist solely within a few samples, precluding detection by conventional recurrence-based statistical approaches. Integrated analysis of somatic mutations and RNA expression data across 12 tumor types reveals that mutations of cancer genes are usually accompanied by substantial changes in expression. We use topological data analysis to leverage this observation and uncover 38 elusive candidate cancer-associated genes, including inactivating mutations of the metalloproteinase ADAMTS12 in lung adenocarcinoma. We show that ADAMTS12-/- mice have a five-fold increase in the susceptibility to develop lung tumors, confirming the role of ADAMTS12 as a tumor suppressor gene. Our results demonstrate that data integration through topological techniques can increase our ability to identify previously unreported cancer-related alterations.
Collapse
Affiliation(s)
- Raúl Rabadán
- Departments of Systems Biology and Biomedical Informatics, Columbia University, 1130 St. Nicholas Ave., New York, NY, 10032, USA.
| | - Yamina Mohamedi
- Departamento de Bioquimica y Biologia Molecular, Universidad de Oviedo, Oviedo, Asturias, Spain
- IUOPA, Instituto Universitario de Oncologia, Oviedo, Asturias, Spain
| | - Udi Rubin
- Departments of Systems Biology and Biomedical Informatics, Columbia University, 1130 St. Nicholas Ave., New York, NY, 10032, USA
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Tim Chu
- Departments of Systems Biology and Biomedical Informatics, Columbia University, 1130 St. Nicholas Ave., New York, NY, 10032, USA
| | - Adam N Alghalith
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Oliver Elliott
- Departments of Systems Biology and Biomedical Informatics, Columbia University, 1130 St. Nicholas Ave., New York, NY, 10032, USA
| | - Luis Arnés
- Departments of Systems Biology and Biomedical Informatics, Columbia University, 1130 St. Nicholas Ave., New York, NY, 10032, USA
| | - Santiago Cal
- Departamento de Bioquimica y Biologia Molecular, Universidad de Oviedo, Oviedo, Asturias, Spain
- IUOPA, Instituto Universitario de Oncologia, Oviedo, Asturias, Spain
| | - Álvaro J Obaya
- IUOPA, Instituto Universitario de Oncologia, Oviedo, Asturias, Spain
- Departamento de Biologia Funcional, Universidad de Oviedo, Oviedo, Asturias, Spain
| | - Arnold J Levine
- The Simons Center for Systems Biology, Institute for Advanced Study, Princeton, NJ, 08540, USA.
| | - Pablo G Cámara
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA.
| |
Collapse
|
18
|
Li L, Yuan S, Zhao X, Luo T. ADAMTS8 is frequently down-regulated in colorectal cancer and functions as a tumor suppressor. Biochem Biophys Res Commun 2020; 524:663-671. [DOI: 10.1016/j.bbrc.2020.01.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/06/2020] [Indexed: 01/06/2023]
|
19
|
Li C, Luo X, Huang B, Wang X, Deng Y, Zhong Z. ADAMTS12 acts as a cancer promoter in colorectal cancer via activating the Wnt/β-catenin signaling pathway in vitro. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:301. [PMID: 32355745 PMCID: PMC7186627 DOI: 10.21037/atm.2020.02.154] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/10/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND ADAMTS12, a member of the ADAMTS family, is reported to be associated with the clinic outcome of colorectal cancer (CRC) patients. However, the functions and precise mechanism in CRC progression have yet to be fully understood. METHODS By analyzing The Cancer Genome Atlas (TCGA) database, we examined the mRNA level of ADAMTS12 and assessed the prognostic value of ADAMTS12 in CRC patients using a tissue microarray containing 41 CRC patient samples. Cell Counting Kit-8 (CCK-8), colony formation, and transwell assays were used to quantify the impact of ADAMTS12 on cell proliferation and migration in ADAMTS12-overexpressing and ADAMTS12-deficient cell lines. The signaling pathways that ADAMTS12 mediated were identified by dual-luciferase reporter assays, and confirmed by western blotting and quantitative teal-time polymerase chain reaction (qRT-PCR). RESULTS The ADAMTS12 mRNA level was upregulated in CRC tissues, and CRC patients with a high level of ADAMTS12 showed worse prognosis when compared with the patients with a low level of ADAMTS12. In vitro functional assays demonstrated that overexpression of ADAMTS12 significantly boosted cell proliferation and migration while ADAMTS12 deficiency remarkably impaired both tumor cell behaviors. Mechanical studies further verified that ADAMTS12 overexpression enhanced the transcriptional activity of β-catenin in the Wnt/β-catenin signaling pathway. In the ADAMTS12-deficient context, the downstream gene expression of myc and cyclin D1 was significantly reduced compared with that in wild-type cancer cells. CONCLUSIONS ADAMTS12 fulfills the tumor-promotor role by activating Wnt/β-catenin signaling pathway in colon cells and may represent a new option in CRC target treatment.
Collapse
Affiliation(s)
- Chunxue Li
- Department of General Surgery, Gastric and Colorectal Surgery Division, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xuelian Luo
- Cancer Center, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Bin Huang
- Department of General Surgery, Gastric and Colorectal Surgery Division, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiangfeng Wang
- Department of General Surgery, Gastric and Colorectal Surgery Division, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yi Deng
- Cancer Center, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhaoyang Zhong
- Cancer Center, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
20
|
Fontanil T, Mohamedi Y, Cobo T, Cal S, Obaya ÁJ. Novel Associations Within the Tumor Microenvironment: Fibulins Meet ADAMTSs. Front Oncol 2019; 9:796. [PMID: 31508361 PMCID: PMC6714394 DOI: 10.3389/fonc.2019.00796] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/06/2019] [Indexed: 01/08/2023] Open
Abstract
The maintenance of tissue homeostasis in any organism is a very complex and delicate process in which numerous factors intervene. Cellular homeostasis not only depends on intrinsic factors but also relies on external factors that compose the microenvironment or cellular niche. Thus, extracellular matrix (ECM) components play a very important role in maintaining cell survival and behavior, and alterations in the ECM composition can lead to different pathologies. Fibulins and ADAMTS metalloproteases play crucial roles in the upkeep and function of the ECM in different tissues. In fact, members of both of these families of secreted multidomain proteins can interact with numerous other ECM components and thus shape or regulate the molecular environment. Individual members of both families have been implicated in tumor-related processes by exhibiting either pro- or antitumor properties. Recent studies have shown both an important relation among members of both families and their participation in several pathologies, including cardiogenesis or cancer. In this review, we summarize the associations among fibulins and ADAMTSs and the effects elicited by those interactions on cellular behavior.
Collapse
Affiliation(s)
- Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain.,Departamento de Investigación, Instituto Órdoñez, Oviedo, Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain
| | - Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Instituto Asturiano de Odontología, Universidad de Oviedo, Oviedo, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain
| | - Álvaro J Obaya
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Oviedo, Spain.,Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
21
|
Li X, Xiao X, Chang R, Zhang C. Comprehensive bioinformatics analysis identifies lncRNA HCG22 as a migration inhibitor in esophageal squamous cell carcinoma. J Cell Biochem 2019; 121:468-481. [PMID: 31236983 DOI: 10.1002/jcb.29218] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022]
Abstract
Esophageal cancer is one of the most lethal malignancies worldwide, and esophageal squamous cell carcinoma (ESCC) is the dominant histological type. However, the long noncoding RNA (lncRNA) alterations in ESCC have not been elucidated to date. In this study, reliable databases from Gene Expression Omnibus (GEO), which analyzed lncRNA expression in ESCC tumor tissues and adjacent normal tissues were searched, and common differentially expressed lncRNAs and genes were analyzed. Next, cis- trans analysis was performed to predict the underlying relationships between altered lncRNAs and mRNAs, and the lncRNA-mRNA regulatory network was established. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of altered lncRNA-related genes were performed. The promising lncRNA HCG22 was validated by quantitative polymerase chain reaction (qPCR), and clinicopathological data were collected to identify the relationship between lncRNA HCG22 expression level and clinical features. Finally, Transwell assays were performed to explore the biological functions of lncRNA HCG22 in ESCC cells. Two hundred forty-one lncRNAs and 835 mRNAs were observed to be remarkably altered between ESCC tumor tissues and adjacent normal tissues. The lncRNA-mRNA regulatory network showed the coexpression association between lncRNA HCG22 and SPINK7 and ADAMTS12. GO and KEGG analyses showed that HCG22 and ADAMTS12 had potential biological functions in the cell migration of ESCC. The downregulation of lncRNA HCG22 in ESCC tumor tissues was validated by qPCR, and the clinicopathological data showed a noticeable correlation between lncRNA HCG22 expression level and the ESCC differentiational degree and clinical TNM stage. Kaplan-Meier analysis showed that patients with ESCC having low lncRNA HCG22 expression in ESCC tissues had considerably shorter overall survival compared with patients with ESCC having high lncRNA HCG22 expression. Following Transwell assays confirmed the migratory role of lncRNA HCG22 in ESCC cells. In conclusion, lncRNA HCG22 was downregulated in ESCC tissues and can be a migration inhibitor of ESCC cells, and SPINK7 and ADAMTS12 are promising to be the regulatory targets of lncRNA HCG22.
