1
|
Nandy D, Shirude MB, S A, Devarajan A, Mukherjee A, Dutta D. Nuclear localization of APLF facilitates breast cancer metastasis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167537. [PMID: 39384105 DOI: 10.1016/j.bbadis.2024.167537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
Most breast cancer deaths result from metastases. We previously reported that DNA repair factor and histone chaperone Aprataxin PNK-like Factor (APLF) is involved in EMT-associated metastasis of triple negative breast cancer (TNBC) cells. However, non-metastatic cells also expressed APLF, the implications of which in disease advancement remain uncertain. Here, we demonstrate that the metastatic prognosis of breast cancer cells may be determined by the cellular localization of APLF. Using TNBC patient samples and cell lines, we discovered that APLF was localized in the nucleus and cytoplasm, whereas other subtypes of breast cancer had cytosolic or perinuclear localization. To investigate metastatic properties in vitro and in vivo, we modeled APLF differential localization by stably producing APLF-tagged nuclear localization signal (NLS) in the luminal subtype MCF7 cells in the absence of putative APLF NLS. Nuclear APLF in non-metastatic MCF7 cells demonstrated pronounced migration, invasion and metastatic potential. We obtained the mechanistic insight from molecular studies that PARP1 could facilitate the transport of APLF from the cytosol to the nucleus, assisting in the metastasis of TNBC cells linked with EMT. Inhibition of PARP1 enzymatic activity with olaparib abrogated the nuclear expression of APLF with loss in expression of genes associated with EMT. Thus, our findings reveal that cellular localization of APLF may predict the risk of breast cancer to metastasize and hence could be exploited to determine the disease progression. We anticipate that the inhibition of cytosolic PARP1-APLF interaction may potentially aid in the prevention of breast cancer metastasis in TNBC patients.
Collapse
Affiliation(s)
- Debparna Nandy
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram 695014, Kerala, India; Manipal Academy of Higher Education, Manipal, Karnataka State 576104, India
| | - Mayur Balkrishna Shirude
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram 695014, Kerala, India; Manipal Academy of Higher Education, Manipal, Karnataka State 576104, India
| | - Archana S
- Rajiv Gandhi Centre for Biotechnology (RGCB), Animal Research Facility, Thycaud PO, Poojappura, Thiruvananthapuram 695014, Kerala, India
| | - Anjali Devarajan
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram 695014, Kerala, India
| | - Ananda Mukherjee
- Rajiv Gandhi Centre for Biotechnology (RGCB), Cancer Research Program, Thycaud PO, Poojappura, Thiruvananthapuram 695014, Kerala, India
| | - Debasree Dutta
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram 695014, Kerala, India.
| |
Collapse
|
2
|
Demir I, Yilmaz I, Horoz E, Calik B, Bilgir O. Matriptase as a potential biomarker and therapeutic target in newly diagnosed type 2 diabetes mellitus. Ir J Med Sci 2024; 193:223-230. [PMID: 37418107 DOI: 10.1007/s11845-023-03441-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/26/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder that affects the processing of carbohydrates, proteins, and lipids. In T2DM, metabolic dysregulation occurs through various pathways caused by increased levels of many adipokines and inflammatory chemokines. Impaired insulin-glucose metabolism occurs in tissues. The proteolytic enzyme matriptase is thought to be closely related to glucose metabolism due to its glycolization sites. AIM Our study aimed to evaluate the correlation between matriptase, a proteolytic enzyme, and metabolic parameters in individuals recently diagnosed with T2DM. We also sought to investigate the potential involvement of matriptase in the development of diabetes. METHODS We measured all participants' metabolic laboratory parameters, including basic biochemical tests, hemograms, high-sensitivity C-reactive protein (hsCRP), and matriptase levels. RESULTS Our results showed a significant increase in circulating matriptase levels in individuals with T2DM compared to the control group. Furthermore, individuals with metabolic syndrome had significantly higher matriptase levels than those without in the T2DM and control groups. We also observed that T2DM patients had elevated levels of Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), hsCRP, and matriptase, which displayed a positive correlation. CONCLUSION Our study is the first to report elevated levels of matriptase in individuals with newly diagnosed T2DM and/or metabolic syndrome. Additionally, we found a significant positive correlation between matriptase levels and metabolic and inflammatory parameters, indicating a potential role for matriptase in the pathogenesis of T2DM and glucose metabolism. Further research on matriptase could lead to its recognition as a novel target for investigation.
Collapse
Affiliation(s)
- Ismail Demir
- Department of Internal Medicine, Health Sciences University, Izmir, Bozyaka Training and Research Hospital, 35170, Karabaglar, Izmir, Turkey.
| | - Ismail Yilmaz
- Faculty of Medicine, Department of Pharmacology and Toxicology, Izmir Kâtip Celebi University, Izmir, Turkey
| | - Ersan Horoz
- Faculty of Medicine, Department of Pharmacology and Toxicology, Izmir Kâtip Celebi University, Izmir, Turkey
| | - Bulent Calik
- Department of General Surgery, Health Sciences University Izmir, Bozyaka Training and Research Hospital, Izmir, Turkey
| | - Oktay Bilgir
- Department of Internal Medicine, Health Sciences University, Izmir, Bozyaka Training and Research Hospital, 35170, Karabaglar, Izmir, Turkey
| |
Collapse
|
3
|
Santos NRM, de Oliveira WF, Cabrera MP, Bezerra Filho CM, Patriota LLS, Napoleão TH, Paiva PMG, Oliva MLV, Cabral Filho PE, Fontes A, Correia MTS. A fluorescent quantum dot conjugate to probe the interaction of Enterolobium contortisiliquum trypsin inhibitor with cancer cells. Int J Biol Macromol 2023; 252:126453. [PMID: 37619683 DOI: 10.1016/j.ijbiomac.2023.126453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/10/2023] [Accepted: 08/20/2023] [Indexed: 08/26/2023]
Abstract
Serine proteases play crucial biological roles and have their activity controlled by inhibitors, such as the EcTI, a serine protease inhibitor purified from Enterolobium contortisiliquum seeds, which has anticancer activity. This study aimed to conjugate EcTI with quantum dots (QDs), fluorophores with outstanding optical properties, and investigate the interaction of QDs-EcTI nanoprobe with cancer cells. The conjugation was evaluated by fluorescence correlation spectroscopy (FCS) and fluorescence microplate assay (FMA). EcTI inhibitory activity after interaction with QDs was also analyzed. From FCS, the conjugate presented a hydrodynamic diameter about 4× greater than bare QDs, suggesting a successful conjugation. This was supported by FMA, which showed a relative fluorescence intensity of ca. 3815% for the nanosystem, concerning bare QDs or EcTI alone. The EcTI inhibitory activity remained intact after its interaction with QDs. From flow cytometry analyses, approximately 62% of MDA-MB-231 and 90% of HeLa cells were labeled with the QD-EcTI conjugate, suggesting that their membranes have different protease levels to which EcTI exhibits an affinity. Concluding, the QD-EcTI represents a valuable nanotool to study the interaction of this inhibitor with cancer cells using fluorescence-based techniques with the potential to unravel the intricate dynamics of interplays between proteases and inhibitors in cancer biology.
Collapse
Affiliation(s)
- Natália R M Santos
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil; Departamento de Biofísica e Radiobiologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil
| | - Weslley F de Oliveira
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil; Departamento de Biofísica e Radiobiologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil
| | - Mariana P Cabrera
- Departamento de Química Fundamental, Centro de Ciências Exatas e da Natureza, Universidade Federal de Pernambuco, Recife, PE, 50740-560, Brazil
| | - Clovis M Bezerra Filho
- Departamento de Bioquímica, Universidade Federal de São Paulo, São Paulo, SP, 04044-020, Brazil; Núcleo de Pesquisas em Ciências Ambientais e Biotecnologia, Universidade Católica de Pernambuco, Recife, PE, 50050-900, Brazil
| | - Leydianne L S Patriota
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil
| | - Thiago H Napoleão
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil
| | - Patrícia M G Paiva
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil
| | - Maria Luiza V Oliva
- Departamento de Bioquímica, Universidade Federal de São Paulo, São Paulo, SP, 04044-020, Brazil
| | - Paulo E Cabral Filho
- Departamento de Biofísica e Radiobiologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil
| | - Adriana Fontes
- Departamento de Biofísica e Radiobiologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil.
| | - Maria T S Correia
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, 50670-901, Brazil.
| |
Collapse
|
4
|
Hatzold J, Nett V, Brantsch S, Zhang JL, Armistead J, Wessendorf H, Stephens R, Humbert PO, Iden S, Hammerschmidt M. Matriptase-dependent epidermal pre-neoplasm in zebrafish embryos caused by a combination of hypotonic stress and epithelial polarity defects. PLoS Genet 2023; 19:e1010873. [PMID: 37566613 PMCID: PMC10446194 DOI: 10.1371/journal.pgen.1010873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/23/2023] [Accepted: 07/17/2023] [Indexed: 08/13/2023] Open
Abstract
Aberrantly up-regulated activity of the type II transmembrane protease Matriptase-1 has been associated with the development and progression of a range of epithelial-derived carcinomas, and a variety of signaling pathways can mediate Matriptase-dependent tumorigenic events. During mammalian carcinogenesis, gain of Matriptase activity often results from imbalanced ratios between Matriptase and its cognate transmembrane inhibitor Hai1. Similarly, in zebrafish, unrestrained Matriptase activity due to loss of hai1a results in epidermal pre-neoplasms already during embryogenesis. Here, based on our former findings of a similar tumor-suppressive role for the Na+/K+-pump beta subunit ATP1b1a, we identify epithelial polarity defects and systemic hypotonic stress as another mode of aberrant Matriptase activation in the embryonic zebrafish epidermis in vivo. In this case, however, a different oncogenic pathway is activated which contains PI3K, AKT and NFkB, rather than EGFR and PLD (as in hai1a mutants). Strikingly, epidermal pre-neoplasm is only induced when epithelial polarity defects in keratinocytes (leading to disturbed Matriptase subcellular localization) occur in combination with systemic hypotonic stress (leading to increased proteolytic activity of Matriptase). A similar combinatorial effect of hypotonicity and loss of epithelial polarity was also obtained for the activity levels of Matriptase-1 in human MCF-10A epithelial breast cells. Together, this is in line with the multi-factor concept of carcinogenesis, with the notion that such factors can even branch off from one and the same initiator (here ATP1a1b) and can converge again at the level of one and the same mediator (here Matriptase). In sum, our data point to tonicity and epithelial cell polarity as evolutionarily conserved regulators of Matriptase activity that upon de-regulation can constitute an alternative mode of Matriptase-dependent carcinogenesis in vivo.
