1
|
Tan K, Jäger C, Geissler S, Schlenzig D, Buchholz M, Ramsbeck D. Synthesis and structure-activity relationships of pyrazole-based inhibitors of meprin α and β. J Enzyme Inhib Med Chem 2023; 38:2165648. [PMID: 36661029 PMCID: PMC9870012 DOI: 10.1080/14756366.2023.2165648] [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] [Indexed: 01/21/2023] Open
Abstract
Targeting metalloproteinases has been in the focus of drug design for a long time. However, meprin α and β emerged as potential drug targets just recently and are linked to several diseases with different pathological background. Nevertheless, the validation of meprins as suitable drug targets still requires highly potent and selective inhibitors as chemical probes to elucidate their role in pathophysiology. Albeit highly selective inhibitors of meprin β have already been reported, only inhibitors of meprin α with modest activity or selectivity are known. Starting from recently reported heteroaromatic scaffolds, the aim of this study was the optimisation of meprin α and/or meprin β inhibition while keeping the favourable off-target inhibition profile over other metalloproteases. We report potent pan-meprin inhibitors as well as highly active inhibitors of meprin α with superior selectivity over meprin β. The latter are suitable to serve as chemical probes and enable further target validation.
Collapse
Affiliation(s)
- Kathrin Tan
- Department of Drug Design and Target Validation MWT, Fraunhofer Institute for Cell Therapy and Immunology IZI, Biocenter, Halle (Saale), Germany
| | | | - Stefanie Geissler
- Department of Drug Design and Target Validation MWT, Fraunhofer Institute for Cell Therapy and Immunology IZI, Biocenter, Halle (Saale), Germany
| | - Dagmar Schlenzig
- Department of Drug Design and Target Validation MWT, Fraunhofer Institute for Cell Therapy and Immunology IZI, Biocenter, Halle (Saale), Germany
| | - Mirko Buchholz
- Department of Drug Design and Target Validation MWT, Fraunhofer Institute for Cell Therapy and Immunology IZI, Biocenter, Halle (Saale), Germany
| | - Daniel Ramsbeck
- Department of Drug Design and Target Validation MWT, Fraunhofer Institute for Cell Therapy and Immunology IZI, Biocenter, Halle (Saale), Germany,CONTACT Daniel Ramsbeck Department of Drug Design and Target Validation MWT, Fraunhofer Institute for Cell Therapy and Immunology IZI, Biocenter, Weinbergweg 22, Halle (Saale), 06120, Germany
| |
Collapse
|
2
|
Li M, Huang C, Wu Y, Zhu L, Zhang Y, Zhou Y, Li H, Liu Z, Pan X, Wang X, Qiu J, Li F, Liao W. Long non-coding RNA CCL14-AS suppresses invasiveness and lymph node metastasis of colorectal cancer cells by regulating MEP1A. Cancer Cell Int 2023; 23:27. [PMID: 36793075 PMCID: PMC9933342 DOI: 10.1186/s12935-023-02866-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play important roles in the biology of colorectal cancer (CRC). There are several lncRNAs associated with invasion and metastasis have been characterized in CRC. However, studies focusing on the precise molecular mechanisms by which lncRNAs function in lymph node (LN) metastasis in CRC are still limited. METHODS In this study, by analyzing TCGA dataset, we identified that AC244100.2 (termed CCL14-AS), a novel lncRNA enriched in the cytoplasm, was negatively correlated with LN metastasis and unfavorable prognosis of CRC. In situ hybridization was used to examine CCL14-AS expression in clinical CRC tissues. Various functional experiments including migration assay and wound-healing assay were used to investigate the effects of CCL14-AS on CRC cells migration. The nude mice popliteal lymph node metastasis model assay further confirmed the effects of CCL14-AS in vivo. RESULTS CCL14-AS expression was significantly downregulated in CRC tissues compared to adjacent normal tissues. In addition, low CCL14-AS expression was correlated with advanced T classification, LN metastasis, distant metastasis, and shorter disease-free survival of CRC patients. Functionally, CCL14-AS overexpression inhibited the invasiveness of CRC cells in vitro and LN metastasis in nude mice. On the contrary, knockdown of CCL14-AS promoted the invasiveness and LN metastasis abilities of CRC cells. Mechanistically, CCL14-AS downregulated the expression of MEP1A via interacting with MEP1A mRNA and reduced its stability. Overexpression of MEP1A rescued the invasiveness and LN metastasis abilities in CCL14-AS-overexpressing CRC cells. Moreover, the expression levels of CCL14-AS was negatively correlated with that of MEP1A in CRC tissues. CONCLUSIONS We identified a novel lncRNA, CCL14-AS, as a potential tumor suppressor in CRC. Our findings supported a model in which the CCL14-AS/MEP1A axis serves as critical regulator in CRC progression, suggesting a novel biomarker and therapeutic target in advanced CRC.
Collapse
Affiliation(s)
- Mingzhou Li
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China ,grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Chengmei Huang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuanyuan Wu
- grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China
| | - Lina Zhu
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yaxin Zhang
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi Zhou
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huali Li
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China ,grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China ,grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Zhihao Liu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China ,grid.284723.80000 0000 8877 7471Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 China ,grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Xinyan Pan
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Wang
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Junfeng Qiu
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Fengtian Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Wenting Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
3
|
Bayly-Jones C, Lupton CJ, Fritz C, Venugopal H, Ramsbeck D, Wermann M, Jäger C, de Marco A, Schilling S, Schlenzig D, Whisstock JC. Helical ultrastructure of the metalloprotease meprin α in complex with a small molecule inhibitor. Nat Commun 2022; 13:6178. [PMID: 36261433 PMCID: PMC9581967 DOI: 10.1038/s41467-022-33893-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
The zinc-dependent metalloprotease meprin α is predominantly expressed in the brush border membrane of proximal tubules in the kidney and enterocytes in the small intestine and colon. In normal tissue homeostasis meprin α performs key roles in inflammation, immunity, and extracellular matrix remodelling. Dysregulated meprin α is associated with acute kidney injury, sepsis, urinary tract infection, metastatic colorectal carcinoma, and inflammatory bowel disease. Accordingly, meprin α is the target of drug discovery programs. In contrast to meprin β, meprin α is secreted into the extracellular space, whereupon it oligomerises to form giant assemblies and is the largest extracellular protease identified to date (~6 MDa). Here, using cryo-electron microscopy, we determine the high-resolution structure of the zymogen and mature form of meprin α, as well as the structure of the active form in complex with a prototype small molecule inhibitor and human fetuin-B. Our data reveal that meprin α forms a giant, flexible, left-handed helical assembly of roughly 22 nm in diameter. We find that oligomerisation improves proteolytic and thermal stability but does not impact substrate specificity or enzymatic activity. Furthermore, structural comparison with meprin β reveal unique features of the active site of meprin α, and helical assembly more broadly.