Collapse
Affiliation(s)
- Xizhe Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxiong Xiao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruimin Chang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
22
|
Mani S, Ghosh J, Lan Y, Senapati S, Ord T, Sapienza C, Coutifaris C, Mainigi M. Epigenetic changes in preterm birth placenta suggest a role for ADAMTS genes in spontaneous preterm birth. Hum Mol Genet 2019; 28:84-95. [PMID: 30239759 DOI: 10.1093/hmg/ddy325] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/13/2018] [Indexed: 12/28/2022] Open
Abstract
Preterm birth (PTB) affects approximately 1 in 10 pregnancies and contributes to approximately 50% of neonatal mortality. However, despite decades of research, little is understood about the etiology of PTB, likely due to the multifactorial nature of the disease. In this study, we examined preterm and term placentas, from unassisted conceptions and those conceived using in vitro fertilization (IVF). IVF increases the risk of PTB and causes epigenetic change in the placenta and fetus; therefore, we utilized these patients as a unique population with a potential common etiology. We investigated genome-wide DNA methylation in placentas from term IVF, preterm IVF, term control (unassisted conception) and preterm control pregnancies and discovered epigenetic dysregulation of multiple genes involved in cell migration, including members of the ADAMTS family, ADAMTS12 and ADAMTS16. These genes function in extracellular matrix regulation and tumor cell invasion, processes replicated by invasive trophoblasts (extravillous trophoblasts (EVTs)) during early placentation. Though expression was similar between term and preterm placentas, we found that both genes demonstrate high expression in first- and second-trimester placenta, specifically in EVTs and syncytiotrophoblasts. When we knocked down ADAMTS12 or ADAMTS16in vitro, there was poor EVT invasion and reduced matrix metalloproteinase activity, reinforcing their critical role in placentation. In conclusion, utilizing a population at high risk for PTB, we have identified a role for ADAMTS gene methylation in regulating early placentation and susceptibility to PTB.
Collapse
Affiliation(s)
- Sneha Mani
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jayashri Ghosh
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, USA
| | - Yemin Lan
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Suneeta Senapati
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Teri Ord
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Carmen Sapienza
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, USA
| | - Christos Coutifaris
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Monica Mainigi
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
23
|
Loss of Forkhead Box O3 Facilitates Inflammatory Colon Cancer: Transcriptome Profiling of the Immune Landscape and Novel Targets. Cell Mol Gastroenterol Hepatol 2018; 7:391-408. [PMID: 30718226 PMCID: PMC6360252 DOI: 10.1016/j.jcmgh.2018.10.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 10/03/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Diminished forkhead box O3 (FOXO3) function drives inflammation and cancer growth; however, mechanisms fostering these pathobiologies are unclear. Here, we aimed to identify in colon loss of FOXO3-dependent cellular and molecular changes that facilitate inflammation-mediated tumor growth. METHODS FOXO3 knockout (KO) and wild-type (WT) mice were used in the AOM/DSS model of inflammation-mediated colon cancer. Bioinformatics were used for profiling of mRNA sequencing data from human and mouse colon and tumors; specific targets were validated in human colon cancer cells (shFOXO3). RESULTS In mice, FOXO3 deficiency led to significantly elevated colonic tumor burden (incidence and size) compared with WT (P < .05). In FOXO3 KO colon, activated molecular pathways overlapped with those associated with mouse and human colonic inflammation and cancer, especially human colonic tumors with inflammatory microsatellite instability (false discovery rate < 0.05). FOXO3 KO colon, similar to tumors, had increased neutrophils, macrophages, B cells, T cells, and decreased natural killer cells (false discovery rate < 0.05). Moreover, in KO colon differentially expressed transcripts were linked to activation of inflammatory nuclear factor kappa B, tumorigenic cMyc, and bacterial Toll-like receptor signaling. Among differentially expressed transcripts, we validated altered expression of integrin subunit alpha 2 (ITGA2), ADAM metallopeptidase with thrombospondin type 1 motif 12, and ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 5 in mouse WT and FOXO3 KO colon and tumors (P < .05). Similarly, their altered expression was found in human inflammatory bowel disease and colon cancer tissues and linked to poor patient survival. Ultimately, in human colon cancer cells, FOXO3 knockdown (shFOXO3) led to significantly increased ITGA2, and silencing ITGA2 (siRNA) alone diminished cell growth. CONCLUSIONS We identified the loss of FOXO3-mediated immune landscape, pathways, and transcripts that could serve as biomarkers and new targets for inflammatory colon cancer treatment.
Collapse
|
24
|
Wei JL, Fu W, Hettinghouse A, He WJ, Lipson KE, Liu CJ. Role of ADAMTS-12 in Protecting Against Inflammatory Arthritis in Mice By Interacting With and Inactivating Proinflammatory Connective Tissue Growth Factor. Arthritis Rheumatol 2018; 70:1745-1756. [PMID: 29750395 DOI: 10.1002/art.40552] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 05/03/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE It has been reported that ADAMTS-12 is a susceptibility gene for rheumatoid arthritis (RA) development, and its level is significantly increased in RA patients. In addition, ADAMTS-12 is reported to be required for inflammation in otherwise healthy subjects. This study was undertaken to determine the role of ADAMTS-12 and the underlying mechanisms in the pathogenesis of inflammatory arthritis. METHODS The collagen-induced arthritis (CIA) model was established in ADAMTS-12-deficient mice and their control littermates to determine the role of ADAMTS-12 in vivo. Micro-computed tomography scanning was used to demonstrate the destruction of the ankle joint; histologic analysis illustrated synovitis, pannus formation, and bone and cartilage destruction; enzyme-linked immunosorbent assay was performed to measure serum levels of inflammatory cytokines; and protein-protein interaction assays were performed to detect the interactions of ADAMTS-12 and its various deletion mutants with connective tissue growth factor (CTGF). RESULTS Deficiency of ADAMTS-12 led to accelerated inflammatory arthritis in the CIA mouse model. Loss of ADAMTS-12 caused enhanced osteoclastogenesis. In vitro and in vivo protein-protein interaction assays demonstrated that ADAMTS-12 bound and processed CTGF, a previously unrecognized substrate of ADAMTS-12. In addition, deletion of ADAMTS-12 enhanced, while overexpression of ADMATS-12 reduced, CTGF-mediated inflammation. Furthermore, ADAMTS-12 regulation of inflammation was largely lost in CTGF-deficient macrophages. Importantly, blocking of CTGF attenuated elevated inflammatory arthritis seen in the ADAMTS-12-deficient CIA mouse model. CONCLUSION This study provides evidence that ADAMTS-12 is a critical regulator of inflammatory arthritis and that this is mediated, at least in part, through control of CTGF turnover.
Collapse
Affiliation(s)
- Jian-Lu Wei
- New York University Medical Center, New York, New York, and Shandong University Qilu Hospital, Jinan, China
| | - Wenyu Fu
- New York University Medical Center, New York, New York
| | | | - Wen-Jun He
- New York University Medical Center, New York, New York
| | | | - Chuan-Ju Liu
- New York University Medical Center and New York University School of Medicine, New York, New York
| |
Collapse
|
25
|
Kordowski F, Kolarova J, Schafmayer C, Buch S, Goldmann T, Marwitz S, Kugler C, Scheufele S, Gassling V, Németh CG, Brosch M, Hampe J, Lucius R, Röder C, Kalthoff H, Siebert R, Ammerpohl O, Reiss K. Aberrant DNA methylation of ADAMTS16 in colorectal and other epithelial cancers. BMC Cancer 2018; 18:796. [PMID: 30081852 PMCID: PMC6080380 DOI: 10.1186/s12885-018-4701-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND ADAMs (a disintegrin and metalloproteinase) have long been associated with tumor progression. Recent findings indicate that members of the closely related ADAMTS (ADAMs with thrombospondin motifs) family are also critically involved in carcinogenesis. Gene silencing through DNA methylation at CpG loci around e.g. transcription start or enhancer sites is a major mechanism in cancer development. Here, we aimed at identifying genes of the ADAM and ADAMTS family showing altered DNA methylation in the development or colorectal cancer (CRC) and other epithelial tumors. METHODS We investigated potential changes of DNA methylation affecting ADAM and ADAMTS genes in 117 CRC, 40 lung cancer (LC) and 15 oral squamous-cell carcinoma (SCC) samples. Tumor tissue was analyzed in comparison to adjacent non-malignant tissue of the same patients. The methylation status of 1145 CpGs in 51 ADAM and ADAMTS genes was measured with the HumanMethylation450 BeadChip Array. ADAMTS16 protein expression was analyzed in CRC samples by immunohistochemistry. RESULTS In CRC, we identified 72 CpGs in 18 genes which were significantly affected by hyper- or hypomethylation in the tumor tissue compared to the adjacent non-malignant tissue. While notable/frequent alterations in methylation patterns within ADAM genes were not observed, conspicuous changes were found in ADAMTS16 and ADAMTS2. To figure out whether these differences would be CRC specific, additional LC and SCC tissue samples were analyzed. Overall, 78 differentially methylated CpGs were found in LC and 29 in SCC. Strikingly, 8 CpGs located in the ADAMTS16 gene were commonly differentially methylated in all three cancer entities. Six CpGs in the promoter region were hypermethylated, whereas 2 CpGs in the gene body were hypomethylated indicative of gene silencing. In line with these findings, ADAMTS16 protein was strongly expressed in globlet cells and colonocytes in control tissue but not in CRC samples. Functional in vitro studies using the colorectal carcinoma cell line HT29 revealed that ADAMTS16 expression restrained tumor cell proliferation. CONCLUSIONS We identified ADAMTS16 as novel gene with cancer-specific promoter hypermethylation in CRC, LC and SCC patients implicating ADAMTS16 as potential biomarker for these tumors. Moreover, our results provide evidence that ADAMTS16 may have tumor suppressor properties.