Collapse
Affiliation(s)
- Julia Hatzold
- Institute of Zoology–Developmental Biology, University of Cologne, Germany
| | - Verena Nett
- Cell and Developmental Biology, Center of Human and Molecular Biology (ZHMB), Saarland University, Faculty of Medicine, Homburg/Saar, Germany
| | - Stephanie Brantsch
- Institute of Zoology–Developmental Biology, University of Cologne, Germany
| | - Jin-Li Zhang
- Institute of Zoology–Developmental Biology, University of Cologne, Germany
| | - Joy Armistead
- Institute of Zoology–Developmental Biology, University of Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Cologne, Germany
| | - Heike Wessendorf
- Institute of Zoology–Developmental Biology, University of Cologne, Germany
| | - Rebecca Stephens
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Patrick O. Humbert
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
- Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Sandra Iden
- Cell and Developmental Biology, Center of Human and Molecular Biology (ZHMB), Saarland University, Faculty of Medicine, Homburg/Saar, Germany
| | - Matthias Hammerschmidt
- Institute of Zoology–Developmental Biology, University of Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne, Cologne, Germany
| |
Collapse
|
5
|
The serine protease matriptase inhibits migration and proliferation in multiple myeloma cells. Oncotarget 2022; 13:1175-1186. [PMID: 36268559 PMCID: PMC9584456 DOI: 10.18632/oncotarget.28300] [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: 11/25/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) is an incurable malignancy of plasma cells. The serine protease matriptase is frequently dysregulated in human carcinomas, which facilitates tumor progression and metastatic dissemination. The importance of matriptase in hematological malignancies is yet to be clarified. In this study, we aimed to characterize the role of matriptase in MM. MATERIALS AND METHODS mRNA expression of matriptase and its inhibitors hepatocyte growth factor activator inhibitor (HAI)-1 and HAI-2 was studied in primary MM cells from patient samples and human myeloma cell lines (HMCLs). We further investigated the effect of matriptase on migration and proliferation of myeloma cells in vitro. By use of the CoMMpass database, we assessed the clinical relevance of matriptase in MM patients. RESULTS Matriptase was expressed in 96% of patient samples and all HMCLs tested. Overexpression of matriptase in vitro reduced proliferation, and significantly decreased cytokine-induced migration. Conversely, matriptase knockdown significantly enhanced migration. Mechanistically, overexpression of matriptase inhibited activation of Src kinase. CONCLUSIONS Our findings may suggest a novel role of matriptase as a tumor suppressor in MM pathogenesis.
Collapse
|
6
|
Jang J, Cho EH, Cho Y, Ganzorig B, Kim KY, Kim MG, Kim C. Environment-Sensitive Ectodomain Shedding of Epithin/PRSS14 Increases Metastatic Potential of Breast Cancer Cells by Producing CCL2. Mol Cells 2022; 45:564-574. [PMID: 35950457 PMCID: PMC9385564 DOI: 10.14348/molcells.2022.2004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/20/2021] [Accepted: 01/28/2022] [Indexed: 11/27/2022] Open
Abstract
Epithin/PRSS14 is a membrane serine protease that plays a key role in tumor progression. The protease exists on the cell surface until its ectodomain shedding, which releases most of the extracellular domain. Previously, we showed that the remaining portion on the membrane undergoes intramembrane proteolysis, which results in the liberation of the intracellular domain and the intracellular domainmediated gene expression. In this study, we investigated how the intramembrane proteolysis for the nuclear function is initiated. We observed that ectodomain shedding of epithin/PRSS14 in mouse breast cancer 4T1 cells increased depending on environmental conditions and was positively correlated with invasiveness of the cells and their proinvasive cytokine production. We identified selenite as an environmental factor that can induce ectodomain shedding of the protease and increase C-C motif chemokine ligand 2 (CCL2) secretion in an epithin/PRSS14-dependent manner. Additionally, by demonstrating that the expression of the intracellular domain of epithin/PRSS14 is sufficient to induce CCL2 secretion, we established that epithin/PRSS14- dependent shedding and its subsequent intramembrane proteolysis are responsible for the metastatic conversion of 4T1 cells under these conditions. Consequently, we propose that epithin/PRSS14 can act as an environment-sensing receptor that promotes cancer metastasis by liberating the intracellular domain bearing transcriptional activity under conditions promoting ectodomain shedding.
Collapse
Affiliation(s)
- Jiyoung Jang
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| | - Eun Hye Cho
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Youngkyung Cho
- Department of Life Sciences, Korea University, Seoul 02841, Korea
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Binderya Ganzorig
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Ki Yeon Kim
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Moon Gyo Kim
- Department of Biological Sciences, Inha University, Incheon 22212, Korea
| | - Chungho Kim
- Department of Life Sciences, Korea University, Seoul 02841, Korea
| |
Collapse
|
7
|
Yamashita F, Kaieda T, Shimomura T, Kawaguchi M, Lin C, Johnson MD, Tanaka H, Kiwaki T, Fukushima T, Kataoka H. Role of the polycystic kidney disease domain in matriptase chaperone activity and localization of hepatocyte growth factor activator inhibitor‐1. FEBS J 2022; 289:3422-3439. [DOI: 10.1111/febs.16348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/16/2021] [Accepted: 01/10/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Fumiki Yamashita
- Section of Oncopathology and Regenerative Biology Department of Pathology, Faculty of Medicine, University of Miyazaki Japan
| | - Takashi Kaieda
- Section of Oncopathology and Regenerative Biology Department of Pathology, Faculty of Medicine, University of Miyazaki Japan
- Chitose Laboratory Corp Kanagawa Japan
| | - Takeshi Shimomura
- Section of Oncopathology and Regenerative Biology Department of Pathology, Faculty of Medicine, University of Miyazaki Japan
| | - Makiko Kawaguchi
- Section of Oncopathology and Regenerative Biology Department of Pathology, Faculty of Medicine, University of Miyazaki Japan
| | - Chen‐Yong Lin
- Lambardi Comprehensive Cancer Center Georgetown University, School of Medicine Washington DC USA
| | - Michael D Johnson
- Lambardi Comprehensive Cancer Center Georgetown University, School of Medicine Washington DC USA
| | - Hiroyuki Tanaka
- Section of Oncopathology and Regenerative Biology Department of Pathology, Faculty of Medicine, University of Miyazaki Japan
| | - Takumi Kiwaki
- Section of Oncopathology and Regenerative Biology Department of Pathology, Faculty of Medicine, University of Miyazaki Japan
| | - Tsuyoshi Fukushima
- Section of Oncopathology and Regenerative Biology Department of Pathology, Faculty of Medicine, University of Miyazaki Japan
| | - Hiroaki Kataoka
- Section of Oncopathology and Regenerative Biology Department of Pathology, Faculty of Medicine, University of Miyazaki Japan
| |
Collapse
|
8
|
Jafari-Oliayi A, Dabiri S. SNHG6 203 RNA May be Involved in the Cell Cycle Progression in HER2-Negative Breast Cancer Cells. IRANIAN JOURNAL OF PATHOLOGY 2022; 17:251-260. [PMID: 36247503 PMCID: PMC9508537 DOI: 10.30699/ijp.2022.525346.2607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/18/2022] [Indexed: 11/06/2022]
Abstract
Background & Objective Long noncoding RNAs (lncRNAs) as challenging molecules are more known than those in the last decade. These transcripts have been validated for carcinogenesis in many types of tissue. Functions of lncRNAs in cancer induction include cell cycle, epithelial to mesenchymal transition progression, apoptosis inhibition, cell migration, and invasion stimulation. LncRNA small nucleolar host gene 6 (SNHG6) have been proven as an oncogenic transcript in many types of cancer. Methods RNA extraction was performed for 47 breast specimens in patients with cancer and cDNAs were synthesized. Relative expression of target variants was determined by qPCR and calculated based on the ΔΔCt method. SNHG6 203 was cloned into pcDNA 3.1+ vector for overexpression in MCF7 (HER2-) and SK-BR3 (HER2+) cells. The cell cycle progression of transfected cells was assessed by flow cytometry. Cell migration ability of transfected cells was evaluated by the scratch method and Image J software. Finally, cell viability was assessed by the MTT method. Results Among four splice variants of SNHG6 (202, 203, 204, and 207), SNHG6 203 was proved as an overexpressed splice variant in breast tumors. This transcript was expressed in HER2-negative breast tumors more frequently than in the positive ones. Overexpression of this variant in target cells resulted in cell cycle progression of MCF7 as HER2-negative cells. Moreover, the overexpression of SNHG6 203 led to lower migration ability of MCF7 cells and a non-significant reduction of their viability as HER2-negative breast cancer cells. Conclusion Our results revealed that SNHG6 203 may be involved in the carcinogenesis of HER2-negative breast cancers via cell cycle progression.
Collapse
Affiliation(s)
- Amin Jafari-Oliayi
- Corresponding Information: Amin Jafari-Oliayi, Pathology and Stem Cell Research Center, Department of Pathology, Afzalipour Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
9
|
Kim JM, Park J, Noh EM, Song HK, Kang SY, Jung SH, Kim JS, Youn HJ, Lee YR. Downregulation of matriptase suppresses the PAR‑2/PLCγ2/PKC‑mediated invasion and migration abilities of MCF‑7 breast cancer cells. Oncol Rep 2021; 46:247. [PMID: 34608498 PMCID: PMC8524316 DOI: 10.3892/or.2021.8198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/13/2021] [Indexed: 12/13/2022] Open
Abstract
Matriptases, members of the type II transmembrane serine protease family, are cell surface proteolytic enzymes that mediate tumor invasion and metastasis. Matriptase is highly expressed in breast cancer and is associated with poor patient outcome. However, the cellular mechanism by which matriptase mediates breast cancer invasion remains unknown. The present study aimed to determine the role of matriptase in the protein kinase C (PKC)‑mediated metastasis of MCF‑7 human breast cancer cells. Matriptase small interfering RNA‑mediated knockdown significantly attenuated the 12‑O‑tetradecanoylphorbol‑13‑acetate (TPA)‑induced invasiveness and migration of MCF‑7 cells, and inhibited the activation of phospholipase C γ2 (PLCγ2)/PKC/MAPK signaling pathways. Matriptase‑knockdown also suppressed the expression of MMP‑9 and inhibited the activation of NF‑κB/activator protein‑1 in MCF‑7 cells. Additionally, GB83 [an inhibitor of protease‑activated receptor‑2 (PAR‑2)] inhibited PKC‑mediated MMP‑9 expression and metastatic ability in MCF‑7 cells. Furthermore, downregulation of matriptase suppressed TPA‑induced MMP‑9 expression and invasiveness via PAR‑2/PLCγ2/PKC/MAPK activation. These findings shed light on the mechanism underlying the role of matriptase in MCF‑7 cell invasion and migration ability, and suggest that matriptase modulation could be a promising therapeutic strategy for preventing breast cancer metastasis.
Collapse
Affiliation(s)
- Jeong-Mi Kim
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju, Jeollabuk 54896, Republic of Korea
| | - Jinny Park
- Department of Internal Medicine, Division of Hematology, Gil Medical Center, Gachon University College of Medicine, Incheon 405‑760, Republic of Korea
| | - Eun-Mi Noh
- Department of Oral Biochemistry, School of Dentistry, Wonkwang University, Iksan, Jeollabuk 570‑749, Republic of Korea
| | - Hyun-Kyung Song
- Department of Oral Biochemistry, School of Dentistry, Wonkwang University, Iksan, Jeollabuk 570‑749, Republic of Korea
| | - Sang Yull Kang
- Department of Surgery, Research Institute of Clinical Medicine, Jeonbuk National University Hospital, Jeonbuk National University and Biomedical Research Institute, Jeonju, Jeollabuk 560‑182, Republic of Korea
| | - Sung Hoo Jung
- Department of Surgery, Research Institute of Clinical Medicine, Jeonbuk National University Hospital, Jeonbuk National University and Biomedical Research Institute, Jeonju, Jeollabuk 560‑182, Republic of Korea
| | - Jong-Suk Kim
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju, Jeollabuk 54896, Republic of Korea
| | - Hyun Jo Youn
- Department of Surgery, Research Institute of Clinical Medicine, Jeonbuk National University Hospital, Jeonbuk National University and Biomedical Research Institute, Jeonju, Jeollabuk 560‑182, Republic of Korea
| | - Young-Rae Lee
- Department of Oral Biochemistry, School of Dentistry, Wonkwang University, Iksan, Jeollabuk 570‑749, Republic of Korea
| |
Collapse
|
10
|
The Effects of Matriptase Inhibition on the Inflammatory and Redox Homeostasis of Chicken Hepatic Cell Culture Models. Biomedicines 2021; 9:biomedicines9050450. [PMID: 33919461 PMCID: PMC8143509 DOI: 10.3390/biomedicines9050450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 01/14/2023] Open
Abstract
The function of the transmembrane serine protease matriptase is well described in mammals, but it has not been elucidated in avian species yet. Hence, the aim of the present study was to assess the effects of the 3-amidinophenylalanine (3-AphA)-type matriptase inhibitors MI432 and MI460 on the inflammatory and oxidative state of chicken primary hepatocyte mono-cultures and hepatocyte-nonparenchymal cell co-cultures, the latter serving as a proper model of hepatic inflammation in birds. Cell cultures were exposed to MI432 and MI460 for 4 and 24 h at 10, 25, and 50 µM concentrations, and thereafter the cellular metabolic activity, extracellular interleukin (IL-)6, IL-8, H2O2 and malondialdehyde concentrations were monitored. Both inhibitors caused a transient moderate reduction in the metabolic activity following 4 h exposure, which was restored after 24 h, reflecting the fast hepatic adaptation potential to matriptase inhibitor administration. Furthermore, MI432 triggered an intense elevation in the cellular proinflammatory IL-6 and IL-8 production after both incubation times in all concentrations, which was not coupled to enhanced oxidative stress and lipid peroxidation based on unchanged H2O2 production, malondialdehyde levels and glutathione peroxidase activity. These data suggest that physiological matriptase activities might have a key function in retaining the metabolic and inflammatory homeostasis of the liver in chicken, without being a major modulator of the hepatocellular redox state.