Collapse
Affiliation(s)
- Charles Bayly-Jones
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia
| | - Christopher J Lupton
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia
| | - Claudia Fritz
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | - Hariprasad Venugopal
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, 3800, VIC, Australia
| | - Daniel Ramsbeck
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | - Michael Wermann
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | | | - Alex de Marco
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia
| | - Stephan Schilling
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
- Hochschule Anhalt, University of Applied Sciences, Köthen, Germany
| | - Dagmar Schlenzig
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany.
| | - James C Whisstock
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia.
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia.
- EMBL Australia, Monash University, Melbourne, VIC, 3800, Australia.
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
4
|
Tan K, Jäger C, Körschgen H, Geissler S, Schlenzig D, Buchholz M, Stöcker W, Ramsbeck D. Heteroaromatic Inhibitors of the Astacin Proteinases Meprin α, Meprin β and Ovastacin Discovered by a Scaffold-Hopping Approach. ChemMedChem 2021; 16:976-988. [PMID: 33369214 PMCID: PMC8048867 DOI: 10.1002/cmdc.202000822] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Indexed: 12/20/2022]
Abstract
Astacin metalloproteinases, in particular meprins α and β, as well as ovastacin, are emerging drug targets. Drug-discovery efforts have led to the development of the first potent and selective inhibitors in the last few years. However, the most recent compounds are based on a highly flexible tertiary amine scaffold that could cause metabolic liabilities or decreased potency due to the entropic penalty upon binding to the target. Thus, the aim of this study was to discover novel conformationally constrained scaffolds as starting points for further inhibitor optimization. Shifting from flexible tertiary amines to rigid heteroaromatic cores resulted in a boost in inhibitory activity. Moreover, some compounds already exhibited higher activity against individual astacin proteinases compared to recently reported inhibitors and also a favorable off-target selectivity profile, thus qualifying them as very suitable chemical probes for target validation.
Collapse
Affiliation(s)
- Kathrin Tan
- Department of Drug Design and Target Validation MWTFraunhofer Institute for Cell Therapy and Immunology IZIBiocenter, Weinbergweg 2206120Halle (Saale)Germany
| | - Christian Jäger
- Department of Drug Design and Target Validation MWTFraunhofer Institute for Cell Therapy and Immunology IZIBiocenter, Weinbergweg 2206120Halle (Saale)Germany
- present address: Vivoryon Therapeutics N.V.Weinbergweg 2206120Halle (Saale)Germany
| | - Hagen Körschgen
- Institute of Molecular PhysiologyCell and Matrix BiologyJohannes Gutenberg-University MainzJohann-Joachim-Becher-Weg 755128MainzGermany
| | - Stefanie Geissler
- Department of Drug Design and Target Validation MWTFraunhofer Institute for Cell Therapy and Immunology IZIBiocenter, Weinbergweg 2206120Halle (Saale)Germany
| | - Dagmar Schlenzig
- Department of Drug Design and Target Validation MWTFraunhofer Institute for Cell Therapy and Immunology IZIBiocenter, Weinbergweg 2206120Halle (Saale)Germany
| | - Mirko Buchholz
- Department of Drug Design and Target Validation MWTFraunhofer Institute for Cell Therapy and Immunology IZIBiocenter, Weinbergweg 2206120Halle (Saale)Germany
| | - Walter Stöcker
- Institute of Molecular PhysiologyCell and Matrix BiologyJohannes Gutenberg-University MainzJohann-Joachim-Becher-Weg 755128MainzGermany
| | - Daniel Ramsbeck
- Department of Drug Design and Target Validation MWTFraunhofer Institute for Cell Therapy and Immunology IZIBiocenter, Weinbergweg 2206120Halle (Saale)Germany
| |
Collapse
|
5
|
Medoro A, Bartollino S, Mignogna D, Marziliano N, Porcile C, Nizzari M, Florio T, Pagano A, Raimo G, Intrieri M, Russo C. Proteases Upregulation in Sporadic Alzheimer's Disease Brain. J Alzheimers Dis 2020; 68:931-938. [PMID: 30814362 DOI: 10.3233/jad-181284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Certain proteases are involved in Alzheimer's disease (AD) and their erroneous control may contribute to the pathology onset and progression. In this study we evaluated the cerebral expression of eight proteases, involved in both AβPP processing and extracellular matrix remodeling. Among these proteases, ADAM10, ADAMTS1, Cathepsin D, and Meprin β show a significantly higher mRNAs expression in sporadic AD subjects versus controls, while ADAMTS1, Cathepsin D, and Meprin β show an increment also at the protein level. These data indicate that transcriptional events affecting brain proteases are activated in AD patients, suggesting a link between proteolysis and AD.
Collapse
Affiliation(s)
- Alessandro Medoro
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Silvia Bartollino
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Donatella Mignogna
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Nicola Marziliano
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy.,Clinical Pathology Laboratory, ASL Taranto, Italy
| | - Carola Porcile
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Mario Nizzari
- Department of Internal Medicine and Centre of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - Tullio Florio
- Department of Internal Medicine and Centre of Excellence for Biomedical Research, University of Genova, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Aldo Pagano
- Department of Experimental Medicine, University of Genova, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gennaro Raimo
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Mariano Intrieri
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Claudio Russo
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| |
Collapse
|
6
|
Abstract
A crucial step for tumor cell extravasation and metastasis is the migration through the extracellular matrix, which requires proteolytic activity. Hence, proteases, particularly matrix metalloproteases (MMPs), have been discussed as therapeutic targets and their inhibition should diminish tumor growth and metastasis. The metalloproteases meprin α and meprin β are highly abundant on intestinal enterocytes and their expression was associated with different stages of colorectal cancer. Due to their ability to cleave extracellular matrix (ECM) components, they were suggested as pro-tumorigenic enzymes. Additionally, both meprins were shown to have pro-inflammatory activity by cleaving cytokines and their receptors, which correlates with chronic intestinal inflammation and associated conditions. On the other hand, meprin β was identified as an essential enzyme for the detachment and renewal of the intestinal mucus, important to prevent bacterial overgrowth and infection. Considering this, it is hard to estimate whether high activity of meprins is generally detrimental or if these enzymes have also protective functions in certain cancer types. For instance, for colorectal cancer, patients with high meprin β expression in tumor tissue exhibit a better survival prognosis, which is completely different to prostate cancer. This demonstrates that the very same enzyme may have contrary effects on tumor initiation and growth, depending on its tissue and subcellular localization. Hence, precise knowledge about proteolytic enzymes is required to design the most efficient therapeutic options for cancer treatment. In this review, we summarize the current findings on meprins' functions, expression, and cancer-associated variants with possible implications for tumor progression and metastasis.