Collapse
Affiliation(s)
- Felix Kordowski
- Department of Dermatology and Allergology, University Hospital Schleswig-Holstein, University of Kiel, Rosalind-Franklin-Straße 7, 24105 Kiel, Germany
| | - Julia Kolarova
- Institute of Human Genetics, University of Kiel, Kiel, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Clemens Schafmayer
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Stephan Buch
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Torsten Goldmann
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
- Research Center Borstel, Leibniz Center for Medicine and Biosciences, Borstel, Germany
| | - Sebastian Marwitz
- Pathology of the University Medical Center Schleswig-Holstein, Campus Luebeck, Lübeck, Germany
- Research Center Borstel, Leibniz Center for Medicine and Biosciences, Borstel, Germany
| | - Christian Kugler
- Thoracic Surgery, LungenClinic Grosshansdorf, Grosshansdorf, Germany
| | | | - Volker Gassling
- Department of Oral and Maxillofacial Surgery, University of Kiel, Kiel, Germany
| | | | - Mario Brosch
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Jochen Hampe
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ralph Lucius
- Anatomical Institute, University of Kiel, Kiel, Germany
| | - Christian Röder
- Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Reiner Siebert
- Institute of Human Genetics, University of Kiel, Kiel, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Ole Ammerpohl
- Institute of Human Genetics, University of Kiel, Kiel, Germany
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Karina Reiss
- Department of Dermatology and Allergology, University Hospital Schleswig-Holstein, University of Kiel, Rosalind-Franklin-Straße 7, 24105 Kiel, Germany
| |
Collapse
|
26
|
Zhang L, Cao H, He T, Yang J, Tao H, Wang Y, Hu Q. Overexpression of PRDM13 inhibits glioma cells via Rho and GTP enzyme activation protein. Int J Mol Med 2018; 42:966-974. [PMID: 29767251 PMCID: PMC6034930 DOI: 10.3892/ijmm.2018.3679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/10/2018] [Indexed: 11/17/2022] Open
Abstract
PR (PRDI-BFI and RIZ) domain containing (PRDM) proteins have been shown to be important in several types of human cancer. PRDM13, a member of the PRDM family, contains transcriptional regulators involved in modulating several cellular processes. However, the function of PRDM13 in glioma remains to be elucidated. The purpose of the present study was to evaluate the expression and effect of PRDM13 on glioma cells. It was found that the expression of PRDM13 was reduced in glioma cells, and the overexpression of PRDM13 significantly decreased the proliferation, migration and invasion of U87 glioma cells. Through validation of RNA-sequencing analysis, genes regulating cell proliferation and migration were classified from Gene Ontology sources. In addition, PRDM13 was shown to be associated with Rho protein and GTP enzyme activation protein. The over expression of PRDM13 upregulated deleted in liver cancer 1 (DLC1) to inhibit the proliferation and invasion of U87 cells. In conclusion, PRDM13 decreased the proliferation and invasion of U87 cells, and may be of potential value for glioma therapy.
Collapse
Affiliation(s)
- Linna Zhang
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Huimei Cao
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Tao He
- Department of Spinal Surgery, Dongfeng General Hospital of Chinese Medicine Affiliated to Hubei Medical University, Shiyan, Hubei 442000, P.R. China
| | - Jijuan Yang
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Hong Tao
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yin Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Basic Medical School of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Qikuan Hu
- Department of Physiology, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
27
|
Li J, Liao Y, Huang J, Sun Y, Chen H, Chen C, Li S, Yang Z. Epigenetic silencing of ADAMTS5 is associated with increased invasiveness and poor survival in patients with colorectal cancer. J Cancer Res Clin Oncol 2018; 144:215-227. [PMID: 29143120 DOI: 10.1007/s00432-017-2545-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/07/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE A disintegrin and metalloprotease with motif 5(ADAMTS5) has been involved in colorectal cancer (CRC) with hypermethylation in the promoter. However, its role in CRC remains unclear. The aim of this study was to explore the clinical significance and biological effect of ADAMTS5 on colorectal carcinogenesis. Through MSP, qRT-PCR, WB and IHC analysis, followed by a variety of in vitro assays, we report the function of ADAMTS5 in CRC. ADAMTS5 was markedly hypermethylaed and downregulated in tumor tissues compared with non-tumor tissues (p < 0.001). Negative expression of ADAMTS5 was much more common in tumor tissues than that in normal tissues (p < 0.001) and correlated with histologic types (p = 0.002), poor OS (p = 0.029) and DFS (p = 0.018). In vitro assay revealed that overexpression of ADAMTS5 inhibited the capabilities of migration and invasion of CRC cells, and no effect on cell growth, cell cycle and apoptosis. ADAMTS5 is hypermethylated and inhibits cancer cells invasion and migration in colorectal cancer, and correlates with OS and DFS, indicating that ADAMTS5 might be a useful biomarker in colorectal cancer therapy.
Collapse
Affiliation(s)
- Jizhen Li
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-Sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China
- Department of General Surgery, The Zhuhai People's Hospital, 79 Kangning Road, Zhuhai, 519000, People's Republic of China
| | - Yi Liao
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-Sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China
- Guangdong Institute of Gastroenterology, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangzhou, People's Republic of China
| | - Jintuan Huang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-Sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China
- Guangdong Institute of Gastroenterology, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangzhou, People's Republic of China
| | - Yi Sun
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-Sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China
- Guangdong Institute of Gastroenterology, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangzhou, People's Republic of China
| | - Hao Chen
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-Sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China
- Guangdong Institute of Gastroenterology, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangzhou, People's Republic of China
| | - Chunyu Chen
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-Sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China
- Guangdong Institute of Gastroenterology, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangzhou, People's Republic of China
| | - Senmao Li
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-Sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China
- Guangdong Institute of Gastroenterology, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangzhou, People's Republic of China
| | - Zuli Yang
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-Sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.
- Guangdong Institute of Gastroenterology, Guangzhou, People's Republic of China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, Guangzhou, People's Republic of China.
| |
Collapse
|
28
|
Fontanil T, Álvarez-Teijeiro S, Villaronga MÁ, Mohamedi Y, Solares L, Moncada-Pazos A, Vega JA, García-Suárez O, Pérez-Basterrechea M, García-Pedrero JM, Obaya AJ, Cal S. Cleavage of Fibulin-2 by the aggrecanases ADAMTS-4 and ADAMTS-5 contributes to the tumorigenic potential of breast cancer cells. Oncotarget 2017; 8:13716-13729. [PMID: 28099917 PMCID: PMC5355132 DOI: 10.18632/oncotarget.14627] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 01/04/2017] [Indexed: 11/30/2022] Open
Abstract
Fibulin-2 participates in the assembly of extracellular matrix components through interactions with multiple ligands and promotes contacts between cells and their surrounding environment. Consequently, identification of processes that could lead to an altered Fibulin-2 could have a major impact not only in the maintenance of tissue architecture and morphogenesis but also in pathological situations including cancer. Herein, we have investigated the ability of the secreted metalloproteases ADAMTS-4 and ADAMTS-5 to digest Fibulin-2. Using in vitro approaches and cultured breast cancer cell lines we demonstrate that Fibulin-2 is a better substrate for ADAMTS-5 than it is for ADAMTS-4. Moreover, Fibulin-2 degradation is associated to an enhancement of the invasive potential of T47D, MCF-7 and SK-BR-3 cells. We have also found that conditioned medium from MCF-7 cells that simultaneously overexpress Fibulin-2 and ADAMTS-5 significantly induced the migratory and invasive ability of normal breast fibroblasts using 3D collagen matrices. Immunohistochemical analysis highlights the close proximity or partial overlap of both Fibulin-2 and ADAMTS-5 in breast tumor samples. Additionally, proteolytic products derived from a potential degradation of Fibulin-2 by ADAMTS-5 were also identified in these samples. Finally, we also show that the cleavage of Fibulin-2 by ADAMTS-5 is counteracted by ADAMTS-12, a metalloprotease that interacts with Fibulin-2. Overall, our results provide direct evidence indicating that Fibulin-2 is a novel substrate of ADAMTS-5 and that this proteolysis could alter the cellular microenvironment affecting the balance between protumor and antitumor effects associated to both Fibulin-2 and the ADAMTSs metalloproteases.