Collapse
|
11
|
Jiang L, Jiang Y, Li L, Zheng K, Yu S, Li J, Yuan C, Huang M. A supramolecular nanocarrier for efficient cancer imaging and therapy by targeting at matriptase. J Control Release 2021; 334:153-163. [PMID: 33894302 DOI: 10.1016/j.jconrel.2021.04.019] [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: 12/03/2020] [Revised: 03/30/2021] [Accepted: 04/18/2021] [Indexed: 11/26/2022]
Abstract
Human serum albumin (HSA), a versatile protein carrier for endogenous and exogenous compounds, is a proven macromolecule to form nanoparticles for drug delivery. To render HSA carrier specificity toward tumors, we designed a recombinant HSA protein fused with Kunitz domain 1 (KD1) of hepatocyte growth factor activator inhibitor type 1, which targets to matriptase, a type II transmembrane serine protease overexpressed on tumor cell surface. The carrier was thus named matriptase targeting carrier (MTC). In this study, we showed that MTC displayed the same inhibitory potency as the KD1 againast matriptase, demonstrating the HSA fusion did not affect the KD1 targeting potency. For tumor optical imaging and ablation, MTC was prepared as nanoparticle drug carrier by a novel method via denaturation and refolding to incorporate photosensitizer, CPZ. This matriptase targeting nanoparticles, CPZ:MTC@NPs, showed high specificity and cytotoxicity for matriptase-overexpressing cancer cells in vitro. In tumor-bearing mice, CPZ:MTC@NPs demonstrated selective accumulation and high retention in matriptase-overexpressing tumor. Under illumination, the nanoparticles significantly reduced tumor volumes (79.6%) as compared to saline control. These findings showed that this supramolecular nanocarrier, a new type of tumor targeting self-assembly nanoparticle, had potential as a highly efficient tumor targeting drug carrier for imaging and therapy.
Collapse
Affiliation(s)
- Libin Jiang
- College of Chemistry, Fuzhou University, Fujian 350116, China
| | - Yunbin Jiang
- State Key Laboratory of Structure Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| | - Linlin Li
- College of Chemistry, Fuzhou University, Fujian 350116, China
| | - Ke Zheng
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, Shandong 266061, China
| | - Shujuan Yu
- College of Chemistry, Fuzhou University, Fujian 350116, China
| | - Jinyu Li
- College of Chemistry, Fuzhou University, Fujian 350116, China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fujian 350116, China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fujian 350116, China.
| |
Collapse
|
12
|
Ozawa S, Matsubayashi M, Nanaura H, Yanagita M, Mori K, Asanuma K, Kajiwara N, Hayashi K, Ohashi H, Kasahara M, Yokoi H, Kataoka H, Mori E, Nakagawa T. Proteolytic cleavage of Podocin by Matriptase exacerbates podocyte injury. J Biol Chem 2020; 295:16002-16012. [PMID: 32907879 DOI: 10.1074/jbc.ra120.013721] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/18/2020] [Indexed: 01/15/2023] Open
Abstract
Podocyte injury is a critical step toward the progression of renal disease and is often associated with a loss of slit diaphragm proteins, including Podocin. Although there is a possibility that the extracellular domain of these slit diaphragm proteins can be a target for a pathological proteolysis, the precise mechanism driving the phenomenon remains unknown. Here we show that Matriptase, a membrane-anchored protein, was activated at podocytes in CKD patients and mice, whereas Matriptase inhibitors slowed the progression of mouse kidney disease. The mechanism could be accounted for by an imbalance favoring Matriptase over its cognate inhibitor, hepatocyte growth factor activator inhibitor type 1 (HAI-1), because conditional depletion of HAI-1 in podocytes accelerated podocyte injury in mouse model. Matriptase was capable of cleaving Podocin, but such a reaction was blocked by either HAI-1 or dominant-negative Matriptase. Furthermore, the N terminus of Podocin, as a consequence of Matriptase cleavage of Podocin, translocated to nucleoli, suggesting that the N terminus of Podocin might be involved in the process of podocyte injury. Given these observations, we propose that the proteolytic cleavage of Podocin by Matriptase could potentially cause podocyte injury and that targeting Matriptase could be a novel therapeutic strategy for CKD patients.
Collapse
Affiliation(s)
- Shota Ozawa
- TMK Project at the Medical Innovation Center, Kyoto University, Kyoto, Japan; Research Unit/Innovative Medical Science, Mitsubishi Tanabe Pharma Corporation, Saitama, Japan
| | - Masaya Matsubayashi
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Hitoki Nanaura
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Motoko Yanagita
- TMK Project at the Medical Innovation Center, Kyoto University, Kyoto, Japan; Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Kiyoshi Mori
- TMK Project at the Medical Innovation Center, Kyoto University, Kyoto, Japan; Department of Molecular and Clinical Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Katsuhiko Asanuma
- TMK Project at the Medical Innovation Center, Kyoto University, Kyoto, Japan; Department of Nephrology, Chiba University, Chiba, Japan
| | | | - Kazuyuki Hayashi
- Department of Nephrology, Ikeda City Hospital, Ikeda, Osaka, Japan
| | - Hiroshi Ohashi
- Department of Pathology, Ikeda City Hospital, Ikeda, Osaka, Japan
| | - Masato Kasahara
- Institute for Clinical and Translational Science, Nara Medical University, Kashihara, Nara, Japan
| | - Hideki Yokoi
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Kataoka
- Department of Pathology, University of Miyazaki, Kihara, Miyazaki, Japan
| | - Eiichiro Mori
- Department of Future Basic Medicine, Nara Medical University, Kashihara, Nara, Japan.
| | - Takahiko Nakagawa
- TMK Project at the Medical Innovation Center, Kyoto University, Kyoto, Japan; Department of Future Basic Medicine, Nara Medical University, Kashihara, Nara, Japan.
| |
Collapse
|
13
|
Geiger M, Stubenrauch KG, Sam J, Richter WF, Jordan G, Eckmann J, Hage C, Nicolini V, Freimoser-Grundschober A, Ritter M, Lauer ME, Stahlberg H, Ringler P, Patel J, Sullivan E, Grau-Richards S, Endres S, Kobold S, Umaña P, Brünker P, Klein C. Protease-activation using anti-idiotypic masks enables tumor specificity of a folate receptor 1-T cell bispecific antibody. Nat Commun 2020; 11:3196. [PMID: 32581215 PMCID: PMC7314773 DOI: 10.1038/s41467-020-16838-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/29/2020] [Indexed: 01/06/2023] Open
Abstract
T-cell bispecific antibodies (TCBs) crosslink tumor and T-cells to induce tumor cell killing. While TCBs are very potent, on-target off-tumor toxicity remains a challenge when selecting targets. Here, we describe a protease-activated anti-folate receptor 1 TCB (Prot-FOLR1-TCB) equipped with an anti-idiotypic anti-CD3 mask connected to the anti-CD3 Fab through a tumor protease-cleavable linker. The potency of this Prot- FOLR1-TCB is recovered following protease-cleavage of the linker releasing the anti-idiotypic anti-CD3 scFv. In vivo, the Prot-FOLR1-TCB mediates antitumor efficacy comparable to the parental FOLR1-TCB whereas a noncleavable control Prot-FOLR1-TCB is inactive. In contrast, killing of bronchial epithelial and renal cortical cells with low FOLR1 expression is prevented compared to the parental FOLR1-TCB. The findings are confirmed for mesothelin as alternative tumor antigen. Thus, masking the anti-CD3 Fab fragment with an anti-idiotypic mask and cleavage of the mask by tumor-specific proteases can be applied to enhance specificity and safety of TCBs.
Collapse
Grants
- SK is supported by grants from the Wilhelm Sander Stiftung (grant number 2014.018.1 to SE and SK), the international doctoral program “i-Target: Immunotargeting of cancer” funded by the Elite Network of Bavaria (to SK and SE), the Melanoma Research Alliance (grant number N269626 to SE and 409510 to SK), the Marie-Sklodowska-Curie “Training Network for the Immunotherapy of Cancer (IMMUTRAIN)” funded by the H2020 program of the European Union (to SE and SK), by LMU Munich‘s Institutional Strategy LMUexcellent within the framework of the German Excellence Initiative (to SE and SK), the Bundesministerium für Bildung und Forschung (project Oncoattract to SE and SK).
- SK and SE are supported by grants from the Wilhelm Sander Stiftung (grant number 2014.018.1 to SE and SK), the international doctoral program “i-Target: Immunotargeting of cancer” funded by the Elite Network of Bavaria (to SK and SE), the Melanoma Research Alliance (grant number N269626 to SE and 409510 to SK), the Marie-Sklodowska-Curie “Training Network for the Immunotherapy of Cancer (IMMUTRAIN)” funded by the H2020 program of the European Union (to SE and SK), the Else Kröner- Fresenius-Stiftung (to SK), the German Cancer Aid (to SK), the Ernst-Jung-Stiftung (to SK), by LMU Munich‘s Institutional Strategy LMUexcellent within the framework of the German Excellence Initiative (to SE and SK), the Bundesministerium für Bildung und Forschung (project Oncoattract to SE and SK), the Deutsche Forschungsgemeinschaft, the José-Carreras Leukämie Stiftung, the Hector-Foundation (all to SK) and the European Research Council (ERC, grant 756017, ARMOR-T to SK).