Collapse
|
7
|
Peters F, Scharfenberg F, Colmorgen C, Armbrust F, Wichert R, Arnold P, Potempa B, Potempa J, Pietrzik CU, Häsler R, Rosenstiel P, Becker-Pauly C. Tethering soluble meprin α in an enzyme complex to the cell surface affects IBD-associated genes. FASEB J 2019; 33:7490-7504. [PMID: 30916990 DOI: 10.1096/fj.201802391r] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Biologic activity of proteases is mainly characterized by the substrate specificity, tissue distribution, and cellular localization. The human metalloproteases meprin α and meprin β share 41% sequence identity and exhibit a similar cleavage specificity with a preference for negatively charged amino acids. However, shedding of meprin α by furin on the secretory pathway makes it a secreted enzyme in comparison with the membrane-bound meprin β. In this study, we identified human meprin α and meprin β as forming covalently linked membrane-tethered heterodimers in the early endoplasmic reticulum, thereby preventing furin-mediated secretion of meprin α. Within this newly formed enzyme complex, meprin α was able to be activated on the cell surface and detected by cleavage of a novel specific fluorogenic peptide substrate. However, the known meprin β substrates amyloid precursor protein and CD99 were not shed by membrane-tethered meprin α. On the other hand, being linked to meprin α, activation of or substrate cleavage by meprin β on the cell surface was not altered. Interestingly, proteolytic activity of both proteases was increased in the heteromeric complex, indicating an increased proteolytic potential at the plasma membrane. Because meprins are susceptibility genes for inflammatory bowel disease (IBD), and to investigate the physiologic impact of the enzyme complex, we performed transcriptome analyses of intestinal mucosa from meprin-knockout mice. Comparison of the transcriptional gene analysis data with gene analyses of IBD patients revealed that different gene subsets were dysregulated if meprin α was expressed alone or in the enzyme complex, demonstrating the physiologic and pathophysiological relevance of the meprin heterodimer formation.-Peters, F., Scharfenberg, F., Colmorgen, C., Armbrust, F., Wichert, R., Arnold, P., Potempa, B., Potempa, J., Pietrzik, C. U., Häsler, R., Rosenstiel, P., Becker-Pauly, C. Tethering soluble meprin α in an enzyme complex to the cell surface affects IBD-associated genes.
Collapse
Affiliation(s)
- Florian Peters
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Franka Scharfenberg
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Cynthia Colmorgen
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Fred Armbrust
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | - Rielana Wichert
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| | | | - Barbara Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of Mainz, Mainz, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Christoph Becker-Pauly
- Unit for Degradomics of the Protease Web, Biochemical Institute, University of Kiel, Kiel, Germany
| |
Collapse
|
8
|
Tracking cellular and molecular changes in a species-specific manner during experimental tumor progression in vivo. Oncotarget 2018; 9:16149-16162. [PMID: 29662633 PMCID: PMC5882324 DOI: 10.18632/oncotarget.24598] [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: 02/12/2018] [Accepted: 02/21/2018] [Indexed: 01/29/2023] Open
Abstract
Hepatoblastoma (HBL) is a pediatric liver cancer with defined molecular alterations driving its progression. Here, we describe an animal model for HBL on the chick chorioallantoic membrane (CAM), which recapitulates relevant features of HBL in patients. Expression of classic tumor-associated proteins such as β-catenin, EpCAM and CK19 was maintained in acini-like organized tumors on CAM, as was synthesis of AFP, a tumor marker used for monitoring patient response. RNA sequencing revealed an unexpected molecular evolution of HBL cells on the CAM, with significant deregulation of more than 6,000 genes including more than half of all HOX genes. Bioinformatic analysis distinguish between tumor cell-expressed genes and chick genes, thereby shedding new light on the complex interactions taking place during HBL progression. Importantly, human tumor suppressive ribosomal genes were downregulated after implantation, whereas mitochondrial genes encoding for anti-apoptotic peptides were strongly induced in vivo. Meprin-1α expression was increased during evolution of CAM tumors and confirmed by immunohistochemistry. Cisplatin, a commonly used chemotherapeutic agent for HBL, showed significant anti-tumoral effects. Our results broaden the understanding of the molecular adaptation process of human cancer cells to the microenvironment and might help to elaborate novel therapeutic concepts for the treatment of this pediatric liver tumor.
Collapse
|
9
|
Breig O, Yates M, Neaud V, Couchy G, Grigoletto A, Lucchesi C, Prox J, Zucman-Rossi J, Becker-Pauly C, Rosenbaum J. Metalloproteinase meprin α regulates migration and invasion of human hepatocarcinoma cells and is a mediator of the oncoprotein Reptin. Oncotarget 2018; 8:7839-7851. [PMID: 27999200 PMCID: PMC5352365 DOI: 10.18632/oncotarget.13975] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/30/2016] [Indexed: 01/13/2023] Open
Abstract
Hepatocellular carcinoma is associated with a high rate of intra-hepatic invasion that carries a poor prognosis. Meprin alpha (Mep1A) is a secreted metalloproteinase with many substrates relevant to cancer invasion. We found that Mep1A was a target of Reptin, a protein that is oncogenic in HCC. We studied Mep1A regulation by Reptin, its role in HCC, and whether it mediates Reptin oncogenic effects. MepA and Reptin expression was measured in human HCC by qRT-PCR and in cultured cells by PCR, western blot and enzymatic activity measurements. Cell growth was assessed by counting and MTS assay. Cell migration was measured in Boyden chambers and wound healing assays, and cell invasion in Boyden chambers. Silencing Reptin decreased Mep1A expression and activity, without affecting meprin β. Mep1A, but not meprin β, was overexpressed in a series of 242 human HCC (2.04 fold, p < 0.0001), and a high expression correlated with a poor prognosis. Mep1A and Reptin expressions were positively correlated (r = 0.39, p < 0.0001). Silencing Mep1A had little effect on cell proliferation, but decreased cell migration and invasion of HuH7 and Hep3B cells. Conversely, overexpression of Mep1A or addition of recombinant Mep1A increased migration and invasion. Finally, overexpression of Mep1A restored a normal cell migration in cells where Reptin was depleted. Mep1A is overexpressed in most HCC and induces HCC cell migration and invasion. Mep1A expression is regulated by Reptin, and Mep1A mediates Reptin-induced migration. Overall, we suggest that Mep1A may be a useful target in HCC.