Collapse
Affiliation(s)
- Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain
| | - Saúl Álvarez-Teijeiro
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Hospital Universitario Central de Asturias, Universidad de Oviedo, Asturias, and CIBERONC, Madrid, Spain
| | - M Ángeles Villaronga
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Hospital Universitario Central de Asturias, Universidad de Oviedo, Asturias, and CIBERONC, Madrid, Spain
| | - Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain
| | - Laura Solares
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain
| | - Angela Moncada-Pazos
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - José A Vega
- Department of Morphology and Cellular Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Olivia García-Suárez
- Department of Morphology and Cellular Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
| | - Marcos Pérez-Basterrechea
- Unidad de Trasplantes, Terapia Celular y Medicina Regenerativa, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Juana M García-Pedrero
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Hospital Universitario Central de Asturias, Universidad de Oviedo, Asturias, and CIBERONC, Madrid, Spain
| | - Alvaro J Obaya
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Departamento de Biología Funcional, Area de Fisiología, Universidad de Oviedo, Asturias, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain
| |
Collapse
|
29
|
Alterations in ADAMTS12 gene expression in salivary glands of radioiodine-131-administered rats. Nucl Med Commun 2017; 37:1010-5. [PMID: 27295306 DOI: 10.1097/mnm.0000000000000556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of this study was to evaluate the alterations in ADAMTS12 expression after radioiodine-131 (RAI)-induced salivary gland damage. MATERIALS AND METHODS A total of 30 Wistar male albino rats (260±45 g, 6 months old) were studied for ADAMTS12 gene expression levels and histological changes in the parotid and submandibular salivary glands of rats after the administration of RAI. A series of healthy rats were used as controls. A 3 mCi (111 MBq) dose of RAI was administered to rats in group 1 (n=6), group 2 (n=6), group 3 (n=6), and group 4 (n=6) to induce salivary gland damage. Evaluations were performed at 24 h in controls and at 4, 24 h, 7, and 30 days after the administration of RAI. Quantitative and statistical analyses were carried out. RESULTS In RAI-administered groups, the mean values of ADAMTS12 gene expression showed a distinct suppression over time for the parotid gland (groups 1-4: 0.38, 0.11, 0.10, and 0.18, respectively; P<0.05), but the values remained similar over time for the submandibular gland (groups 1-4: 1.59, 1.57, 1.03, and 1.00, respectively; P>0.05) compared with the controls. Histological evaluation indicated that RAI-administered groups had significant common nuclear coarsening and focal subnuclear vacuolization, but not in the control samples. Histological changes were more prominent in the parotid gland samples. CONCLUSION Alterations in ADAMTS12 gene expression may play a role in RAI-induced salivary gland damage in rats.
Collapse
|
30
|
Binder MJ, McCoombe S, Williams ED, McCulloch DR, Ward AC. The extracellular matrix in cancer progression: Role of hyalectan proteoglycans and ADAMTS enzymes. Cancer Lett 2016; 385:55-64. [PMID: 27838414 DOI: 10.1016/j.canlet.2016.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/28/2016] [Accepted: 11/01/2016] [Indexed: 01/08/2023]
Abstract
Remodelling of the extracellular matrix (ECM) has emerged as a key factor in cancer progression. Proteoglycans, including versican and other hyalectans, represent major structural elements of the ECM where they interact with other important molecules, including the glycosaminoglycan hyaluronan and the CD44 cell surface receptor. The hyalectan proteoglycans are regulated through cleavage by the proteolytic actions of A Disintegrin-like And Metalloproteinase domain with Thrombospondin-1 motif (ADAMTS) family members. Alteration in the balance between hyalectan proteoglycans and ADAMTS enzymes has been proposed to be a crucial factor in cancer progression either in a positive or negative manner depending on the context. Further complexity arises due to the formation of bioactive cleavage products, such as versikine, which may also play a role, and non-enzymatic functions for ADAMTS proteins. This research is providing fresh insights into cancer biology and opportunities for the development of new diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Marley J Binder
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Scott McCoombe
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre, Institute of Health and Biomedical Innovation, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland 4000, Australia
| | - Daniel R McCulloch
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia; Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Waurn Ponds, Victoria 3216, Australia; Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria 3216, Australia.
| |
Collapse
|
31
|
Sol narae (Sona) is a Drosophila ADAMTS involved in Wg signaling. Sci Rep 2016; 6:31863. [PMID: 27535473 PMCID: PMC4989167 DOI: 10.1038/srep31863] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/28/2016] [Indexed: 01/06/2023] Open
Abstract
ADAMTS (a disintegrin and metalloproteases with thrombospondin motif) family consists of secreted proteases, and is shown to cleave extracellular matrix proteins. Their malfunctions result in cancers and disorders in connective tissues. We report here that a Drosophila ADAMTS named Sol narae (Sona) promotes Wnt/Wingless (Wg) signaling. sona loss-of-function mutants are lethal and rare escapers had malformed appendages, indicating that sona is essential for fly development and survival. sona exhibited positive genetic interaction with wntless (wls) that encodes a cargo protein for Wg. Loss of sona decreased the level of extracellular Wg, and also reduced the expression level of Wg effector proteins such as Senseless (Sens), Distalless (Dll) and Vestigial (Vg). Sona and Wg colocalized in Golgi and endosomal vesicles, and were in the same protein complex. Furthermore, co-expression of Wg and Sona generated ectopic wing margin bristles. This study suggests that Sona is involved in Wg signaling by regulating the level of extracellular Wg.
Collapse
|
32
|
Pérez-García S, Gutiérrez-Cañas I, Seoane IV, Fernández J, Mellado M, Leceta J, Tío L, Villanueva-Romero R, Juarranz Y, Gomariz RP. Healthy and Osteoarthritic Synovial Fibroblasts Produce a Disintegrin and Metalloproteinase with Thrombospondin Motifs 4, 5, 7, and 12: Induction by IL-1β and Fibronectin and Contribution to Cartilage Damage. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2449-61. [PMID: 27449198 DOI: 10.1016/j.ajpath.2016.05.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 12/22/2022]
Abstract
Current description of osteoarthritis includes the involvement of synovial inflammation. Studies contributing to understanding the mechanisms of cross-talk and feedback among the joint tissues could be relevant to the development of therapies that block disease progression. During osteoarthritis, synovial fibroblasts exposed to anomalous mechanical forces and an inflammatory microenvironment release factors such as a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) metalloproteinases that mediate tissue damage and perpetuate inflammation. We therefore studied the production of ADAMTS by synovial fibroblasts and their contribution to cartilage degradation. Moreover, we analyzed the implication of two mediators present in the osteoarthritis joint, IL-1β as proinflammatory cytokine, and 45-kDa fibronectin fragments as products of matrix degradation. We reported that synovial fibroblasts constitutively express and release ADAMTS 4, 5, 7, and 12. Despite the contribution of both mediators to the stimulation of Runx2 and Wnt/β-catenin signaling pathways, as well as to ADAMTS expression, promoting the degradation of aggrecan and cartilage oligomeric matrix protein from cartilage, fibronectin fragments rather than IL-1β played the major pathological role in osteoarthritis, contributing to the maintenance of the disease. Moreover, higher levels of ADAMTS 4 and 7 and a specific regulation of ADAMTS-12 were observed in osteoarthritis, suggesting them as new potential therapeutic targets. Therefore, synovial fibroblasts provide the biochemical tools to the chronicity and destruction of the osteoarthritic joints.