Collapse
Affiliation(s)
- Martina Geiger
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952, Schlieren, Switzerland
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Lindwurmstraße 2a, Member of the German Center for Lung Research (DZL), 80337, Munich, Germany
| | - Kay-Gunnar Stubenrauch
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Nonnenwald 2, 82372, Penzberg, Germany
| | - Johannes Sam
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Wolfgang F Richter
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Gregor Jordan
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Nonnenwald 2, 82372, Penzberg, Germany
| | - Jan Eckmann
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Nonnenwald 2, 82372, Penzberg, Germany
| | - Carina Hage
- Roche Pharma Research & Early Development, Roche Innovation Center Munich, Nonnenwald 2, 82372, Penzberg, Germany
| | - Valeria Nicolini
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Anne Freimoser-Grundschober
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Mirko Ritter
- Roche Diagnostics, CPS Research and Development, Nonnenwald 2, 82372, Penzberg, Germany
| | - Matthias E Lauer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Henning Stahlberg
- Center for Cellular Imaging and Nano Analytics, Biozentrum, University of Basel, 4070, Basel, Switzerland
| | - Philippe Ringler
- Center for Cellular Imaging and Nano Analytics, Biozentrum, University of Basel, 4070, Basel, Switzerland
| | - Jigar Patel
- Roche Sequencing, NimbleGen, Madison, WI, 53719, USA
- Nimble Therapeutics Inc., 500S Rosa Rd, Madison, WI, 53719, USA
| | - Eric Sullivan
- Roche Sequencing, NimbleGen, Madison, WI, 53719, USA
- Nimble Therapeutics Inc., 500S Rosa Rd, Madison, WI, 53719, USA
| | - Sandra Grau-Richards
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Lindwurmstraße 2a, Member of the German Center for Lung Research (DZL), 80337, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Lindwurmstraße 2a, Member of the German Center for Lung Research (DZL), 80337, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, Munich, Germany
| | - Pablo Umaña
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Peter Brünker
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952, Schlieren, Switzerland
| | - Christian Klein
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich, Wagistrasse 10, 8952, Schlieren, Switzerland.
| |
Collapse
|
14
|
Abstract
Over the last two decades, a novel subgroup of serine proteases, the cell surface-anchored serine proteases, has emerged as an important component of the human degradome, and several members have garnered significant attention for their roles in cancer progression and metastasis. A large body of literature describes that cell surface-anchored serine proteases are deregulated in cancer and that they contribute to both tumor formation and metastasis through diverse molecular mechanisms. The loss of precise regulation of cell surface-anchored serine protease expression and/or catalytic activity may be contributing to the etiology of several cancer types. There is therefore a strong impetus to understand the events that lead to deregulation at the gene and protein levels, how these precipitate in various stages of tumorigenesis, and whether targeting of selected proteases can lead to novel cancer intervention strategies. This review summarizes current knowledge about cell surface-anchored serine proteases and their role in cancer based on biochemical characterization, cell culture-based studies, expression studies, and in vivo experiments. Efforts to develop inhibitors to target cell surface-anchored serine proteases in cancer therapy will also be summarized.
Collapse
|
15
|
Hu P, Shang L, Chen J, Chen X, Chen C, Hong W, Huang M, Xu P, Chen Z. A nanometer-sized protease inhibitor for precise cancer diagnosis and treatment. J Mater Chem B 2020; 8:504-514. [PMID: 31840729 DOI: 10.1039/c9tb02081k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inhibition of pro-cancer proteases is a potent anticancer strategy. However, protease inhibitors are mostly developed in the forms of small molecules or peptides, which normally suffer from insufficient metabolic stability. The fast clearance significantly impairs the antitumor effects of these inhibitors. In this study, we report a nanometer-sized inhibitor of a pro-cancer protease, suppressor of tumorigenicity 14 (st14), which has been reported as a potent prognostic marker for multiple cancers. This st14 inhibitor was fabricated by conjugating a recombinant st14 inhibitor (KD1) with carbon quantum dots (CQDs). CQD-KD1 not only demonstrated high potency of inhibiting st14 activity in biochemical experiments, but also remarkably suppressed the invasion of breast cancer cells. In contrast to the original recombinant KD1, CQD-KD1 demonstrated a prolonged retention time in plasma and at the tumor site because of the reduced renal clearance. Consistently, CQD-KD1 demonstrated enhanced efficacies of suppressing tumor growth and cancer metastases in vivo. In addition, CQD-KD1 precisely imaged tumor tissues in cancer-grafted mice by specifically targeting the over-expressed st14 on the tumor cell surface, which indicates CQD-KD1 as a potent probe for the fluorescence guided surgery of tumor resection. In conclusion, this study demonstrates that CQD-KD1 is a highly potent diagnostic and therapeutic agent for cancer treatments.
Collapse
Affiliation(s)
- Ping Hu
- State Key Laboratory of Structural Chemistry, and CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Barna RF, Pomothy JM, Paréj Z, Pásztiné Gere E. Investigation of sphingosin-1-phosphate-triggered matriptase activation using a rat primary hepatocyte model. Acta Vet Hung 2019; 67:578-587. [PMID: 31842605 DOI: 10.1556/004.2019.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Sphingosine-1-phosphate (S1P) has been reported as a matriptase activator. The aim of this study was to reveal if S1P can influence hepcidin production. Furthermore, we investigated how S1P can affect the viability and the redox status of primary hepatocytes. Rat primary hepatocytes were cultivated for 72 h and were treated with 50, 200, 1000 ng/ml S1P. Cell-free supernatants were collected every 24 h. Cell viability was tested by a colorimetric method using tetrazolium compound (MTS). The hepcidin levels in the cell-free supernatants were examined with hepcidin sandwich ELISA to determine the effect of S1P on the hepcidin-modulating ability of matriptase. In order to estimate the extent of S1P-generated oxidative stress, extracellular H2O2 measurements were performed by the use of fluorescent dye. Based on the findings, S1P treatment did not cause cell death for 72 h at concentrations up to 1000 ng/ml. S1P did not influence the extracellular H2O2 production for 72 h. The hepcidin levels were significantly suppressed in hepatocytes exposed to S1P treatment. Further studies would be needed to explore the exact mechanism of action of S1P.
Collapse
Affiliation(s)
- Réka Fanni Barna
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, István u. 2, H-1078 Budapest, Hungary
| | - Judit Mercédesz Pomothy
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, István u. 2, H-1078 Budapest, Hungary
| | - Zsuzsanna Paréj
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, István u. 2, H-1078 Budapest, Hungary
| | - Erzsébet Pásztiné Gere
- Department of Pharmacology and Toxicology, University of Veterinary Medicine Budapest, István u. 2, H-1078 Budapest, Hungary
| |
Collapse
|
17
|
Tracking genome-editing and associated molecular perturbations by SWATH mass spectrometry. Sci Rep 2019; 9:15240. [PMID: 31645615 PMCID: PMC6811567 DOI: 10.1038/s41598-019-51612-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/02/2019] [Indexed: 12/19/2022] Open
Abstract
Advances in gene editing now allow reverse genetics to be applied to a broad range of biological systems. Ultimately, any modification to coding sequences requires confirmation at the protein level, although immunoblotting is often hampered by antibody quality or availability especially in non-model species. Sequential Window Acquisition of All Theoretical Spectra (SWATH), a mass spectrometry (MS) technology with exceptional quantitative reproducibility and accuracy, offers an ideal alternative for protein-based confirmation. Here, using genome edits in mouse, zebrafish and Bicyclus anynana butterflies produced using either homologous recombination or targeted nucleases, we demonstrate absence of the targeted proteins using SWATH, thus confirming successful editing. We show that SWATH is a robust antibody-independent alternative for monitoring gene editing at the protein level and broadly applicable across diverse organisms and targeted genome manipulation techniques. Moreover, SWATH concomitantly defines the global proteome response in the edited organism, which may provide pertinent biological insights.
Collapse
|
18
|
Rather GM, Lin SY, Lin H, Szekely Z, Bertino JR. A Novel Antibody-Toxin Conjugate to Treat Mantle Cell Lymphoma. Front Oncol 2019; 9:258. [PMID: 31024856 PMCID: PMC6467949 DOI: 10.3389/fonc.2019.00258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 03/21/2019] [Indexed: 11/18/2022] Open
Abstract
Matriptase is a transmembrane serine protease, synthesized as an inactive single-chain zymogen on the endoplasmic reticulum and transported to the plasma membrane. Matriptase is activated in different epithelial and some B-cell malignancies and changes its conformation and activity is inhibited mainly by its endogenous inhibitor HAI-1. Activated matriptase plays a key role in tumor initiation as well as tumor progression, including invasiveness, and metastasis. To target the anti-mitotic toxin (monomethyl auristatin-E) to activated matriptase, a novel antibody to activated matriptase was conjugated with this toxin via a valine-citrulline-PABA linker. In a previous study, this antibody-toxin conjugate was found to be effective against triple negative breast cancer cell lines and xenografts, alone, or in combination with cisplatin (1). In this study, we examined the anti-tumor effect of the antibody toxin conjugate (ADC) against activated matriptase positive mantle cell lymphoma cell lines (JeKo-1, Maver, Mino, and Z138). This ADC was cytotoxic to these cell lines with IC50s between 5 and 14 μg/mL. The ADC also showed a dose dependent anti-tumor effect on the JeKo-1 xenograft in mice without toxicity.
Collapse
Affiliation(s)
- Gulam M Rather
- Departments of Pharmacology and Medicine, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Siang-Yo Lin
- Departments of Pharmacology and Medicine, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Hongxia Lin
- Departments of Pharmacology and Medicine, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Zoltan Szekely
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Joseph R Bertino
- Departments of Pharmacology and Medicine, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
19
|
Recent progress on inhibitors of the type II transmembrane serine proteases, hepsin, matriptase and matriptase-2. Future Med Chem 2019; 11:743-769. [DOI: 10.4155/fmc-2018-0446] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Members of the type II transmembrane serine proteases (TTSP) family play a vital role in cell growth and development but many are also implicated in disease. Two of the well-studied TTSPs, matriptase and hepsin proteolytically process multiple protein substrates such as the inactive single-chain zymogens pro-HGF and pro-macrophage stimulating protein into the active heterodimeric forms, HGF and macrophage stimulating protein. These two proteases also have many other substrates which are associated with cancer and tumor progression. Another related TTSP, matriptase-2 is expressed in the liver and functions by regulating iron homoeostasis through the cleavage of hemojuvelin and thus is implicated in iron overload diseases. In the present review, we will discuss inhibitor design strategy and Structure activity relationships of TTSP inhibitors, which have been reported in the literature.
Collapse
|
20
|
Liu YL, Chou CK, Kim M, Vasisht R, Kuo YA, Ang P, Liu C, Perillo EP, Chen YA, Blocher K, Horng H, Chen YI, Nguyen DT, Yankeelov TE, Hung MC, Dunn AK, Yeh HC. Assessing metastatic potential of breast cancer cells based on EGFR dynamics. Sci Rep 2019; 9:3395. [PMID: 30833579 PMCID: PMC6399327 DOI: 10.1038/s41598-018-37625-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 11/22/2018] [Indexed: 01/05/2023] Open
Abstract
Derailed transmembrane receptor trafficking could be a hallmark of tumorigenesis and increased tumor invasiveness, but receptor dynamics have not been used to differentiate metastatic cancer cells from less invasive ones. Using single-particle tracking techniques, we developed a phenotyping asssay named Transmembrane Receptor Dynamics (TReD), studied the dynamics of epidermal growth factor receptor (EGFR) in seven breast epithelial cell lines and developed a phenotyping assay named Transmembrane Receptor Dynamics (TReD). Here we show a clear evidence that increased EGFR diffusivity and enlarged EGFR confinement size in the plasma membrane (PM) are correlated with the enhanced metastatic potential in these cell lines. By comparing the TReD results with the gene expression profiles, we found a clear negative correlation between the EGFR diffusivities and the breast cancer luminal differentiation scores (r = -0.75). Upon the induction of epithelial-mesenchymal transition (EMT), EGFR diffusivity significantly increased for the non-tumorigenic MCF10A (99%) and the non-invasive MCF7 (56%) cells, but not for the highly metastatic MDA-MB-231 cell. We believe that the reorganization of actin filaments during EMT modified the PM structures, causing the receptor dynamics to change. TReD can thus serve as a new biophysical marker to probe the metastatic potential of cancer cells and even to monitor the transition of metastasis.