Collapse
Affiliation(s)
- Osman Breig
- University Bordeaux, INSERM, U1053, BordeAux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Maïlyn Yates
- University Bordeaux, INSERM, U1053, BordeAux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Véronique Neaud
- University Bordeaux, INSERM, U1053, BordeAux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Gabrielle Couchy
- Inserm, U1162, Génomique Fonctionnelle des Tumeurs Solides, Université Paris Diderot, Université Paris Descartes, Université Paris 13, Paris, France
| | - Aude Grigoletto
- University Bordeaux, INSERM, U1053, BordeAux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | | | - Johannes Prox
- Unit for Degradomics of the Protease Web, University of Kiel, Germany
| | - Jessica Zucman-Rossi
- Inserm, U1162, Génomique Fonctionnelle des Tumeurs Solides, Université Paris Diderot, Université Paris Descartes, Université Paris 13, Paris, France
| | | | - Jean Rosenbaum
- University Bordeaux, INSERM, U1053, BordeAux Research in Translational Oncology, BaRITOn, Bordeaux, France
| |
Collapse
|
10
|
Hou H, Gou X, Bu J, Su Y, Wei X, Wang X, Hou B. Meprin α combined with CEA and CA19-9 improves prognostic prediction for surgically treated colorectal cancer patients. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:10441-10450. [PMID: 31966381 PMCID: PMC6965749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/16/2017] [Indexed: 06/10/2023]
Abstract
BACKGROUND Carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) are generally used as tumour markers in patients with colorectal cancer (CRC), and meprin α might be an additional marker. METHODS The preoperative expression of serum CEA and CA19-9 was evaluated using a C12 protein biochip system, and tissue meprin α expression in CRC cells was detected by immunohistochemistry. The relationships of these indexes with clinicopathological parameters and the survival of CRC patients were analysed. RESULTS Of the 147 CRC patients, the preoperative seropositive rates for CEA and CA19-9 were 51.70% and 44.22%, respectively, and the tissue meprin α positive rate was 39.46%. Preoperative seropositivity for CEA was correlated with tumour size (P = 0.019), T stage (P = 0.005) and staging of CRC based on the American Joint Committee on Cancer (AJCC) guidelines (P = 0.032). The preoperative seropositive rate for CA19-9 was correlated with AJCC tumour stage (P = 0.031). High expression of meprin α was significantly correlated with distant CRC metastasis (P = 0.003), serum CEA (P = 0.002) and serum CA19-9 (P = 0.001). The combination of the three markers was an independent prognostic factor in patients with CRC (HR 3.985, 95% CI 1.106-14.361, P = 0.035 for overall survival). CONCLUSIONS Tissue meprin α expression may be a useful predictor of metastasis and prognosis in CRC. The combined detection of the three markers may also be helpful to improve the accuracy of CRC prognosis monitoring.
Collapse
Affiliation(s)
- Hongfa Hou
- Department of Gastrointestinal Surgery, Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Guangdong Province, China
| | - Xinmin Gou
- Department of Pathology, Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Guangdong Province, China
| | - Juyuan Bu
- Department of Gastrointestinal Surgery, Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Guangdong Province, China
| | - Yonghui Su
- Department of Gastrointestinal Surgery, Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Guangdong Province, China
| | - Xinying Wei
- Department of Gastrointestinal Surgery, Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Guangdong Province, China
| | - Xiao Wang
- Department of Gastrointestinal Surgery, Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Guangdong Province, China
| | - Bingzong Hou
- Department of Gastrointestinal Surgery, Fifth Affiliated Hospital of Sun Yat-sen UniversityZhuhai, Guangdong Province, China
| |
Collapse
|
11
|
Xu Y, Wang Y, Liu H, Shi Q, Zhu D, Amos CI, Fang S, Lee JE, Hyslop T, Li X, Han J, Wei Q. Genetic variants in the metzincin metallopeptidase family genes predict melanoma survival. Mol Carcinog 2017; 57:22-31. [PMID: 28796414 DOI: 10.1002/mc.22716] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/08/2017] [Indexed: 12/31/2022]
Abstract
Metzincins are key molecules in the degradation of the extracellular matrix and play an important role in cellular processes such as cell migration, adhesion, and cell fusion of malignant tumors, including cutaneous melanoma (CM). We hypothesized that genetic variants of the metzincin metallopeptidase family genes would be associated with CM-specific survival (CMSS). To test this hypothesis, we first performed Cox proportional hazards regression analysis to evaluate the associations between genetic variants of 75 metzincin metallopeptidase family genes and CMSS using the dataset from the genome-wide association study (GWAS) from The University of Texas MD Anderson Cancer Center (MDACC) which included 858 non-Hispanic white patients with CM, and then validated using the dataset from the Harvard GWAS study which had 409 non-Hispanic white patients with invasive CM. Four independent SNPs (MMP16 rs10090371 C>A, ADAMTS3 rs788935 T>C, TLL2 rs10882807 T>C and MMP9 rs3918251 A>G) were identified as predictors of CMSS, with a variant-allele attributed hazards ratio (HR) of 1.73 (1.32-2.29, 9.68E-05), 1.46 (1.15-1.85, 0.002), 1.68 (1.31-2.14, 3.32E-05) and 0.67 (0.51-0.87, 0.003), respectively, in the meta-analysis of these two GWAS studies. Combined analysis of risk genotypes of these four SNPs revealed a decreased CMSS in a dose-response manner as the number of risk genotypes increased (Ptrend < 0.001). An improvement was observed in the prediction model (area under the curve [AUC] = 81.4% vs. 78.6%), when these risk genotypes were added to the model containing non-genotyping variables. Our findings suggest that these genetic variants may be promising prognostic biomarkers for CMSS.