Collapse
Affiliation(s)
- Selene Pérez-García
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Irene Gutiérrez-Cañas
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Iria V Seoane
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Julián Fernández
- Traumatology Service, Hospital Universitario de La Princesa, Medical Research Institute, Madrid, Spain
| | - Mario Mellado
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Javier Leceta
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Laura Tío
- Cellular Inflammation and Cartilage Research Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Raúl Villanueva-Romero
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Yasmina Juarranz
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
| | - Rosa P Gomariz
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
33
|
Alonso S, González B, Ruiz-Larroya T, Durán Domínguez M, Kato T, Matsunaga A, Suzuki K, Strongin AY, Gimènez-Bonafé P, Perucho M. Epigenetic inactivation of the extracellular matrix metallopeptidase ADAMTS19 gene and the metastatic spread in colorectal cancer. Clin Epigenetics 2015; 7:124. [PMID: 26634009 PMCID: PMC4667455 DOI: 10.1186/s13148-015-0158-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/24/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND ADAMTS19 encodes a member of the ADAMTS (a disintegrin and metalloproteinase domain with thrombospondin motifs) protein family with emerging roles in carcinogenesis and metastasis. ADAMTS shares several distinct protein modules including a propeptide region, a metalloproteinase domain, a disintegrin-like domain, and a thrombospondin type 1 (TS) motif. In a previous work, we found ADAMTS19 frequently hypermethylated in colorectal cancer (CRC). We explored the association of methylation with tumor genotype and phenotype. RESULTS The methylation status of the CpG island in the promoter of ADAMTS19 was determined in 252 colorectal, 65 pancreatic, 33 breast and 169 ovarian primary tumors, 70 CRC metastases, and 10 CRC cell lines. Tumor-specific methylation of ADAMTS19 was significantly more frequent in gastrointestinal than in gynecological cancers (odds ratio (OR) = 2.9, confidence interval (CI) = (1.9-4.7), p = 5.2 × 10(-7)) and was independent of the methylation of adjacent loci in CRC. Hypermethylation associated with CRC with mutated BRAF oncogene (OR = 10.1, CI = (3.1-42.9), p = 6.3 × 10(-6)) and with the mucinous phenotype in CRC (OR = 2.1, CI = (1.1-4.1), p = 0.023) and ovarian cancer (OR = 60, CI = (16-346), p = 4 × 10(-16)). Methylation was significantly more frequent in CRC metastases homing to the ovary and omentum than in those homing to the liver and lung (OR = 6.1, CI = (1.8-22.2), p = 0.001). Differentiating local from distant metastatic spread, methylation negatively associated with tumor progression (p = 0.031) but positively with depth of invasion (p = 0.030). Hypermethylation associated with transcriptional repression in CRC cell lines, and treatment with 5'-AZA-2'-deoxycytidine led to reactivation of mRNA expression. shRNA-mediated silencing of ADAMTS19 had no effect on the in vitro proliferation rate of CRC cells but significantly diminished their collective migration speed (56 %, p = 3.3 × 10(-4)) and potential to migrate in collagen I (64 %, p = 4.3 × 10(-10)). CONCLUSIONS Our results highlight the frequent involvement of ADAMTS19 epigenetic silencing in CRC and mucinous ovarian cancer. The mechanistic preferences for the target organ of metastatic spread may lead to the development of diagnostic CRC biomarkers. The association with the mucinous phenotype also may have diagnostic applications for ovarian cancer.
Collapse
Affiliation(s)
- Sergio Alonso
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Institut d'investigació en ciéncies de la salut Germans Trias I Pujol, (IGTP), Campus Can Ruti, 08916 Badalona, Barcelona Spain
| | - Beatriz González
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Institut d'investigació en ciéncies de la salut Germans Trias I Pujol, (IGTP), Campus Can Ruti, 08916 Badalona, Barcelona Spain
| | - Tatiana Ruiz-Larroya
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Institut d'investigació en ciéncies de la salut Germans Trias I Pujol, (IGTP), Campus Can Ruti, 08916 Badalona, Barcelona Spain ; Sanford Burnham Prebys Medical Dicovery Institute, 10901 N. Torrey Pines Rd. La Jolla, San Diego, CA 92037 USA
| | | | - Takaharu Kato
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Institut d'investigació en ciéncies de la salut Germans Trias I Pujol, (IGTP), Campus Can Ruti, 08916 Badalona, Barcelona Spain ; Department of Surgery, Saitama Medical Center, Jichi Medical University, 1-847, Amanuma-cho, Omiya-ku, Saitama, 330-8503 Japan
| | - Akihiro Matsunaga
- Sanford Burnham Prebys Medical Dicovery Institute, 10901 N. Torrey Pines Rd. La Jolla, San Diego, CA 92037 USA
| | - Koichi Suzuki
- Department of Surgery, Saitama Medical Center, Jichi Medical University, 1-847, Amanuma-cho, Omiya-ku, Saitama, 330-8503 Japan
| | - Alex Y Strongin
- Sanford Burnham Prebys Medical Dicovery Institute, 10901 N. Torrey Pines Rd. La Jolla, San Diego, CA 92037 USA
| | - Pepita Gimènez-Bonafé
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Institut d'investigació en ciéncies de la salut Germans Trias I Pujol, (IGTP), Campus Can Ruti, 08916 Badalona, Barcelona Spain ; Departament de Ciències Fisiològiques II, Campus Ciènces de Salut de Bellvitge, IDIBELL, University of Barcelona, Barcelona, 08907 Spain
| | - Manuel Perucho
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Institut d'investigació en ciéncies de la salut Germans Trias I Pujol, (IGTP), Campus Can Ruti, 08916 Badalona, Barcelona Spain ; Sanford Burnham Prebys Medical Dicovery Institute, 10901 N. Torrey Pines Rd. La Jolla, San Diego, CA 92037 USA ; Institució Catalana de Recerca i Estudis Avançats (ICREA), Catalan Institution for Research and Advanced Studies. Pg. Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
34
|
Zhang P, Shen M, Fernandez-Patron C, Kassiri Z. ADAMs family and relatives in cardiovascular physiology and pathology. J Mol Cell Cardiol 2015; 93:186-99. [PMID: 26522853 DOI: 10.1016/j.yjmcc.2015.10.031] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/26/2015] [Accepted: 10/28/2015] [Indexed: 12/21/2022]
Abstract
A disintegrin and metalloproteinases (ADAMs) are a family of membrane-bound proteases. ADAM-TSs (ADAMs with thrombospondin domains) are a close relative of ADAMs that are present in soluble form in the extracellular space. Dysregulated production or function of these enzymes has been associated with pathologies such as cancer, asthma, Alzheimer's and cardiovascular diseases. ADAMs contribute to angiogenesis, hypertrophy and apoptosis in a stimulus- and cell type-dependent manner. Among the ADAMs identified so far (34 in mouse, 21 in human), ADAMs 8, 9, 10, 12, 17 and 19 have been shown to be involved in cardiovascular development or cardiomyopathies; and among the 19 ADAM-TSs, ADAM-TS1, 5, 7 and 9 are important in development of the cardiovascular system, while ADAM-TS13 can contribute to vascular disorders. Meanwhile, there remain a number of ADAMs and ADAM-TSs whose function in the cardiovascular system has not been yet explored. The current knowledge about the role of ADAMs and ADAM-TSs in the cardiovascular pathologies is still quite limited. The most detailed studies have been performed in other cell types (e.g. cancer cells) and organs (nervous system) which can provide valuable insight into the potential functions of ADAMs and ADAM-TSs, their mechanism of action and therapeutic potentials in cardiomyopathies. Here, we review what is currently known about the structure and function of ADAMs and ADAM-TSs, and their roles in development, physiology and pathology of the cardiovascular system.
Collapse
Affiliation(s)
- Pu Zhang
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Mengcheng Shen
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Carlos Fernandez-Patron
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
35
|
Kelwick R, Desanlis I, Wheeler GN, Edwards DR. The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin motifs) family. Genome Biol 2015; 16:113. [PMID: 26025392 PMCID: PMC4448532 DOI: 10.1186/s13059-015-0676-3] [Citation(s) in RCA: 433] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The ADAMTS (A Disintegrin and Metalloproteinase with Thrombospondin motifs) enzymes are secreted, multi-domain matrix-associated zinc metalloendopeptidases that have diverse roles in tissue morphogenesis and patho-physiological remodeling, in inflammation and in vascular biology. The human family includes 19 members that can be sub-grouped on the basis of their known substrates, namely the aggrecanases or proteoglycanases (ADAMTS1, 4, 5, 8, 9, 15 and 20), the procollagen N-propeptidases (ADAMTS2, 3 and 14), the cartilage oligomeric matrix protein-cleaving enzymes (ADAMTS7 and 12), the von-Willebrand Factor proteinase (ADAMTS13) and a group of orphan enzymes (ADAMTS6, 10, 16, 17, 18 and 19). Control of the structure and function of the extracellular matrix (ECM) is a central theme of the biology of the ADAMTS, as exemplified by the actions of the procollagen-N-propeptidases in collagen fibril assembly and of the aggrecanases in the cleavage or modification of ECM proteoglycans. Defects in certain family members give rise to inherited genetic disorders, while the aberrant expression or function of others is associated with arthritis, cancer and cardiovascular disease. In particular, ADAMTS4 and 5 have emerged as therapeutic targets in arthritis. Multiple ADAMTSs from different sub-groupings exert either positive or negative effects on tumorigenesis and metastasis, with both metalloproteinase-dependent and -independent actions known to occur. The basic ADAMTS structure comprises a metalloproteinase catalytic domain and a carboxy-terminal ancillary domain, the latter determining substrate specificity and the localization of the protease and its interaction partners; ancillary domains probably also have independent biological functions. Focusing primarily on the aggrecanases and proteoglycanases, this review provides a perspective on the evolution of the ADAMTS family, their links with developmental and disease mechanisms, and key questions for the future.