Collapse
Affiliation(s)
- Yen-Liang Liu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Chao-Kai Chou
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mirae Kim
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Rohan Vasisht
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Yu-An Kuo
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Phyllis Ang
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Cong Liu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Evan P Perillo
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Yu-An Chen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Katherine Blocher
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hannah Horng
- Department of Bioengineering, The University of Maryland, College Park, MD, USA
| | - Yuan-I Chen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Duc Trung Nguyen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Computational Engineering and Sciences, The University of Texas, Austin, TX, USA
- Department of Diagnostic Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Molecular Medicine and Graduate Institute of Cancer Biology, China Medical University, Taichung, Taiwan
| | - Andrew K Dunn
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Hsin-Chih Yeh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Texas Materials Institute, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
21
|
Chiu YL, Wu YY, Barndt RB, Yeo YH, Lin YW, Sytwo HP, Liu HC, Xu Y, Jia B, Wang JK, Johnson MD, Lin CY. Aberrant regulation favours matriptase proteolysis in neoplastic B-cells that co-express HAI-2. J Enzyme Inhib Med Chem 2019; 34:692-702. [PMID: 30777474 PMCID: PMC6383611 DOI: 10.1080/14756366.2019.1577831] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Matriptase is ectopically expressed in neoplastic B-cells, in which matriptase activity is enhanced by negligible expression of its endogenous inhibitor, hepatocyte growth factor activator inhibitor (HAI)-1. HAI-1, however, is also involved in matriptase synthesis and intracellular trafficking. The lack of HAI-1 indicates that other related inhibitor, such as HAI-2, might be expressed. Here, we show that HAI-2 is commonly co-expressed in matriptase-expressing neoplastic B-cells. The level of active matriptase shed after induction of matriptase zymogen activation in 7 different neoplastic B-cells was next determined and characterised. Our data reveal that active matriptase can only be generated and shed by those cells able to activate matriptase and in a rough correlation with the levels of matriptase protein. While HAI-2 can potently inhibit matriptase, the levels of active matriptase are not proportionally suppressed in those cells with high HAI-2. Our survey suggests that matriptase proteolysis might aberrantly remain high in neoplastic B-cells regardless of the levels of HAI-2.
Collapse
Affiliation(s)
- Yi-Lin Chiu
- a Lombardi Comprehensive Cancer Center, Department of Oncology , Georgetown University , Washington , DC, USA.,b Department of Biochemistry , National Defense Medical Center , Taipei , Taiwan
| | - Yi-Ying Wu
- c Division of Hematology/Oncology, Department of Internal Medicine , Tri-Service General Hospital, National Defense Medical Center , Taipei , Taiwan
| | - Robert B Barndt
- a Lombardi Comprehensive Cancer Center, Department of Oncology , Georgetown University , Washington , DC, USA
| | - Yee Hui Yeo
- a Lombardi Comprehensive Cancer Center, Department of Oncology , Georgetown University , Washington , DC, USA
| | - Yu-Wen Lin
- a Lombardi Comprehensive Cancer Center, Department of Oncology , Georgetown University , Washington , DC, USA.,b Department of Biochemistry , National Defense Medical Center , Taipei , Taiwan
| | - Hou-Ping Sytwo
- d School of Medicine , National Defense Medical Center , Taipei , Taiwan
| | - Huan-Cheng Liu
- a Lombardi Comprehensive Cancer Center, Department of Oncology , Georgetown University , Washington , DC, USA.,e Langley High School , McLean , VA, USA
| | - Yuan Xu
- a Lombardi Comprehensive Cancer Center, Department of Oncology , Georgetown University , Washington , DC, USA
| | - Bailing Jia
- a Lombardi Comprehensive Cancer Center, Department of Oncology , Georgetown University , Washington , DC, USA.,f Department of Gastroenterology , Henan Provincial People's Hospital , Zhengzhou , China
| | - Jehng-Kang Wang
- b Department of Biochemistry , National Defense Medical Center , Taipei , Taiwan
| | - Michael D Johnson
- a Lombardi Comprehensive Cancer Center, Department of Oncology , Georgetown University , Washington , DC, USA
| | - Chen-Yong Lin
- a Lombardi Comprehensive Cancer Center, Department of Oncology , Georgetown University , Washington , DC, USA
| |
Collapse
|
22
|
Fang JD, Tung HH, Lee SL. Mitochondrial localization of St14-encoding transmembrane serine protease is involved in neural stem/progenitor cell bioenergetics through binding to F 0F 1-ATP synthase complex. FASEB J 2018; 33:4327-4340. [PMID: 30566397 DOI: 10.1096/fj.201801307r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Knockdown of the suppression of tumorigenicity 14-encoding type II transmembrane serine protease matriptase (MTP) in neural stem/progenitor (NS/P) cells impairs cell mobility, response to chemo-attractants, and neurovascular niche interaction. In the present study, we showed by Western blot that a portion of MTP can be detected in the mitochondrial fraction of mouse NS/P cells by immunostaining that it is co-stained with the mitochondrial dye MitoTracker (Thermo Fisher Scientific, Waltham, MA, USA) inside the cells. Co-immunoprecipitation showed that MTP is bound to the β subunit of mitochondrial F0F1-ATP synthase complex (ATP-β). Cyto-immunofluorescence staining and an in situ proximity ligation assay further confirmed a physical interaction between MTP and ATP-β. This interaction relied on the presence of both Cls/Clr urchin embryonic growth factor, bone morphogenic protein 1 and low-density lipoprotein receptor motifs of MTP. We found that NS/P cell mitochondrial membrane potential is impaired by MTP knockdown, and ATP synthesis and oxygen consumption rate are significantly reduced in MTP-knockdown NS/P cells. Among the oxidative phosphorylation functions, the greatest effect of MTP knockdown is the reduction by over 50% in the mitochondrial energy reserve capacity. This made MTP-knockdown NS/P cells unable to overcome hydrogen peroxide stress, which leads to cessation of cell growth. This work identifies 2 previously unknown functions for MTP: first as a binding protein in the mitochondrial F1F0-ATP synthase complex and second as a regulatory mechanism of mitochondrial bioenergetics. Mitochondrial MTP may serve a protective function for NS/P cells in response to stress.-Fang, J.-D., Tung, H.-H., Lee, S.-L. Mitochondrial localization of St14-encoding transmembrane serine protease is involved in neural stem/progenitor cell bioenergetics through binding to F0F1-ATP synthase complex.
Collapse
Affiliation(s)
- Jung-Da Fang
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan Town, Taiwan, Republic of China
| | - Hsiu-Hui Tung
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan Town, Taiwan, Republic of China
| | - Sheau-Ling Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan Town, Taiwan, Republic of China
| |
Collapse
|
23
|
Kreitman M, Noronha A, Yarden Y. Irreversible modifications of receptor tyrosine kinases. FEBS Lett 2018; 592:2199-2212. [PMID: 29790151 DOI: 10.1002/1873-3468.13095] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/12/2018] [Accepted: 05/08/2018] [Indexed: 01/18/2023]
Abstract
Each group of the 56 receptor tyrosine kinases (RTK) binds with one or more soluble growth factors and coordinates a vast array of cellular functions. These outcomes are tightly regulated by inducible post-translational events, such as tyrosine phosphorylation, ubiquitination, ectodomain shedding, and regulated intramembrane proteolysis. Because of the delicate balance required for appropriate RTK function, cells may become pathogenic upon dysregulation of RTKs themselves or their post-translational covalent modifications. For example, reduced ectodomain shedding and decreased ubiquitination of the cytoplasmic region, both of which enhance growth factor signals, characterize malignant cells. Whereas receptor phosphorylation and ubiquitination are reversible, proteolytic cleavage events are irreversible, and either modification might alter the subcellular localization of RTKs. Herein, we focus on ectodomain shedding by metalloproteinases (including ADAM family proteases), cleavage within the membrane or cytoplasmic regions of RTKs (by gamma-secretases and caspases, respectively), and complete receptor proteolysis in lysosomes and proteasomes. Roles of irreversible modifications in RTK signaling, pathogenesis, and pharmacology are highlighted.
Collapse
Affiliation(s)
- Matthew Kreitman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Ashish Noronha
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
24
|
Activated matriptase as a target to treat breast cancer with a drug conjugate. Oncotarget 2018; 9:25983-25992. [PMID: 29899836 PMCID: PMC5995259 DOI: 10.18632/oncotarget.25414] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023] Open
Abstract
The antitumor effects of a novel antibody drug conjugate (ADC) was tested against human solid tumor cell lines and against human triple negative breast cancer (TNBC) xenografts in immunosuppressed mice. The ADC targeting activated matriptase of tumor cells was synthesized by using the potent anti-tubulin toxin, monomethyl auristatin-E linked to the activated matriptase-specific monoclonal antibody (M69) via a lysosomal protease-cleavable dipeptide linker. This ADC was found to be cytotoxic against multiple activated matriptase-positive epithelial carcinoma cell lines in vitro and markedly inhibited growth of triple negative breast cancer xenografts and a primary human TNBC (PDX) in vivo. Overexpression of activated matriptase may be a biomarker for response to this ADC. The ADC had potent anti-tumor activity, while the unconjugated M69 antibody was ineffective in a mouse model study using MDA-MB-231 xenografts in mice. Treatment of a human TNBC (MDA-MB-231) showed potent anti-tumor effects in combination with cisplatin in mice. This ADC alone or in combination with cisplatin has the potential to improve the treatment outcomes of patients with TNBC as well as other tumors overexpressing activated matriptase.
Collapse
|
25
|
Sun P, Xue L, Song Y, Mao X, Chen L, Dong B, Braicu EL, Sehouli J. Regulation of matriptase and HAI-1 system, a novel therapeutic target in human endometrial cancer cells. Oncotarget 2018; 9:12682-12694. [PMID: 29560101 PMCID: PMC5849165 DOI: 10.18632/oncotarget.23913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 11/01/2017] [Indexed: 12/25/2022] Open
Abstract
The effects of specific and non-specific regulation of matriptase on endometrial cancer cells in vitro were investigated. Messenger ribonucleic acid (mRNA) and protein expression of matriptase and hepatocyte growth factor activator inhibitor-1 (HAI-1) in RL-952, HEC-1A, and HEC-1B endometrial cancer cells were detected by real-time quantitative PCR (RT-qPCR) and western blot. The cells were infected with lentivirus-mediated small-interfering RNA (siRNA) targeted on matriptase (MA-siRNA) or treated with different cisplatin (DDP) concentrations. After treatment, invasion, migration, and cellular apoptosis were analyzed. Matriptase mRNA and protein expression significantly decreased to 80% after infection with MA-siRNA (P < 0.01), and scratch and trans-well chamber assays showed significant inhibition of invasiveness and metastasis. Upon incubation with cisplatin at concentrations higher than the therapeutic dose for 24 h, the expressions of matriptase and HAI-1 significantly decreased (P < 0.001). Moreover, the invasiveness, metastasis, and survival rate of HEC-1A and RL-952 endometrial cancer cells were significantly decreased (P < 0.001) due to the down-regulation of matriptase and HAI-1 upon increasing cisplatin concentration. However, a slight increase in matriptase and HAI-1 expression was observed in cells treated with low cisplatin concentration (P = 0.01). Moreover, matriptase expression was associated with metastasis and invasiveness. Down-regulation of matriptase by specific Ma-SiRNA or non-specific cisplatin in matriptase/HAI-1-positive endometrial cancer cells showed promising therapeutic features.