Collapse
Affiliation(s)
- Yinghui Xu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China.,Department of Medicine, Duke University School of Medicine, Durham, North Carolina.,Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Yanru Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China.,Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Hongliang Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China.,Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Qiong Shi
- Department of Dermatology, Xijing Hospital, Xi'an, Shanxi, China
| | - Dakai Zhu
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Christopher I Amos
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Terry Hyslop
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China.,Department of Biostatistics and Bioinformatics, Duke University and Duke Clinical Research Institute, Durham, North Carolina
| | - Xin Li
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Jiali Han
- Department of Epidemiology, Fairbanks School of Public Health, and Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Qingyi Wei
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China.,Department of Medicine, Duke University School of Medicine, Durham, North Carolina.,Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
12
|
Mao YQ, Houry WA. The Role of Pontin and Reptin in Cellular Physiology and Cancer Etiology. Front Mol Biosci 2017; 4:58. [PMID: 28884116 PMCID: PMC5573869 DOI: 10.3389/fmolb.2017.00058] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/03/2017] [Indexed: 12/29/2022] Open
Abstract
Pontin (RUVBL1, TIP49, TIP49a, Rvb1) and Reptin (RUVBL2, TIP48, TIP49b, Rvb2) are highly conserved ATPases of the AAA+ (ATPases Associated with various cellular Activities) superfamily and are involved in various cellular processes that are important for oncogenesis. First identified as being upregulated in hepatocellular carcinoma and colorectal cancer, their overexpression has since been shown in multiple cancer types such as breast, lung, gastric, esophageal, pancreatic, kidney, bladder as well as lymphatic, and leukemic cancers. However, their exact functions are still quite unknown as they interact with many molecular complexes with vastly different downstream effectors. Within the nucleus, Pontin and Reptin participate in the TIP60 and INO80 complexes important for chromatin remodeling. Although not transcription factors themselves, Pontin and Reptin modulate the transcriptional activities of bona fide proto-oncogenes such as MYC and β-catenin. They associate with proteins involved in DNA damage repair such as PIKK complexes as well as with the core complex of Fanconi anemia pathway. They have also been shown to be important for cell cycle progression, being involved in assembly of telomerase, mitotic spindle, RNA polymerase II, and snoRNPs. When the two ATPases localize to the cytoplasm, they were reported to promote cancer cell invasion and metastasis. Due to their various roles in carcinogenesis, it is not surprising that Pontin and Reptin are proving to be important biomarkers for diagnosis and prognosis of various cancers. They are also current targets for the development of new therapeutic anticancer drugs.
Collapse
Affiliation(s)
- Yu-Qian Mao
- Department of Biochemistry, University of TorontoToronto, ON, Canada
| | - Walid A Houry
- Department of Biochemistry, University of TorontoToronto, ON, Canada.,Department of Chemistry, University of TorontoToronto, ON, Canada
| |
Collapse
|
13
|
Arnold P, Boll I, Rothaug M, Schumacher N, Schmidt F, Wichert R, Schneppenheim J, Lokau J, Pickhinke U, Koudelka T, Tholey A, Rabe B, Scheller J, Lucius R, Garbers C, Rose-John S, Becker-Pauly C. Meprin Metalloproteases Generate Biologically Active Soluble Interleukin-6 Receptor to Induce Trans-Signaling. Sci Rep 2017; 7:44053. [PMID: 28276471 PMCID: PMC5343444 DOI: 10.1038/srep44053] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/03/2017] [Indexed: 12/27/2022] Open
Abstract
Soluble Interleukin-6 receptor (sIL-6R) mediated trans-signaling is an important pro-inflammatory stimulus associated with pathological conditions, such as arthritis, neurodegeneration and inflammatory bowel disease. The sIL-6R is generated proteolytically from its membrane bound form and A Disintegrin And Metalloprotease (ADAM) 10 and 17 were shown to perform ectodomain shedding of the receptor in vitro and in vivo. However, under certain conditions not all sIL-6R could be assigned to ADAM10/17 activity. Here, we demonstrate that the IL-6R is a shedding substrate of soluble meprin α and membrane bound meprin β, resulting in bioactive sIL-6R that is capable of inducing IL-6 trans-signaling. We determined cleavage within the N-terminal part of the IL-6R stalk region, distinct from the cleavage site reported for ADAM10/17. Interestingly, meprin β can be shed from the cell surface by ADAM10/17 and the observation that soluble meprin β is not capable of shedding the IL-6R suggests a regulatory mechanism towards trans-signaling. Additionally, we observed a significant negative correlation of meprin β expression and IL-6R levels on human granulocytes, providing evidence for in vivo function of this proteolytic interaction.
Collapse
Affiliation(s)
- Philipp Arnold
- Institute of Anatomy, University of Kiel, 24118 Kiel, Germany
| | - Inga Boll
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | - Michelle Rothaug
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | - Neele Schumacher
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | | | - Rielana Wichert
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | | | - Juliane Lokau
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | - Ute Pickhinke
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | - Tomas Koudelka
- Systematic Proteomics &Bioanalytics; Institute of Experimental Medicine; University of Kiel, 24105 Kiel, Germany
| | - Andreas Tholey
- Systematic Proteomics &Bioanalytics; Institute of Experimental Medicine; University of Kiel, 24105 Kiel, Germany
| | - Björn Rabe
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Ralph Lucius
- Institute of Anatomy, University of Kiel, 24118 Kiel, Germany
| | | | - Stefan Rose-John
- Institute of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | | |
Collapse
|
14
|
Kistowski M, Dębski J, Karczmarski J, Paziewska A, Olędzki J, Mikula M, Ostrowski J, Dadlez M. A Strong Neutrophil Elastase Proteolytic Fingerprint Marks the Carcinoma Tumor Proteome. Mol Cell Proteomics 2016; 16:213-227. [PMID: 27927741 DOI: 10.1074/mcp.m116.058818] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 11/12/2016] [Indexed: 12/18/2022] Open
Abstract
Proteolytic cascades are deeply involved in critical stages of cancer progression. During the course of peptide-wise analysis of shotgun proteomic data sets representative of colon adenocarcinoma (AC) and ulcerative colitis (UC), we detected a cancer-specific proteolytic fingerprint composed of a set of numerous protein fragments cleaved C-terminally to V, I, A, T, or C residues, significantly overrepresented in AC. A peptide set linked by a common VIATC cleavage consensus was the only prominent cancer-specific proteolytic fingerprint detected. This sequence consensus indicated neutrophil elastase as a source of the fingerprint. We also found that a large fraction of affected proteins are RNA processing proteins associated with the nuclear fraction and mostly cleaved within their functionally important RNA-binding domains. Thus, we detected a new class of cancer-specific peptides that are possible markers of tumor-infiltrating neutrophil activity, which often correlates with the clinical outcome. Data are available via ProteomeXchange with identifiers: PXD005274 (Data set 1) and PXD004249 (Data set 2). Our results indicate the value of peptide-wise analysis of large global proteomic analysis data sets as opposed to protein-wise analysis, in which outlier differential peptides are usually neglected.