Collapse
Affiliation(s)
- Richard Kelwick
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| | - Ines Desanlis
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| | - Grant N Wheeler
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| | - Dylan R Edwards
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
36
|
Cal S, López-Otín C. ADAMTS proteases and cancer. Matrix Biol 2015; 44-46:77-85. [PMID: 25636539 DOI: 10.1016/j.matbio.2015.01.013] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 01/19/2015] [Accepted: 01/19/2015] [Indexed: 11/17/2022]
Abstract
ADAMTSs (A disintegrin and metalloprotease domains with thrombospondins motifs) are complex extracellular proteases that have been related to both oncogenic and tumor-protective functions. These enzymes can be secreted by cancer and stromal cells and may contribute to modify the tumor microenvironment by multiple mechanisms. Thus, ADAMTSs can cleave or interact with a wide range of extracellular matrix components or regulatory factors, and therefore affect cell adhesion, migration, proliferation and angiogenesis. The balance of protumor versus antitumor effects of ADAMTSs may depend on the nature of their substrates or interacting-partners upon secretion from the cell. Moreover, different ADAMTS genes have been found overexpressed, mutated or epigenetically silenced in tumors from different origins, suggesting the direct impact of these metalloproteases in cancer development. However, despite the important advances on the tumor biology of ADAMTSs in recent years, more mechanistic and functional studies are necessary to fully understand how these proteases can influence tumor microenvironment to potentiate cancer growth or to induce tumor regression. This review outlines current and emerging connections between ADAMTSs and cancer.
Collapse
Affiliation(s)
- Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología del Principado de Asturias, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain.
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología del Principado de Asturias, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| |
Collapse
|
37
|
Fontanil T, Rúa S, Llamazares M, Moncada-Pazos A, Quirós PM, García-Suárez O, Vega JA, Sasaki T, Mohamedi Y, Esteban MM, Obaya AJ, Cal S. Interaction between the ADAMTS-12 metalloprotease and fibulin-2 induces tumor-suppressive effects in breast cancer cells. Oncotarget 2015; 5:1253-64. [PMID: 24457941 PMCID: PMC4012729 DOI: 10.18632/oncotarget.1690] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Balance between pro-tumor and anti-tumor effects may be affected by molecular interactions within tumor microenvironment. On this basis we searched for molecular partners of ADAMTS-12, a secreted metalloprotease that shows both oncogenic and tumor-suppressive effects. Using its spacer region as a bait in a yeast two-hybrid screen, we identified fibulin-2 as a potential ADAMTS-12-interacting protein. Fibulins are components of basement membranes and elastic matrix fibers in connective tissue. Besides this structural function, fibulins also play crucial roles in different biological events, including tumorigenesis. To examine the functional consequences of the ADAMTS-12/fibulin-2 interaction, we performed different in vitro assays using two breast cancer cell lines: the poorly invasive MCF-7 and the highly invasive MDA-MB-231. Overall our data indicate that this interaction promotes anti-tumor effects in breast cancer cells. To assess the in vivo relevance of this interaction, we induced tumors in nude mice using MCF-7 cells expressing both ADAMTS-12 and fibulin-2 that showed a remarkable growth deficiency. Additionally, we also found that ADAMTS-12 may elicit pro-tumor effects in the absence of fibulin-2. Immunohistochemical staining of breast cancer samples allowed the detection of both ADAMTS-12 and fibulin-2 in the connective tissue surrounding tumor area in less aggressive carcinomas. However, both proteins are hardly detected in more aggressive tumors. These data and survival analysis plots of breast cancer patients suggest that concomitant detection of ADAMTS-12 and fibulin-2 could be a good prognosis marker in breast cancer diagnosis.
Collapse
Affiliation(s)
- Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Oviedo, Asturias, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kelwick R, Wagstaff L, Decock J, Roghi C, Cooley LS, Robinson SD, Arnold H, Gavrilović J, Jaworski DM, Yamamoto K, Nagase H, Seubert B, Krüger A, Edwards DR. Metalloproteinase-dependent and -independent processes contribute to inhibition of breast cancer cell migration, angiogenesis and liver metastasis by a disintegrin and metalloproteinase with thrombospondin motifs-15. Int J Cancer 2014; 136:E14-26. [PMID: 25099234 DOI: 10.1002/ijc.29129] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 07/15/2014] [Accepted: 07/18/2014] [Indexed: 01/10/2023]
Abstract
The ADAMTS proteinases are a family of secreted, matrix-associated enzymes that have diverse roles in the regulation of tissue organization and vascular homeostasis. Several of the 19 human family members have been identified as having either tumor promoting or suppressing roles. We previously demonstrated that decreased ADAMTS15 expression correlated with a worse clinical outcome in mammary carcinoma (e.g., Porter et al., Int J Cancer 2006;118:1241-7). We have explored the effects of A Disintegrin and Metalloproteinase with Thrombospondin motifs-15 (ADAMTS-15) on the behavior of MDA-MB-231 and MCF-7 breast cancer cells by stable expression of either a wild-type (wt) or metalloproteinase-inactive (E362A) protein. No effects on mammary cancer cell proliferation or apoptosis were observed for either form of ADAMTS-15. However, both forms reduced cell migration on fibronectin or laminin matrices, though motility on a Type I collagen matrix was unimpaired. Knockdown of syndecan-4 attenuated the inhibitory effects of ADAMTS-15 on cell migration. In contrast to its effects on cell migration, wt ADAMTS-15 but not the E362A inactive mutant inhibited endothelial tubulogenesis in 3D collagen gels and angiogenesis in the aortic ring assay. In experimental metastasis assays in nude mice, MDA-MB-231 cells expressing either form of ADAMTS-15 showed reduced spread to the liver, though lung colonization was enhanced for cells expressing wt ADAMTS-15. These studies indicate that extracellular ADAMTS-15 has multiple actions on tumor pathophysiology. Via modulation of cell-ECM interactions, which likely involve syndecan-4, it attenuates mammary cancer cell migration independent of its metalloproteinase activity; however, its antiangiogenic action requires catalytic functionality, and its effects on metastasis in vivo are tissue niche-dependent.
Collapse
Affiliation(s)
- Richard Kelwick
- School of Biological Sciences, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
ADAMTS-12: a multifaced metalloproteinase in arthritis and inflammation. Mediators Inflamm 2014; 2014:649718. [PMID: 24876675 PMCID: PMC4020202 DOI: 10.1155/2014/649718] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/28/2014] [Accepted: 04/07/2014] [Indexed: 12/12/2022] Open
Abstract
ADAMTS-12 is a member of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family of proteases, which were known to play important roles in various biological and pathological processes, such as development, angiogenesis, inflammation, cancer, arthritis, and atherosclerosis. In this review, we briefly summarize the structural organization of ADAMTS-12; concentrate on the emerging role of ADAMTS-12 in several pathophysiological conditions, including intervertebral disc degeneration, tumorigenesis and angioinhibitory effects, pediatric stroke, gonad differentiation, trophoblast invasion, and genetic linkage to schizophrenia and asthma, with special focus on its role in arthritis and inflammation; and end with the perspective research of ADAMTS-12 and its potential as a promising diagnostic and therapeutic target in various kinds of diseases and conditions.
Collapse
|
40
|
Stoll M, Rühle F, Nowak-Göttl U. Advances in understanding stroke risk in children--a geneticist's view. Br J Haematol 2013; 164:636-45. [PMID: 24354735 DOI: 10.1111/bjh.12706] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 11/13/2013] [Indexed: 12/28/2022]
Abstract
The advent of the genomic era has provided novel insights into the genetic architecture of common complex diseases, such as thrombophilia and stroke. Since 2006, a growing number of genome wide association studies (GWAS) for common complex diseases have revealed new candidate loci and genomic regions that play an important role in disease aetiology and progression. While GWAS on the above mentioned traits are abundant in adults, similar studies in paediatric study cohorts are lagging behind. However, genetic research in this important clinical area has gained momentum and starts to provide us with exciting insights into the genetic underpinnings of stroke with paediatric onset. Here we review recent advances in genetic association studies underlying stroke in children and aim to translate the results to clinical utility. These studies comprise candidate gene approaches and GWAS, and represent the current status on what we have learnt about the genetic architecture underlying paediatric stroke, and how this may affect medical practice in the years to come.