Collapse
Affiliation(s)
- Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children Hospital, Affiliate Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, P.R. of China.,Department of Gynecology, Fujian Provincial Maternity and Children Hospital, Affiliate Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, P.R. of China
| | - Lifang Xue
- Department of Gynecology, Fujian Provincial Maternity and Children Hospital, Affiliate Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, P.R. of China
| | - Yiyi Song
- Department of Gynecology, Fujian Provincial Maternity and Children Hospital, Affiliate Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, P.R. of China
| | - Xiaodan Mao
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children Hospital, Affiliate Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, P.R. of China
| | - Lili Chen
- Department of Gynecology, Fujian Provincial Maternity and Children Hospital, Affiliate Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, P.R. of China
| | - Binhua Dong
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children Hospital, Affiliate Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, P.R. of China
| | - Elena Loana Braicu
- Department of Gynecologic Oncology and Gynecology, Charité, Campus Virchow-Klinikum, European Competence Center for Ovarian Cancer University of Berlin, 13353 Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecologic Oncology and Gynecology, Charité, Campus Virchow-Klinikum, European Competence Center for Ovarian Cancer University of Berlin, 13353 Berlin, Germany
| |
Collapse
|
26
|
Kim S, Yang JW, Kim C, Kim MG. Impact of suppression of tumorigenicity 14 (ST14)/serine protease 14 (Prss14) expression analysis on the prognosis and management of estrogen receptor negative breast cancer. Oncotarget 2017; 7:34643-63. [PMID: 27167193 PMCID: PMC5085182 DOI: 10.18632/oncotarget.9155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 04/16/2016] [Indexed: 01/06/2023] Open
Abstract
To elucidate the role of a type II transmembrane serine protease, ST14/Prss14, during breast cancer progression, we utilized publically accessible databases including TCGA, GEO, NCI-60, and CCLE. Survival of breast cancer patients with high ST14/Prss14 expression is significantly poor in estrogen receptor (ER) negative populations regardless of the ratios of ST14/Prss14 to its inhibitors, SPINT1 or SPINT2. In a clustering of 1085 selected EMT signature genes, ST14/Prss14 is located in the same cluster with CDH3, and closer to post-EMT markers, CDH2, VIM, and FN1 than to the pre-EMT marker, CDH1. Coexpression analyses of known ST14/Prss14 substrates and transcription factors revealed context dependent action. In cell lines, paradoxically, ST14/Prss14 expression is higher in the ER positive group and located closer to CDH1 in clustering. This apparent contradiction is not likely due to ST14/Prss14 expression in a cancer microenvironment, nor due to negative regulation by ER. Genes consistently coexpressed with ST14/Prss14 include transcription factors, ELF5, GRHL1, VGLL1, suggesting currently unknown mechanisms for regulation. Here, we report that ST14/Prss14 is an emerging therapeutic target for breast cancer where HER2 is not applicable. In addition we suggest that careful conclusions should be drawn not exclusively from the cell line studies for target development.
Collapse
Affiliation(s)
- Sauryang Kim
- Inha University, Department of Biological Sciences, Incheon, Republic of Korea
| | - Jae Woong Yang
- Inha University, Department of Biological Sciences, Incheon, Republic of Korea
| | - Chungho Kim
- Department of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Moon Gyo Kim
- Inha University, Department of Biological Sciences, Incheon, Republic of Korea.,Convergent Research Institute for Metabolism and Immunoregulation, Incheon, Republic of Korea
| |
Collapse
|
27
|
Wu SR, Teng CH, Tu YT, Ko CJ, Cheng TS, Lan SW, Lin HY, Lin HH, Tu HF, Hsiao PW, Huang HP, Chen CH, Lee MS. The Kunitz Domain I of Hepatocyte Growth Factor Activator Inhibitor-2 Inhibits Matriptase Activity and Invasive Ability of Human Prostate Cancer Cells. Sci Rep 2017; 7:15101. [PMID: 29118397 PMCID: PMC5678078 DOI: 10.1038/s41598-017-15415-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 10/26/2017] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of pericellular proteolysis is often required for tumor invasion and cancer progression. It has been shown that down-regulation of hepatocyte growth factor activator inhibitor-2 (HAI-2) results in activation of matriptase (a membrane-anchored serine protease), human prostate cancer cell motility and tumor growth. In this study, we further characterized if HAI-2 was a cognate inhibitor for matriptase and identified which Kunitz domain of HAI-2 was required for inhibiting matriptase and human prostate cancer cell motility. Our results show that HAI-2 overexpression suppressed matriptase-induced prostate cancer cell motility. We demonstrate that HAI-2 interacts with matriptase on cell surface and inhibits matriptase proteolytic activity. Moreover, cellular HAI-2 harnesses its Kunitz domain 1 (KD1) to inhibit matriptase activation and prostate cancer cell motility although recombinant KD1 and KD2 of HAI-2 both show an inhibitory activity and interaction with matriptase protease domain. The results together indicate that HAI-2 is a cognate inhibitor of matriptase, and KD1 of HAI-2 plays a major role in the inhibition of cellular matritptase activation as well as human prostate cancer invasion.
Collapse
Affiliation(s)
- Shang-Ru Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Hsin Teng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Ting Tu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Jung Ko
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tai-Shan Cheng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shao-Wei Lan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Ying Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Hsien Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Fang Tu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chung-Hsin Chen
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Shyue Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
28
|
Lanchec E, Désilets A, Béliveau F, Flamier A, Mahmoud S, Bernier G, Gris D, Leduc R, Lavoie C. The type II transmembrane serine protease matriptase cleaves the amyloid precursor protein and reduces its processing to β-amyloid peptide. J Biol Chem 2017; 292:20669-20682. [PMID: 29054928 DOI: 10.1074/jbc.m117.792911] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/16/2017] [Indexed: 11/06/2022] Open
Abstract
Recent studies have reported that many proteases, besides the canonical α-, β-, and γ-secretases, cleave the amyloid precursor protein (APP) and modulate β-amyloid (Aβ) peptide production. Moreover, specific APP isoforms contain Kunitz protease-inhibitory domains, which regulate the proteolytic activity of serine proteases. This prompted us to investigate the role of matriptase, a member of the type II transmembrane serine protease family, in APP processing. Using quantitative RT-PCR, we detected matriptase mRNA in several regions of the human brain with an enrichment in neurons. RNA sequencing data of human dorsolateral prefrontal cortex revealed relatively high levels of matriptase RNA in young individuals, whereas lower levels were detected in older individuals. We further demonstrate that matriptase and APP directly interact with each other and that matriptase cleaves APP at a specific arginine residue (Arg-102) both in vitro and in cells. Site-directed (Arg-to-Ala) mutagenesis of this cleavage site abolished matriptase-mediated APP processing. Moreover, we observed that a soluble, shed matriptase form cleaves endogenous APP in SH-SY5Y cells and that this cleavage significantly reduces APP processing to Aβ40. In summary, this study identifies matriptase as an APP-cleaving enzyme, an activity that could have important consequences for the abundance of Aβ and in Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Erwan Lanchec
- From the Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec J1H5N4, Canada
| | - Antoine Désilets
- From the Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec J1H5N4, Canada
| | - François Béliveau
- From the Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec J1H5N4, Canada
| | - Anthony Flamier
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont, 5415 Boulevard de l'Assomption, Montréal, Quebec H1T 2M4, Canada
| | - Shaimaa Mahmoud
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec J1H5N4, Canada, and
| | - Gilbert Bernier
- Stem Cell and Developmental Biology Laboratory, Hôpital Maisonneuve-Rosemont, 5415 Boulevard de l'Assomption, Montréal, Quebec H1T 2M4, Canada.,Department of Neurosciences, Université de Montréal, Montréal, Quebec H3C 3J7, Canada
| | - Denis Gris
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec J1H5N4, Canada, and
| | - Richard Leduc
- From the Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec J1H5N4, Canada,
| | - Christine Lavoie
- From the Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec J1H5N4, Canada,
| |
Collapse
|
29
|
Ishikawa T, Kimura Y, Hirano H, Higashi S. Matrix metalloproteinase-7 induces homotypic tumor cell aggregation via proteolytic cleavage of the membrane-bound Kunitz-type inhibitor HAI-1. J Biol Chem 2017; 292:20769-20784. [PMID: 29046355 DOI: 10.1074/jbc.m117.796789] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 10/11/2017] [Indexed: 12/22/2022] Open
Abstract
Matrix metalloproteinase-7 (MMP-7) plays important roles in tumor progression and metastasis. Our previous studies have demonstrated that MMP-7 binds to colon cancer cells via cell surface-bound cholesterol sulfate and induces significant cell aggregation by cleaving cell-surface protein(s). These aggregated cells exhibit a dramatically enhanced metastatic potential. However, the molecular mechanism inducing this cell-cell adhesion through the proteolytic action of MMP-7 remained to be clarified. Here, we explored MMP-7 substrates on the cell surface; the proteins on the cell surface were first biotinylated, and a labeled protein fragment specifically released from the cells after MMP-7 treatment was analyzed using LC-MS/MS. We found that hepatocyte growth factor activator inhibitor type 1 (HAI-1), a membrane-bound Kunitz-type serine protease inhibitor, is an MMP-7 substrate. We also found that the cell-bound MMP-7 cleaves HAI-1 mainly between Gly451 and Leu452 and thereby releases the extracellular region as soluble HAI-1 (sHAI-1). We further demonstrated that this sHAI-1 can induce cancer cell aggregation and determined that the HAI-1 region corresponding to amino acids 141-249, which does not include the serine protease inhibitor domain, has the cell aggregation-inducing activity. Interestingly, a cell-surface cholesterol sulfate-independent proteolytic action of MMP-7 is critical for the sHAI-1-mediated induction of cell aggregation, whereas cholesterol sulfate is needed for the MMP-7-catalyzed generation of sHAI-1. Considering that MMP-7-induced cancer cell aggregation is an important mechanism in cancer metastasis, we propose that sHAI-1 is an essential component of MMP-7-induced stimulation of cancer metastasis and may therefore represent a suitable target for antimetastatic therapeutic strategies.
Collapse
Affiliation(s)
- Tomohiro Ishikawa
- From the Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027 and
| | - Yayoi Kimura
- the Advanced Medical Research Center, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Hisashi Hirano
- the Advanced Medical Research Center, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Shouichi Higashi
- From the Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027 and
| |
Collapse
|
30
|
Murray AS, Varela FA, Hyland TE, Schoenbeck AJ, White JM, Tanabe LM, Todi SV, List K. Phosphorylation of the type II transmembrane serine protease, TMPRSS13, in hepatocyte growth factor activator inhibitor-1 and -2-mediated cell-surface localization. J Biol Chem 2017; 292:14867-14884. [PMID: 28710277 DOI: 10.1074/jbc.m117.775999] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 06/12/2017] [Indexed: 12/22/2022] Open
Abstract
TMPRSS13 is a member of the type II transmembrane serine protease (TTSP) family. Although various TTSPs have been characterized in detail biochemically and functionally, the basic properties of TMPRSS13 remain unclear. Here, we investigate the activation, inhibition, post-translational modification, and localization of TMPRSS13. We show that TMPRSS13 is a glycosylated, active protease and that its own proteolytic activity mediates zymogen cleavage. Full-length, active TMPRSS13 exhibits impaired cell-surface expression in the absence of the cognate Kunitz-type serine protease inhibitors, hepatocyte growth factor activator inhibitor (HAI)-1 or HAI-2. Concomitant presence of TMPRSS13 with either HAI-1 or -2 mediates phosphorylation of residues in the intracellular domain of the protease, and it coincides with efficient transport of the protease to the cell surface and its subsequent shedding. Cell-surface labeling experiments indicate that the dominant form of TMPRSS13 on the cell surface is phosphorylated, whereas intracellular TMPRSS13 is predominantly non-phosphorylated. These data provide novel insight into the cellular properties of TMPRSS13 and highlight phosphorylation of TMPRSS13 as a novel post-translational modification of this TTSP family member and potentially other members of this family of proteases.