Collapse
Affiliation(s)
- Michał Kistowski
- From the ‡Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5A, 02-106 Warsaw
| | - Janusz Dębski
- From the ‡Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5A, 02-106 Warsaw
| | - Jakub Karczmarski
- §Department of Genetics, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Wilhelma Konrada Roentgena 5, 02-781 Warsaw, Poland
| | - Agnieszka Paziewska
- §Department of Genetics, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Wilhelma Konrada Roentgena 5, 02-781 Warsaw, Poland
| | - Jacek Olędzki
- From the ‡Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5A, 02-106 Warsaw
| | - Michał Mikula
- §Department of Genetics, Maria Skłodowska-Curie Memorial Cancer Center and Institute of Oncology, Wilhelma Konrada Roentgena 5, 02-781 Warsaw, Poland
| | - Jerzy Ostrowski
- ¶Department of Gastroenterology Hepatology and Clinical Oncology, Medical Center for Postgraduate Education, Warsaw, Poland
| | - Michał Dadlez
- From the ‡Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5A, 02-106 Warsaw;
| |
Collapse
|
15
|
OuYang HY, Xu J, Luo J, Zou RH, Chen K, Le Y, Zhang YF, Wei W, Guo RP, Shi M. MEP1A contributes to tumor progression and predicts poor clinical outcome in human hepatocellular carcinoma. Hepatology 2016; 63:1227-39. [PMID: 26660154 DOI: 10.1002/hep.28397] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 12/10/2015] [Indexed: 12/24/2022]
Abstract
UNLABELLED Although many staging classifications have been proposed for hepatocellular carcinoma (HCC), determining a patient's prognosis in clinical practice is a challenge due to the molecular diversity of HCC. We investigated the relationship between MEP1A, a candidate oncogene, and clinical outcomes of HCC patients; furthermore, we explored the role of MEP1A in HCC. In this report, it was demonstrated by quantitative real-time polymerase chain reaction that MEP1A messenger RNA levels were significantly elevated in HCC tumor tissues compared with matched adjacent nonneoplastic tissues and nonmalignant liver disease tissues. Immunohistochemical analyses of tissue samples from two independent groups of 394 HCC patients showed that positive expression of MEP1A in tumor cells was an independent and significant risk factor affecting survival after curative resection in both cohort 1 (hazard ratio = 2.05, 95% confidence interval 1.427-2.946; P < 0.001) and cohort 2 (hazard ratio = 1.89, 95% confidence interval 1.260-2.833; P = 0.002). Analysis of Barcelona Clinic Liver Cancer stage 0-A subgroup further showed that patients with positive MEP1A expression in tumor cells had poorer surgical prognoses than those with negative MEP1A expression in tumor cells (cohort 1 P = 0.001, cohort 2 P < 0.001). Both in vitro and in vivo assays showed that MEP1A promoted HCC cell proliferation, migration, and invasion. Further analyses found that MEP1A played an important role in regulating cytoskeletal events and induced epithelial-mesenchymal transition in HCC cells. CONCLUSION MEP1A is a novel prognostic predictor in HCC and plays an important role in the development and progression of HCC.
Collapse
Affiliation(s)
- Han-Yue OuYang
- Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China.,State Key Laboratory of Oncology in South China, Guangzhou, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China
| | - Jing Xu
- State Key Laboratory of Oncology in South China, Guangzhou, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China
| | - Jun Luo
- Department of Proctology, The Sixth Affiliated Hospital of Sun Yat-sen University (Gastrointestinal and Anal Hospital of Sun Yat-sen University), Guangzhou, P.R. China
| | - Ru-Hai Zou
- Department of Ultrasound, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China
| | - Keng Chen
- Department of Hepatopathy, The Eighth People's Hospital of Guangzhou, Guangzhou, P.R. China
| | - Yong Le
- Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yong-Fa Zhang
- Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China
| | - Wei Wei
- Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China
| | - Rong-Ping Guo
- Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China
| | - Ming Shi
- Department of Hepatobiliary Oncology, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China.,State Key Laboratory of Oncology in South China, Guangzhou, P.R. China.,Collaborative Innovation Center for Cancer Medicine, Cancer Center, Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
16
|
Kumar N, Nakagawa P, Janic B, Romero CA, Worou ME, Monu SR, Peterson EL, Shaw J, Valeriote F, Ongeri EM, Niyitegeka JMV, Rhaleb NE, Carretero OA. The anti-inflammatory peptide Ac-SDKP is released from thymosin-β4 by renal meprin-α and prolyl oligopeptidase. Am J Physiol Renal Physiol 2016; 310:F1026-34. [PMID: 26962108 DOI: 10.1152/ajprenal.00562.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/07/2016] [Indexed: 11/22/2022] Open
Abstract
N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is a natural tetrapeptide with anti-inflammatory and antifibrotic properties. Previously, we have shown that prolyl oligopeptidase (POP) is involved in the Ac-SDKP release from thymosin-β4 (Tβ4). However, POP can only hydrolyze peptides shorter than 30 amino acids, and Tβ4 is 43 amino acids long. This indicates that before POP hydrolysis takes place, Tβ4 is hydrolyzed by another peptidase that releases NH2-terminal intermediate peptide(s) with fewer than 30 amino acids. Our peptidase database search pointed out meprin-α metalloprotease as a potential candidate. Therefore, we hypothesized that, prior to POP hydrolysis, Tβ4 is hydrolyzed by meprin-α. In vitro, we found that the incubation of Tβ4 with both meprin-α and POP released Ac-SDKP, whereas no Ac-SDKP was released when Tβ4 was incubated with either meprin-α or POP alone. Incubation of Tβ4 with rat kidney homogenates significantly released Ac-SDKP, which was blocked by the meprin-α inhibitor actinonin. In addition, kidneys from meprin-α knockout (KO) mice showed significantly lower basal Ac-SDKP amount, compared with wild-type mice. Kidney homogenates from meprin-α KO mice failed to release Ac-SDKP from Tβ4. In vivo, we observed that rats treated with the ACE inhibitor captopril increased plasma concentrations of Ac-SDKP, which was inhibited by the coadministration of actinonin (vehicle, 3.1 ± 0.2 nmol/l; captopril, 15.1 ± 0.7 nmol/l; captopril + actinonin, 6.1 ± 0.3 nmol/l; P < 0.005). Similar results were obtained with urinary Ac-SDKP after actinonin treatment. We conclude that release of Ac-SDKP from Tβ4 is mediated by successive hydrolysis involving meprin-α and POP.