Collapse
Affiliation(s)
- Monika Stoll
- Genetic Epidemiology, University of Muenster, Muenster, Germany
| | | | | |
Collapse
|
41
|
Choi GCG, Li J, Wang Y, Li L, Zhong L, Ma B, Su X, Ying J, Xiang T, Rha SY, Yu J, Sung JJY, Tsao SW, Chan ATC, Tao Q. The Metalloprotease ADAMTS8 Displays Antitumor Properties through Antagonizing EGFR–MEK–ERK Signaling and Is Silenced in Carcinomas by CpG Methylation. Mol Cancer Res 2013; 12:228-38. [PMID: 24184540 DOI: 10.1158/1541-7786.mcr-13-0195] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Gigi C G Choi
- Room 315, Cancer Center, PWH, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Przemyslaw L, Boguslaw HA, Elzbieta S, Malgorzata SM. ADAM and ADAMTS family proteins and their role in the colorectal cancer etiopathogenesis. BMB Rep 2013; 46:139-50. [PMID: 23527857 PMCID: PMC4133867 DOI: 10.5483/bmbrep.2013.46.3.176] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The ADAM and ADAMTS families, also called adamalysins belong to an important group of extracellular matrix proteins. The ADAMs family belong to both the transmembrane and secreted proteins, while ADAMTS family only contains secreted forms. Adamalysins play an important role in the cell phenotype regulation via their activities in signaling pathways, cell adhesion and migration. The human proteome contains 21 ADAM, and 19 ADAMTS proteins, which are involved in extracellular matrix remodeling, shedding of various substrates such as: adhesion ligands, growth factors, their receptors and diverse cytokines. Recent studies provide evidence that adamalysins play a crucial role in colorectal cancer (CRC) etiopathogenesis. It seems possible that adamalysins might be used as CRC prediction markers or potential pharmaceutical targets. [BMB Reports 2013; 46(3): 139-150]
Collapse
|
43
|
Wasserkort R, Kalmar A, Valcz G, Spisak S, Krispin M, Toth K, Tulassay Z, Sledziewski AZ, Molnar B. Aberrant septin 9 DNA methylation in colorectal cancer is restricted to a single CpG island. BMC Cancer 2013; 13:398. [PMID: 23988185 PMCID: PMC3837632 DOI: 10.1186/1471-2407-13-398] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 08/28/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The septin 9 gene (SEPT9) codes for a GTP-binding protein associated with filamentous structures and cytoskeleton formation. SEPT9 plays a role in multiple cancers as either an oncogene or a tumor suppressor gene. Regulation of SEPT9 expression is complex and not well understood; however, hypermethylation of the gene was recently introduced as a biomarker for early detection of colorectal cancer (CRC) and has been linked to cancer of the breast and of the head and neck. Because the DNA methylation landscape of different regions of SEPT9 is poorly understood in cancer, we analyzed the methylation patterns of this gene in distinct cell populations from normal and diseased colon mucosa. METHODS Laser capture microdissection was performed to obtain homogeneous populations of epithelial and stromal cells from normal, adenomatous, and tumorous colon mucosa. Microdissected samples were analyzed using direct bisulfite sequencing to determine the DNA methylation status of eight regions within and near the SEPT9 gene. Septin-9 protein expression was assessed using immunohistochemistry (IHC). RESULTS Regions analyzed in SEPT9 were unmethylated in normal tissue except for a methylation boundary detected downstream of the largest CpG island. In adenoma and tumor tissues, epithelial cells displayed markedly increased DNA methylation levels (>80%, p <0.0001) in only one of the CpG islands investigated. SEPT9 methylation in stromal cells increased in adenomatous and tumor tissues (≤50%, p <0.0001); however, methylation did not increase in stromal cells of normal tissue close to the tumor. IHC data indicated a significant decrease (p <0.01) in Septin-9 protein levels in epithelial cells derived from adenoma and tumor tissues; Septin-9 protein levels in stromal cells were low in all tissues. CONCLUSIONS Hypermethylation of SEPT9 in adenoma and CRC specimens is confined to one of several CpG islands of this gene. Tumor-associated aberrant methylation originates in epithelial cells; stromal cells appear to acquire hypermethylation subsequent to epithelial cells, possibly through field effects. The region in SEPT9 with disease-related hypermethylation also contains the CpGs targeted by a novel blood-based screening test (Epi proColon®), providing further support for the clinical relevance of this biomarker.
Collapse
Affiliation(s)
- Reinhold Wasserkort
- Epigenomics AG, Berlin, Germany
- Current address: Delta-Vir GmbH, Leipzig, Germany
| | - Alexandra Kalmar
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Gabor Valcz
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Sandor Spisak
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Manuel Krispin
- Epigenomics AG, Berlin, Germany
- Current address: Zymo Research, Irvine CA 92614, USA
| | - Kinga Toth
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Zsolt Tulassay
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
- Molecular Medicine Research Unit, Hungarian Academy of Science, Budapest, Hungary
| | | | - Bela Molnar
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
- Molecular Medicine Research Unit, Hungarian Academy of Science, Budapest, Hungary
| |
Collapse
|
44
|
Cifola I, Pietrelli A, Consolandi C, Severgnini M, Mangano E, Russo V, De Bellis G, Battaglia C. Comprehensive genomic characterization of cutaneous malignant melanoma cell lines derived from metastatic lesions by whole-exome sequencing and SNP array profiling. PLoS One 2013; 8:e63597. [PMID: 23704925 PMCID: PMC3660556 DOI: 10.1371/journal.pone.0063597] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 04/04/2013] [Indexed: 02/08/2023] Open
Abstract
Cutaneous malignant melanoma is the most fatal skin cancer and although improved comprehension of its pathogenic pathways allowed to realize some effective molecular targeted therapies, novel targets and drugs are still needed. Aiming to add genetic information potentially useful for novel targets discovery, we performed an extensive genomic characterization by whole-exome sequencing and SNP array profiling of six cutaneous melanoma cell lines derived from metastatic patients. We obtained a total of 3,325 novel coding single nucleotide variants, including 2,172 non-synonymous variants. We catalogued the coding mutations according to Sanger COSMIC database and to a manually curated list including genes involved in melanoma pathways identified by mining recent literature. Besides confirming the presence of known melanoma driver mutations (BRAF(V600E), NRAS(Q61R) ), we identified novel mutated genes involved in signalling pathways crucial for melanoma pathogenesis and already addressed by current targeted therapies (such as MAPK and glutamate pathways). We also identified mutations in four genes (MUC19, PAICS, RBMXL1, KIF23) never reported in melanoma, which might deserve further investigations. All data are available to the entire research community in our Melanoma Exome Database (at https://155.253.6.64/MExDB/). In summary, these cell lines are valuable biological tools to improve the genetic comprehension of this complex cancer disease and to study functional relevance of individual mutational events, and these findings could provide insights potentially useful for identification of novel therapeutic targets for cutaneous malignant melanoma.
Collapse
Affiliation(s)
- Ingrid Cifola
- Institute for Biomedical Technologies, National Research Council, Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Peng L, Yang Z, Tan C, Ren G, Chen J. Epigenetic inactivation of ADAMTS9 via promoter methylation in multiple myeloma. Mol Med Rep 2013; 7:1055-61. [PMID: 23358566 DOI: 10.3892/mmr.2013.1291] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 01/21/2013] [Indexed: 11/06/2022] Open
Abstract
A disintegrin‑like and metalloprotease with thrombospondin type Ⅰ motifs (ADAMTS) are a family of 19 secreted mammalian metalloproteases. ADAMTS9 was reported to be a novel tumor suppressor gene and is downregulated in various types of human cancer due to hypermethylation of promoter CpG islands. In the present study, the silencing mechanism of the ADAMTS9 gene was analyzed in the multiple myeloma (MM) cell lines, KM3 and RPMI‑8226. Control and MM samples were obtained by conventional bone marrow (BM) biopsy of normal and MM adult BM, respectively. RT‑PCR revealed a high expression of the ADAMTS9 gene in normal samples and RPMI‑8226 cells while marked gene silencing of ADAMTS9 was observed in MM patients and KM3 cells. Promoter methylation of ADAMTS9 was detected in the KM3 cell line and 66% (37/56) MM patients by methylation‑specific PCR. In addition, the DNA demethylating agent, 5‑aza‑2'‑deoxycytidine and trichostatin A restored ADAMTS9 expression by suppressing promoter methylation in KM3 cells. Ectopic expression of ADAMTS9 in ADAMTS9‑silenced MM cells was found to significantly suppress cell colony formation and proliferation. In the present study, DNA methylation was found to play a key role in ADAMTS9 gene silencing and the biological behavior of myeloma cells. The results demonstrate that ADAMTS9 silencing by methylation may be a novel tumor marker for MM and the applicability of demethylating agents in the treatment of MM.