Collapse
Affiliation(s)
- Andrew S Murray
- From the Departments of Pharmacology.,Oncology, and.,Cancer Biology Graduate Program, and.,the Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
| | - Fausto A Varela
- From the Departments of Pharmacology.,Pharmacology Graduate Program, and
| | | | | | - Jordan M White
- From the Departments of Pharmacology.,Oncology, and.,Cancer Biology Graduate Program, and.,the Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
| | | | | | - Karin List
- From the Departments of Pharmacology, .,Oncology, and.,the Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201
| |
Collapse
|
31
|
Chen LM, Chai KX. Proteolytic cleavages in the extracellular domain of receptor tyrosine kinases by membrane-associated serine proteases. Oncotarget 2017; 8:56490-56505. [PMID: 28915606 PMCID: PMC5593577 DOI: 10.18632/oncotarget.17009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 03/21/2017] [Indexed: 12/21/2022] Open
Abstract
The epithelial extracellular membrane-associated serine proteases matriptase, hepsin, and prostasin are proteolytic modifying enzymes of the extracellular domain (ECD) of the epidermal growth factor receptor (EGFR). Matriptase also cleaves the ECD of the vascular endothelial growth factor receptor 2 (VEGFR2) and the angiopoietin receptor Tie2. In this study we tested the hypothesis that these serine proteases may cleave the ECD of additional receptor tyrosine kinases (RTKs). We co-expressed the proteases in an epithelial cell line with Her2, Her3, Her4, insulin receptor (INSR), insulin-like growth factor I receptor (IGF-1R), the platelet-derived growth factor receptors (PDGFRs) α and β, or nerve growth factor receptor A (TrkA). Western blot analysis was performed to detect the carboxyl-terminal fragments (CTFs) of the RTKs. Matriptase and hepsin were found to cleave the ECD of all RTKs tested, while TMPRSS6/matriptase-2 cleaves the ECD of Her4, INSR, and PDGFR α and β. Prostasin was able to cleave the ECD of Her3 and PDGFRα. Matriptase cleaves phosphorylated Her2 at Arg558 and Arg599 and the Arg599 cleavage produces a CTF not recognized by the monoclonal antibody trastuzumab/Herceptin. Her2 cleavages by matriptase can be inhibited by the hepatocyte growth factor activator inhibitor 1 (HAI-1) in the MDA-MB-231 human breast cancer cells. Matriptase silencing in the Her2, matriptase, and HAI-1 triple-positive SKBR3 human breast cancer cells enhanced Her2 protein down-regulation induced by a sustained exposure to phorbol 12-myristate 13-acetate (PMA), which down-regulated matriptase protein. The novel Her2 cleavage and expression regulation mechanisms mediated by matriptase may have potential impacts in Her2-targeting therapies.
Collapse
Affiliation(s)
- Li-Mei Chen
- Burnett School of Biomedical Sciences, Division of Cancer Research, University of Central Florida College of Medicine, Orlando, FL 32816-2364, USA
| | - Karl X Chai
- Burnett School of Biomedical Sciences, Division of Cancer Research, University of Central Florida College of Medicine, Orlando, FL 32816-2364, USA
| |
Collapse
|
32
|
Tanabe LM, List K. The role of type II transmembrane serine protease-mediated signaling in cancer. FEBS J 2016; 284:1421-1436. [PMID: 27870503 DOI: 10.1111/febs.13971] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/29/2016] [Accepted: 11/18/2016] [Indexed: 12/31/2022]
Abstract
Pericellular proteases have long been implicated in carcinogenesis. Previous research focused on these proteins, primarily as extracellular matrix (ECM) protein-degrading enzymes which allowed cancer cells to breach the basement membrane and invade surrounding tissue. However, recently, there has been a shift in the view of cell surface proteases, including serine proteases, as proteolytic modifiers of particular targets, including growth factors and protease-activated receptors, which are critical for the activation of oncogenic signaling pathways. Of the 176 human serine proteases currently identified, a subset of 17, known as type II transmembrane serine proteases (TTSPs). Many have been shown to be relevant to cancer progression since they were first identified as a family around the turn of the century. To this end, altered expression of TTSPs appeared as a trademark of several tumor types. However, the substrates and underlying signaling pathways remained unclear. Localization of these proteins to the cell surface places them in the unique position to mediate signal transduction between the cell and its surrounding environment. Many of the TTSPs have already been shown to play key roles in processes such as postnatal development, tissue homeostasis, and tumor progression, which share overlapping molecular mechanisms. In this review, we summarize the current knowledge regarding the role of the TTSP family in pro-oncogenic signaling.
Collapse
Affiliation(s)
- Lauren M Tanabe
- Department of Pharmacology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Karin List
- Department of Pharmacology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
33
|
Natural Endogenous Human Matriptase and Prostasin Undergo Zymogen Activation via Independent Mechanisms in an Uncoupled Manner. PLoS One 2016; 11:e0167894. [PMID: 27936035 PMCID: PMC5148038 DOI: 10.1371/journal.pone.0167894] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022] Open
Abstract
The membrane-associated serine proteases matriptase and prostasin are believed to function in close partnership. Their zymogen activation has been reported to be tightly coupled, either as a matriptase-initiated proteolytic cascade or through a mutually dependent mechanism involving the formation of a reciprocal zymogen activation complex. Here we show that this putative relationship may not apply in the context of human matriptase and prostasin. First, the tightly coupled proteolytic cascade between matriptase and prostasin might not occur when modest matriptase activation is induced by sphingosine 1-phospahte in human mammary epithelial cells. Second, prostasin is not required and/or involved in matriptase autoactivation because matriptase can undergo zymogen activation in cells that do not endogenously express prostasin. Third, matriptase is not required for and/or involved in prostasin activation, since activated prostasin can be detected in cells expressing no endogenous matriptase. Finally, matriptase and prostasin both undergo zymogen activation through an apparently un-coupled mechanism in cells endogenously expressing both proteases, such as in Caco-2 cells. In these human enterocytes, matriptase is detected primarily in the zymogen form and prostasin predominantly as the activated form, either in complexes with protease inhibitors or as the free active form. The negligible levels of prostasin zymogen with high levels of matriptase zymogen suggests that the reciprocal zymogen activation complex is likely not the mechanism for matriptase zymogen activation. Furthermore, high level prostasin activation still occurs in Caco-2 variants with reduced or absent matriptase expression, indicating that matriptase is not required and/or involved in prostasin zymogen activation. Collectively, these data suggest that any functional relationship between natural endogenous human matriptase and prostasin does not occur at the level of zymogen activation.
Collapse
|
34
|
Martin EW, Buzza MS, Driesbaugh KH, Liu S, Fortenberry YM, Leppla SH, Antalis TM. Targeting the membrane-anchored serine protease testisin with a novel engineered anthrax toxin prodrug to kill tumor cells and reduce tumor burden. Oncotarget 2016; 6:33534-53. [PMID: 26392335 PMCID: PMC4741784 DOI: 10.18632/oncotarget.5214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/03/2015] [Indexed: 02/04/2023] Open
Abstract
The membrane-anchored serine proteases are a unique group of trypsin-like serine proteases that are tethered to the cell surface via transmembrane domains or glycosyl-phosphatidylinositol-anchors. Overexpressed in tumors, with pro-tumorigenic properties, they are attractive targets for protease-activated prodrug-like anti-tumor therapies. Here, we sought to engineer anthrax toxin protective antigen (PrAg), which is proteolytically activated on the cell surface by the proprotein convertase furin to instead be activated by tumor cell-expressed membrane-anchored serine proteases to function as a tumoricidal agent. PrAg's native activation sequence was mutated to a sequence derived from protein C inhibitor (PCI) that can be cleaved by membrane-anchored serine proteases, to generate the mutant protein PrAg-PCIS. PrAg-PCIS was resistant to furin cleavage in vitro, yet cytotoxic to multiple human tumor cell lines when combined with FP59, a chimeric anthrax toxin lethal factor-Pseudomonas exotoxin fusion protein. Molecular analyses showed that PrAg-PCIS can be cleaved in vitro by several serine proteases including the membrane-anchored serine protease testisin, and mediates increased killing of testisin-expressing tumor cells. Treatment with PrAg-PCIS also potently attenuated the growth of testisin-expressing xenograft tumors in mice. The data indicates PrAg can be engineered to target tumor cell-expressed membrane-anchored serine proteases to function as a potent tumoricidal agent.
Collapse
Affiliation(s)
- Erik W Martin
- Center for Vascular and Inflammatory Diseases and the Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Marguerite S Buzza
- Center for Vascular and Inflammatory Diseases and the Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kathryn H Driesbaugh
- Center for Vascular and Inflammatory Diseases and the Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shihui Liu
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yolanda M Fortenberry
- Division of Pediatric Hematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephen H Leppla
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Toni M Antalis
- Center for Vascular and Inflammatory Diseases and the Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
35
|
Jin JS, Chen A, Hsieh DS, Yao CW, Cheng MF, Lin YF. Expression of Serine Protease Matriptase in Renal Cell Carcinoma: Correlation of Tissue Microarray Immunohistochemical Expression Analysis Results with Clinicopathological Parameters. Int J Surg Pathol 2016; 14:65-72. [PMID: 16501837 DOI: 10.1177/106689690601400111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Serine protease matriptase (matriptase) cleaves and activates proteins implicated in the progression of cancer and represents a potential therapeutic target. Immunohistochemical analysis of matriptase was performed in tissue microarrays of 168 renal cell carcinomas (RCCs). All subtypes of RCC showed significant immunohistochemical expression of matriptase. In contrast, no expression occurred in areas of RCC with sarcomatous differentiation (SRCC) and in normal collecting tubules. The matriptase scores were significantly higher in papillary RCC (341) and clear cell RCC with granular cell differentiation (GRCC; 324) than in other histologic subtypes of RCC. In GRCC, matriptase scores were correlated with TNM staging and nuclear grading. Matriptase was overexpressed in all subtypes of RCC, and matriptase scores could distinguish between conventional clear cell RCC, GRCC, and SRCC.
Collapse
Affiliation(s)
- Jong-Shiaw Jin
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, No. 325, Sec. 2, Cheng-Gong Road, Taipei, Taiwan, R.O.C
| | | | | | | | | | | |
Collapse
|
36
|
Chen YW, Yin S, Lai YJJ, Johnson MD, Lin CY. Plasminogen-Dependent Matriptase Activation Accelerates Plasmin Generation by Differentiating Primary Human Keratinocytes. J Invest Dermatol 2016; 136:1210-1218. [DOI: 10.1016/j.jid.2016.01.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 01/19/2016] [Accepted: 01/25/2016] [Indexed: 11/25/2022]
|
37
|
Inhibition of Matriptase Activity Results in Decreased Intestinal Epithelial Monolayer Integrity In Vitro. PLoS One 2015; 10:e0141077. [PMID: 26488575 PMCID: PMC4619522 DOI: 10.1371/journal.pone.0141077] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/04/2015] [Indexed: 02/08/2023] Open
Abstract
Barrier dysfunction in inflammatory bowel diseases implies enhanced paracellular flux and lowered transepithelial electrical resistance (TER) causing effective invasion of enteropathogens or altered intestinal absorption of toxins and drug compounds. To elucidate the role of matriptase-driven cell surface proteolysis in the maintenance of intestinal barrier function, the 3-amidinophenylalanine-derived matriptase inhibitor, MI-432 was used on porcine IPEC-J2 cell monolayer. Studies with two fluorescent probes revealed that short (2 h) treatment with MI-432 caused an altered distribution of oxidative species between intracellular and extracellular spaces in IPEC-J2 cells. This perturbation was partially compensated when administration of inhibitor continued for up to 48 h. Significant decrease in TER between apical and basolateral compartments of MI-432-treated IPEC-J2 cell monolayers proved that matriptase is one of the key effectors in the maintenance of barrier integrity. Changes in staining pattern of matriptase and in localization of the junctional protein occludin were observed suggesting that inhibition of matriptase by MI-432 can also exert an effect on paracellular gate opening via modulation of tight junctional protein assembly. This study confirms that non-tumorigenic IPEC-J2 cells can be used as an appropriate small intestinal model for the in vitro characterization of matriptase-related effects on intestinal epithelium. These findings demonstrate indirectly that matriptase plays a pivotal role in the development of barrier integrity; thus matriptase dysfunction can facilitate the occurence of leaky gut syndrome observed in intestinal inflammatory diseases.