Collapse
Affiliation(s)
- Nitin Kumar
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Pablo Nakagawa
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Branislava Janic
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Cesar A Romero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Morel E Worou
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Sumit R Monu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Edward L Peterson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, Michigan
| | - Jiajiu Shaw
- 21st Century Therapeutics, Inc., Detroit, Michigan
| | - Frederick Valeriote
- Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan; and
| | - Elimelda M Ongeri
- Department of Biology, North Carolina A & T State University, Greensboro, North Carolina
| | | | - Nour-Eddine Rhaleb
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Oscar A Carretero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan;
| |
Collapse
|
17
|
Jodal A, Pape F, Becker-Pauly C, Maas O, Schibli R, Béhé M. Evaluation of ¹¹¹in-labelled exendin-4 derivatives containing different meprin β-specific cleavable linkers. PLoS One 2015; 10:e0123443. [PMID: 25855967 PMCID: PMC4391719 DOI: 10.1371/journal.pone.0123443] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/04/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cleavable linkers, which are specifically cleaved by defined conditions or enzymes, are powerful tools that can be used for various purposes. Amongst other things, they have been successfully used to deliver toxic payloads as prodrugs into target tissues. In this work novel linker sequences targeting meprin β, a metalloprotease expressed in the kidney brush-border membrane, were designed and included in the sequence of three radiolabelled exendin-4 derivatives. As radiolabelled exendin-4 derivatives strongly accumulate in the kidneys, we hypothesised that specific cleavage of the radiolabelled moiety at the kidney brush-border membrane would allow easier excretion of the activity into the urine and therefore improve the pharmacological properties of the peptide. RESULTS The insertion of a cleavable linker did not negatively influence the in vitro properties of the peptides. They showed a good affinity to the GLP-1 receptor expressed in CHL cells, a high internalisation and sufficiently high stability in fresh human blood plasma. In vitro digestion with recombinant meprin β rapidly metabolised the corresponding linker sequences. After 60 min the majority of the corresponding peptides were digested and at the same time the anticipated fragments were formed. The peptides were also quickly metabolised in CD1 nu/nu mouse kidney homogenates. Immunofluorescence staining of meprin β in kidney sections confirmed the expression of the protease in the kidney brush-border membrane. Biodistribution in GLP-1 receptor positive tumour-xenograft bearing mice revealed high specific uptake of the 111In-labelled tracers in receptor positive tissue. Accumulation in the kidneys, however, was still high and comparable to the lead compound 111In-Ex4NOD40. CONCLUSION In conclusion, we show that the concept of cleavable linkers specific for meprin β is feasible, as the peptides are rapidly cleaved by the enzyme while retaining their biological properties.
Collapse
Affiliation(s)
- Andreas Jodal
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
| | - Fabienne Pape
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | | - Ole Maas
- Department of Radiology and Nuclear Medicine, Division of Nuclear Medicine, University Hospital Basel, Basel, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Martin Béhé
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
- * E-mail:
| |
Collapse
|
18
|
Urinary signatures of Renal Cell Carcinoma investigated by peptidomic approaches. PLoS One 2014; 9:e106684. [PMID: 25202906 PMCID: PMC4159280 DOI: 10.1371/journal.pone.0106684] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 07/31/2014] [Indexed: 01/16/2023] Open
Abstract
Renal Cell Carcinoma (RCC) is typically asymptomatic and surgery usually increases patient's lifespan only for early stage tumours. Moreover, solid renal masses cannot be confidently differentiated from RCC. Therefore, markers to distinguish malignant kidney tumours and for their detection are needed. Two different peptide signatures were obtained by a MALDI-TOF profiling approach based on urine pre-purification by C8 magnetic beads. One cluster of 12 signals could differentiate malignant tumours (n = 137) from benign renal masses and controls (n = 153) with sensitivity of 76% and specificity of 87% in the validation set. A second cluster of 12 signals distinguished clear cell RCC (n = 118) from controls (n = 137) with sensitivity and specificity values of 84% and 91%, respectively. Most of the peptide signals used in the two models were observed at higher abundance in patient urines and could be identified as fragments of proteins involved in tumour pathogenesis and progression. Among them: the Meprin 1α with a pro-angiogenic activity, the Probable G-protein coupled receptor 162, belonging to the GPCRs family and known to be associated with several key functions in cancer, the Osteopontin that strongly correlates to tumour stages and invasiveness, the Phosphorylase b kinase regulatory subunit alpha and the SeCreted and TransMembrane protein 1.
Collapse
|
19
|
Identification of interconnected markers for T-cell acute lymphoblastic leukemia. BIOMED RESEARCH INTERNATIONAL 2013; 2013:210253. [PMID: 23956970 PMCID: PMC3727179 DOI: 10.1155/2013/210253] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/04/2013] [Indexed: 12/11/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a complex disease, resulting from proliferation of differentially arrested immature T cells. The molecular mechanisms and the genes involved in the generation of T-ALL remain largely undefined. In this study, we propose a set of genes to differentiate individuals with T-ALL from the nonleukemia/healthy ones and genes that are not differential themselves but interconnected with highly differentially expressed ones. We provide new suggestions for pathways involved in the cause of T-ALL and show that network-based classification techniques produce fewer genes with more meaningful and successful results than expression-based approaches. We have identified 19 significant subnetworks, containing 102 genes. The classification/prediction accuracies of subnetworks are considerably high, as high as 98%. Subnetworks contain 6 nondifferentially expressed genes, which could potentially participate in pathogenesis of T-ALL. Although these genes are not differential, they may serve as biomarkers if their loss/gain of function contributes to generation of T-ALL via SNPs. We conclude that transcription factors, zinc-ion-binding proteins, and tyrosine kinases are the important protein families to trigger T-ALL. These potential disease-causing genes in our subnetworks may serve as biomarkers, alternative to the traditional ones used for the diagnosis of T-ALL, and help understand the pathogenesis of the disease.
Collapse
|
20
|
The metalloproteases meprin α and meprin β: unique enzymes in inflammation, neurodegeneration, cancer and fibrosis. Biochem J 2013; 450:253-64. [PMID: 23410038 PMCID: PMC3573791 DOI: 10.1042/bj20121751] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The metalloproteases meprin α and meprin β exhibit structural and functional features that are unique among all extracellular proteases. Although meprins were discovered more than 30 years ago, their precise substrates and physiological roles have been elusive. Both enzymes were originally found to be highly expressed in kidney and intestine, which focused research on these particular tissues and associated pathologies. Only recently it has become evident that meprins exhibit a much broader expression pattern, implicating functions in angiogenesis, cancer, inflammation, fibrosis and neurodegenerative diseases. Different animal models, as well as proteomics approaches for the identification of protease substrates, have helped to reveal more precise molecular signalling events mediated by meprin activity, such as activation and release of pro-inflammatory cytokines. APP (amyloid precursor protein) is cleaved by meprin β in vivo, reminiscent of the β-secretase BACE1 (β-site APP-cleaving enzyme 1). The subsequent release of Aβ (amyloid β) peptides is thought to be the major cause of the neurodegenerative Alzheimer's disease. On the other hand, ADAM10 (a disintegrin and metalloprotease domain 10), which is the constitutive α-secretase, was shown to be activated by meprin β, which is itself shed from the cell surface by ADAM10. In skin, both meprins are overexpressed in fibrotic tumours, characterized by massive accumulation of fibrillar collagens. Indeed, procollagen III is processed to its mature form by meprin α and meprin β, an essential step in collagen fibril assembly. The recently solved crystal structure of meprin β and the unique cleavage specificity of these proteases identified by proteomics will help to generate specific inhibitors that could be used as therapeutics to target meprins under certain pathological conditions.