Collapse
Affiliation(s)
- Ling Peng
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | | | | | | | | |
Collapse
|
46
|
Kumar S, Rao N, Ge R. Emerging Roles of ADAMTSs in Angiogenesis and Cancer. Cancers (Basel) 2012; 4:1252-99. [PMID: 24213506 PMCID: PMC3712723 DOI: 10.3390/cancers4041252] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 11/21/2012] [Accepted: 11/23/2012] [Indexed: 12/18/2022] Open
Abstract
A Disintegrin-like And Metalloproteinase with ThromboSpondin motifs—ADAMTSs—are a multi-domain, secreted, extracellular zinc metalloproteinase family with 19 members in humans. These extracellular metalloproteinases are known to cleave a wide range of substrates in the extracellular matrix. They have been implicated in various physiological processes, such as extracellular matrix turnover, melanoblast development, interdigital web regression, blood coagulation, ovulation, etc. ADAMTSs are also critical in pathological processes such as arthritis, atherosclerosis, cancer, angiogenesis, wound healing, etc. In the past few years, there has been an explosion of reports concerning the role of ADAMTS family members in angiogenesis and cancer. To date, 10 out of the 19 members have been demonstrated to be involved in regulating angiogenesis and/or cancer. The mechanism involved in their regulation of angiogenesis or cancer differs among different members. Both angiogenesis-dependent and -independent regulation of cancer have been reported. This review summarizes our current understanding on the roles of ADAMTS in angiogenesis and cancer and highlights their implications in cancer therapeutic development.
Collapse
Affiliation(s)
- Saran Kumar
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore.
| | | | | |
Collapse
|
47
|
Bernal T, Moncada-Pazos A, Soria-Valles C, Gutiérrez-Fernández A. Effects of azacitidine on matrix metalloproteinase-9 in acute myeloid leukemia and myelodysplasia. Exp Hematol 2012; 41:172-9. [PMID: 23085464 DOI: 10.1016/j.exphem.2012.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 09/22/2012] [Accepted: 10/10/2012] [Indexed: 12/29/2022]
Abstract
Matrix metalloprotease-9 (MMP9) plays a critical role in acute myeloid leukemia (AML) by increasing the invasive properties of malignant myeloblasts. The role of this enzyme in high-risk myelodysplastic diseases (MDS) and the effect of azacitidine on its expression in MDS and AML have not been studied in detail. In this work, we have analyzed the effect of different concentrations of azacitidine in two well-established, MDS-derived, acute myeloid leukemic cell lines: MOLM-13 and SKM-1. We have demonstrated that 1 μmol/L azacitidine decreases MMP9 DNA methylation levels and that this is correlated with a significant increase in messenger RNA expression in both cell lines. Surprisingly, changes in protein levels were minor. This paradoxic effect is explained by the drug-dependent induction of apoptosis that reduces the amount of active secreting cells. A balance between induced expression and apoptosis was established at an azacitidine concentration of 0.2 μmol/L in MOLM-13 cells. This dose significantly increased the invasive capacity of viable cells, as measured in the Matrigel assay. To evaluate the clinical relevance of this observation, we have examined the effect of azacitidine on MMP9 expression in bone marrow from five patients with MDS, with the finding that this drug significantly increased MMP9 protein levels in all analyzed patients after six cycles of treatment. Based on these results, we conclude that azacitidine increases MMP9 expression and may enhance invasiveness in vitro. Because all five patients relapsed, these findings might explain, at least partially, the clinical failure of the drug and the progression to a more aggressive disease.
Collapse
Affiliation(s)
- Teresa Bernal
- Servicio de Hematología, Hospital Universitario Central de Asturias, Oviedo, Spain.
| | | | | | | |
Collapse
|
48
|
Moncada-Pazos A, Obaya AJ, Llamazares M, Heljasvaara R, Suárez MF, Colado E, Noël A, Cal S, López-Otín C. ADAMTS-12 metalloprotease is necessary for normal inflammatory response. J Biol Chem 2012; 287:39554-63. [PMID: 23019333 DOI: 10.1074/jbc.m112.408625] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
ADAMTSs (a disintegrin and metalloprotease with thrombospondin domains) are a family of enzymes with both proteolytic and protein interaction functions, which have been implicated in distinct pathologies. In this work, we have investigated the putative role of ADAMTS-12 in inflammation by using a mouse model deficient in this metalloprotease. Control and mutant mice were subjected to different experimental conditions to induce colitis, endotoxic sepsis, and pancreatitis. We have observed that Adamts12-deficient mice exhibit more severe inflammation and a delayed recovery from these challenges compared with their wild-type littermates. These changes are accompanied by an increase in inflammatory markers including several cytokines, as assessed by microarray expression analysis and proteomic-based approaches. Interestingly, the clinical symptoms observed in Adamts12-deficient mice are also concomitant with an elevation in the number of neutrophils in affected tissues. Finally, isolation and in vitro culture of human neutrophils demonstrate that the presence of ADAMTS-12 induces neutrophil apoptosis. On the basis of these results, we propose that ADAMTS-12 is implicated in the inflammatory response by modulating normal neutrophil apoptosis.
Collapse
Affiliation(s)
- Angela Moncada-Pazos
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vong S, Kalluri R. The role of stromal myofibroblast and extracellular matrix in tumor angiogenesis. Genes Cancer 2012; 2:1139-45. [PMID: 22866205 DOI: 10.1177/1947601911423940] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tumor angiogenesis, the building of blood vessels in an expanding tumor mass, is an elegantly coordinated process that dictates tumor growth and progression. Stromal components of the tumor microenvironment, such as myofibroblasts and the extracellular matrix, collaborate with tumor cells in regulating development. Such myofibroblasts and the extracellular matrix have ever-expanding roles in the angiogenic process as well. This review summarizes how stromal myofibroblasts and the extracellular matrix can modulate tumor angiogenesis, highlighting recent findings.
Collapse
Affiliation(s)
- Sylvia Vong
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
50
|
Butchar JP, Cain D, Manivannan SN, McCue AD, Bonanno L, Halula S, Truesdell S, Austin CL, Jacobsen TL, Simcox A. New negative feedback regulators of Egfr signaling in Drosophila. Genetics 2012; 191:1213-26. [PMID: 22595244 PMCID: PMC3416002 DOI: 10.1534/genetics.112.141093] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 05/07/2012] [Indexed: 12/18/2022] Open
Abstract
The highly conserved epidermal growth factor receptor (Egfr) pathway is required in all animals for normal development and homeostasis; consequently, aberrant Egfr signaling is implicated in a number of diseases. Genetic analysis of Drosophila melanogaster Egfr has contributed significantly to understanding this conserved pathway and led to the discovery of new components and targets. Here we used microarray analysis of third instar wing discs, in which Egfr signaling was perturbed, to identify new Egfr-responsive genes. Upregulated transcripts included five known targets, suggesting the approach was valid. We investigated the function of 29 previously uncharacterized genes, which had pronounced responses. The Egfr pathway is important for wing-vein patterning and using reverse genetic analysis we identified five genes that showed venation defects. Three of these genes are expressed in vein primordia and all showed transcriptional changes in response to altered Egfr activity consistent with being targets of the pathway. Genetic interactions with Egfr further linked two of the genes, Sulfated (Sulf1), an endosulfatase gene, and CG4096, an A Disintegrin And Metalloproteinase with ThromboSpondin motifs (ADAMTS) gene, to the pathway. Sulf1 showed a strong genetic interaction with the neuregulin-like ligand vein (vn) and may influence binding of Vn to heparan-sulfated proteoglycans (HSPGs). How Drosophila Egfr activity is modulated by CG4096 is unknown, but interestingly vertebrate EGF ligands are regulated by a related ADAMTS protein. We suggest Sulf1 and CG4096 are negative feedback regulators of Egfr signaling that function in the extracellular space to influence ligand activity.
Collapse
Affiliation(s)
- Jonathan P. Butchar
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210
| | - Donna Cain
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210
| | | | - Andrea D. McCue
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210
| | - Liana Bonanno
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210
| | - Sarah Halula
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210
| | - Sharon Truesdell
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210
| | - Christina L. Austin
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210
| | - Thomas L. Jacobsen
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210
| | - Amanda Simcox
- Department of Molecular Genetics, Ohio State University, Columbus, Ohio 43210
| |
Collapse
|