Collapse
|
21
|
Geurts N, Becker-Pauly C, Martens E, Proost P, Van den Steen PE, Stöcker W, Opdenakker G. Meprins process matrix metalloproteinase-9 (MMP-9)/gelatinase B and enhance the activation kinetics by MMP-3. FEBS Lett 2012; 586:4264-9. [PMID: 23123160 DOI: 10.1016/j.febslet.2012.10.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 10/18/2012] [Indexed: 01/22/2023]
Abstract
Meprin α and β, members of the astacin family of zinc metalloproteinases, are unique plasma membrane and secreted proteases known to cleave a wide range of biological substrates involved in inflammation, cancer and fibrosis. In this study, we identified proMMP-9 as a novel substrate and show that aminoterminal meprin-mediated clipping improves the activation kinetics of proMMP-9 by MMP-3, an efficient activator of proMMP-9. Interestingly, the NH(2)-terminus LVLFPGDL, generated by incubation with meprin α, is identical to the form produced in conditioned media from human neutrophils and monocytes. Hence, this meprin-mediated processing and enhancement of MMP-9 activation kinetics may have biological relevance in the context of in vivo inflammatory processes.
Collapse
Affiliation(s)
- Nathalie Geurts
- Laboratory of Immunobiology, Rega Institute for Medical Research, University of Leuven, Minderbroedersstraat 10, 3000 Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
22
|
Structural basis for the sheddase function of human meprin β metalloproteinase at the plasma membrane. Proc Natl Acad Sci U S A 2012; 109:16131-6. [PMID: 22988105 DOI: 10.1073/pnas.1211076109] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Ectodomain shedding at the cell surface is a major mechanism to regulate the extracellular and circulatory concentration or the activities of signaling proteins at the plasma membrane. Human meprin β is a 145-kDa disulfide-linked homodimeric multidomain type-I membrane metallopeptidase that sheds membrane-bound cytokines and growth factors, thereby contributing to inflammatory diseases, angiogenesis, and tumor progression. In addition, it cleaves amyloid precursor protein (APP) at the β-secretase site, giving rise to amyloidogenic peptides. We have solved the X-ray crystal structure of a major fragment of the meprin β ectoprotein, the first of a multidomain oligomeric transmembrane sheddase, and of its zymogen. The meprin β dimer displays a compact shape, whose catalytic domain undergoes major rearrangement upon activation, and reveals an exosite and a sugar-rich channel, both of which possibly engage in substrate binding. A plausible structure-derived working mechanism suggests that substrates such as APP are shed close to the plasma membrane surface following an "N-like" chain trace.
Collapse
|
23
|
Jefferson T, Auf dem Keller U, Bellac C, Metz VV, Broder C, Hedrich J, Ohler A, Maier W, Magdolen V, Sterchi E, Bond JS, Jayakumar A, Traupe H, Chalaris A, Rose-John S, Pietrzik CU, Postina R, Overall CM, Becker-Pauly C. The substrate degradome of meprin metalloproteases reveals an unexpected proteolytic link between meprin β and ADAM10. Cell Mol Life Sci 2012; 70:309-33. [PMID: 22940918 PMCID: PMC3535375 DOI: 10.1007/s00018-012-1106-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 07/19/2012] [Accepted: 07/23/2012] [Indexed: 01/10/2023]
Abstract
The in vivo roles of meprin metalloproteases in pathophysiological conditions remain elusive. Substrates define protease roles. Therefore, to identify natural substrates for human meprin α and β we employed TAILS (terminal amine isotopic labeling of substrates), a proteomics approach that enriches for N-terminal peptides of proteins and cleavage fragments. Of the 151 new extracellular substrates we identified, it was notable that ADAM10 (a disintegrin and metalloprotease domain-containing protein 10)-the constitutive α-secretase-is activated by meprin β through cleavage of the propeptide. To validate this cleavage event, we expressed recombinant proADAM10 and after preincubation with meprin β, this resulted in significantly elevated ADAM10 activity. Cellular expression in murine primary fibroblasts confirmed activation. Other novel substrates including extracellular matrix proteins, growth factors and inhibitors were validated by western analyses and enzyme activity assays with Edman sequencing confirming the exact cleavage sites identified by TAILS. Cleavages in vivo were confirmed by comparing wild-type and meprin(-/-) mice. Our finding of cystatin C, elafin and fetuin-A as substrates and natural inhibitors for meprins reveal new mechanisms in the regulation of protease activity important for understanding pathophysiological processes.
Collapse
Affiliation(s)
- Tamara Jefferson
- Institute of Biochemistry, Christian-Albrechts-University, 24118, Kiel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Minder P, Bayha E, Becker-Pauly C, Sterchi EE. Meprinα transactivates the epidermal growth factor receptor (EGFR) via ligand shedding, thereby enhancing colorectal cancer cell proliferation and migration. J Biol Chem 2012; 287:35201-35211. [PMID: 22923609 DOI: 10.1074/jbc.m112.368910] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Meprinα, an astacin-type metalloprotease is overexpressed in colorectal cancer cells and is secreted in a non-polarized fashion, leading to the accumulation of meprinα in the tumor stroma. The transition from normal colonocytes to colorectal cancer correlates with increased meprinα activity at primary tumor sites. A role for meprinα in invasion and metastatic dissemination is supported by its pro-angiogenic and pro-migratory activity. In the present study, we provide evidence for a meprinα-mediated transactivation of the EGFR signaling pathway and suggest that this mechanism is involved in colorectal cancer progression. Using alkaline phosphatase-tagged EGFR ligands and an ELISA assay, we demonstrate that meprinα is capable of shedding epidermal growth factor (EGF) and transforming growth factor-α (TGFα) from the plasma membrane. Shedding was abrogated using actinonin, an inhibitor for meprinα. The physiological effects of meprinα-mediated shedding of EGF and TGFα were investigated with human colorectal adenocarcinoma cells (Caco-2). Proteolytically active meprinα leads to an increase in EGFR and ERK1/2 phosphorylation and subsequently enhances cell proliferation and migration. In conclusion, the implication of meprinα in the EGFR/MAPK signaling pathway indicates a role of meprinα in colorectal cancer progression.
Collapse
Affiliation(s)
- Petra Minder
- Institute of Biochemistry and Molecular Medicine,University of Bern, Buehlstrasse 28, CH-3012 Bern, Switzerland
| | - Elke Bayha
- Institute of Biochemistry and Molecular Medicine,University of Bern, Buehlstrasse 28, CH-3012 Bern, Switzerland
| | - Christoph Becker-Pauly
- Institute of Biochemistry, Christian-Albrechts-University, Rudolf-Hoeber-Strasse 1, 24118 Kiel, Germany
| | - Erwin E Sterchi
- Institute of Biochemistry and Molecular Medicine,University of Bern, Buehlstrasse 28, CH-3012 Bern, Switzerland.
| |
Collapse
|