Collapse
|
38
|
Zoratti GL, Tanabe LM, Varela FA, Murray AS, Bergum C, Colombo É, Lang JE, Molinolo AA, Leduc R, Marsault E, Boerner J, List K. Targeting matriptase in breast cancer abrogates tumour progression via impairment of stromal-epithelial growth factor signalling. Nat Commun 2015; 6:6776. [PMID: 25873032 PMCID: PMC4749267 DOI: 10.1038/ncomms7776] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 02/24/2015] [Indexed: 02/07/2023] Open
Abstract
Matriptase is an epithelia-specific membrane-anchored serine protease that has received considerable attention in recent years due to its consistent dysregulation in human epithelial tumors, including breast cancer. Mice with reduced levels of matriptase display a significant delay in oncogene-induced mammary tumor formation and blunted tumor growth. The abated tumor growth is associated with a decrease in cancer cell proliferation. Here we demonstrate by genetic deletion and silencing that the proliferation impairment in matriptase deficient breast cancer cells is caused by their inability to initiate activation of the c-Met signaling pathway in response to fibroblast-secreted pro-HGF. Similarly, inhibition of matriptase catalytic activity using a selective small-molecule inhibitor abrogates the activation of c-Met, Gab1 and AKT, in response to pro-HGF, which functionally leads to attenuated proliferation in breast carcinoma cells. We conclude that matriptase is critically involved in breast cancer progression and represents a potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Gina L Zoratti
- 1] Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA [2] Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA [3] Cancer Biology Graduate Program, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 110 E. Warren Avenue, Suite 2215, Detroit, Michigan 48201, USA
| | - Lauren M Tanabe
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA
| | - Fausto A Varela
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA
| | - Andrew S Murray
- 1] Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA [2] Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA [3] Cancer Biology Graduate Program, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 110 E. Warren Avenue, Suite 2215, Detroit, Michigan 48201, USA
| | - Christopher Bergum
- Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA
| | - Éloïc Colombo
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Av Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | - Julie E Lang
- Department of Surgery, Norris Comprehensive Cancer Center, University of Southern California, 1510 San Pablo Street, Suite 412, Los Angeles, California 90033, USA
| | - Alfredo A Molinolo
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 211, Bethesda, Maryland 20892, USA
| | - Richard Leduc
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Av Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | - Eric Marsault
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001 12e Av Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | - Julie Boerner
- Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA
| | - Karin List
- 1] Department of Pharmacology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA [2] Department of Oncology, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 540 E Canfield, Scott Hall Room 6332, Detroit, Michigan 48201, USA [3] Cancer Biology Graduate Program, Wayne State University School of Medicine and Barbara Ann Karmanos Cancer Institute, 110 E. Warren Avenue, Suite 2215, Detroit, Michigan 48201, USA
| |
Collapse
|
39
|
Fang JD, Lee SL. Matriptase is required for the active form of hepatocyte growth factor induced Met, focal adhesion kinase and protein kinase B activation on neural stem/progenitor cell motility. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1285-94. [DOI: 10.1016/j.bbamcr.2014.03.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/19/2014] [Accepted: 03/23/2014] [Indexed: 12/31/2022]
|
40
|
Wang CY, Meynard D, Lin HY. The role of TMPRSS6/matriptase-2 in iron regulation and anemia. Front Pharmacol 2014; 5:114. [PMID: 24966834 PMCID: PMC4053654 DOI: 10.3389/fphar.2014.00114] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/29/2014] [Indexed: 01/12/2023] Open
Abstract
Matriptase-2, encoded by the TMPRSS6 gene, is a member of the type II transmembrane serine protease family. Matriptase-2 has structural and enzymatic similarities to matriptase-1, which has been implicated in cancer progression. Matriptase-2 was later established to be essential in iron homeostasis based on the phenotypes of iron-refractory iron deficiency anemia identified in mouse models as well as in human patients with TMPRSS6 mutations. TMPRSS6 is expressed mainly in the liver and negatively regulates the production of hepcidin, the systemic iron regulatory hormone. This review focuses on the current understanding of matriptase-2 biochemistry, and its role in iron metabolism and cancer progression. In light of recent investigations, the function of matriptase-2 in hepcidin regulation, how it is being regulated, as well as the therapeutic potential of matriptase-2 are also discussed.
Collapse
Affiliation(s)
- Chia-Yu Wang
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School Boston, MA, USA
| | - Delphine Meynard
- INSERM, U1043, CNRS, U5282, Université Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan Toulouse, France
| | - Herbert Y Lin
- Program in Anemia Signaling Research, Division of Nephrology, Program in Membrane Biology, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School Boston, MA, USA
| |
Collapse
|
41
|
Chu LL, Xu Y, Yang JR, Hu YA, Chang HH, Lai HY, Tseng CC, Wang HY, Johnson MD, Wang JK, Lin CY. Human cancer cells retain modest levels of enzymatically active matriptase only in extracellular milieu following induction of zymogen activation. PLoS One 2014; 9:e92244. [PMID: 24663123 PMCID: PMC3963879 DOI: 10.1371/journal.pone.0092244] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 02/09/2014] [Indexed: 11/18/2022] Open
Abstract
The type 2 transmembrane serine protease matriptase is broadly expressed in human carcinomas and hematological cancers. The proteolytic activity of matriptase is a potential target of drugs and imaging probes. We assessed the fate of active matriptase following the induction of matriptase zymogen activation. Exposing eight human carcinoma cells to pH 6.0 buffer induced robust matriptase zymogen activation followed by rapid inhibition of the nascent active matriptase by hepatocyte growth factor activator inhibitor (HAI)-1. Consequently, no enzymatically active matriptase was detected in these cells. Some active matriptase is, however, rapidly shed to the extracellular milieu by these carcinoma cells. The lack of cell-associated active matriptase and the shedding of active matriptase were also observed in two hematological cancer lines. Matriptase shedding is correlated closely with the induction of matriptase activation, suggesting that matriptase activation and shedding are kinetically coupled. The coupling allows a proportion of active matriptase to survive HAI-1 inhibition by rapid shedding from cell surface. Our study suggests that cellular free, active matriptase is scarce and might not be an effective target for in vivo imaging and drug development.
Collapse
Affiliation(s)
- Li-Ling Chu
- Department of Pharmacy, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yuan Xu
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, D.C., United States of America
| | - Jie-Ru Yang
- Department of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Yi-An Hu
- Department of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hsiang-Hua Chang
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, D.C., United States of America
- Department of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Hong-Yu Lai
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, D.C., United States of America
| | - Chun-Che Tseng
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, D.C., United States of America
- Department of Biology, Carleton College, Northfield, Minnesota, United States of America
| | - Hue-Yu Wang
- Department of Pharmacy, Chi-Mei Medical Center, Tainan, Taiwan
| | - Michael D. Johnson
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, D.C., United States of America
| | - Jehng-Kang Wang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
- * E-mail: (C-YL); (J-KW)
| | - Chen-Yong Lin
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, D.C., United States of America
- * E-mail: (C-YL); (J-KW)
| |
Collapse
|
42
|
Dorn J, Beaufort N, Schmitt M, Diamandis EP, Goettig P, Magdolen V. Function and clinical relevance of kallikrein-related peptidases and other serine proteases in gynecological cancers. Crit Rev Clin Lab Sci 2014; 51:63-84. [PMID: 24490956 DOI: 10.3109/10408363.2013.865701] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gynecological cancers, including malignant tumors of the ovaries, the endometrium and the cervix, account for approximately 10% of tumor-associated deaths in women of the Western world. For screening, diagnosis, prognosis, and therapy response prediction, the group of enzymes known as serine (Ser-)proteases show great promise as biomarkers. In the present review, following a summary of the clinical facts regarding malignant tumors of the ovaries, the endometrium and the cervix, and characterization of the most important Ser-proteases, we thoroughly review the current state of knowledge relating to the use of proteases as biomarkers of the most frequent gynecological cancers. Within the Ser-protease group, the kallikrein-related peptidase (KLK) family, which encompasses a subgroup of 15 members, holds particular promise, with some acting via a tumor-promoting mechanism and others behaving as protective factors. Further, the urokinase-type plasminogen activator (uPA) and its inhibitor PAI-1 (plasminogen activator inhibitor-1) seem to play an unfavorable role in gynecological tumors, while down-regulation of high-temperature requirement proteins A 1, 2 and 3 (HtrA1,2,3) is associated with malignant disease and cancer progression. Expression/activity levels of other Ser-proteases, including the type II transmembrane Ser-proteases (TTSPs) matriptase, hepsin (TMPRSS1), and the hepsin-related protease (TMPRSS3), as well as the glycosyl-phosphatidylinositol (GPI)-anchored Ser-proteases prostasin and testisin, may be of clinical relevance in gynecological cancers. In conclusion, proteases are a rich source of biomarkers of gynecological cancer, though the enzymes' exact roles and functions merit further investigation.
Collapse
Affiliation(s)
- Julia Dorn
- Klinische Forschergruppe der Frauenklinik der Technischen Universität München, Klinikum rechts der Isar , Munich , Germany
| | | | | | | | | | | |
Collapse
|
43
|
Sales KU, Friis S, Konkel JE, Godiksen S, Hatakeyama M, Hansen KK, Rogatto SR, Szabo R, Vogel LK, Chen W, Gutkind JS, Bugge TH. Non-hematopoietic PAR-2 is essential for matriptase-driven pre-malignant progression and potentiation of ras-mediated squamous cell carcinogenesis. Oncogene 2014; 34:346-56. [PMID: 24469043 PMCID: PMC4112178 DOI: 10.1038/onc.2013.563] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/06/2013] [Accepted: 12/10/2013] [Indexed: 01/27/2023]
Abstract
The membrane-anchored serine protease, matriptase, is consistently dysregulated in a range of human carcinomas, and high matriptase activity correlates with poor prognosis. Furthermore, matriptase is unique among tumor-associated proteases in that epithelial stem cell expression of the protease suffices to induce malignant transformation. Here, we use genetic epistasis analysis to identify proteinase-activated receptor (PAR)-2-dependent inflammatory signaling as an essential component of matriptase-mediated oncogenesis. In cell-based assays, matriptase was a potent activator of PAR-2, and PAR-2 activation by matriptase caused robust induction of nuclear factor (NF)κB through Gαi. Importantly, genetic elimination of PAR-2 from mice completely prevented matriptase-induced pre-malignant progression, including inflammatory cytokine production, inflammatory cell recruitment, epidermal hyperplasia and dermal fibrosis. Selective ablation of PAR-2 from bone marrow-derived cells did not prevent matriptase-driven pre-malignant progression, indicating that matriptase activates keratinocyte stem cell PAR-2 to elicit its pro-inflammatory and pro-tumorigenic effects. When combined with previous studies, our data suggest that dual induction of PAR-2-NFκB inflammatory signaling and PI3K-Akt-mTor survival/proliferative signaling underlies the transforming potential of matriptase and may contribute to pro-tumorigenic signaling in human epithelial carcinogenesis.
Collapse
Affiliation(s)
- K U Sales
- 1] Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA [2] Clinical Research Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S Friis
- 1] Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA [2] Department of Cellular and Molecular Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - J E Konkel
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S Godiksen
- 1] Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA [2] Department of Cellular and Molecular Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark [3] Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - M Hatakeyama
- 1] Department of Urology, Faculty of Medicine, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, Brazil [2] AC Camargo Cancer Center, Sao Paulo, Brazil
| | - K K Hansen
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S R Rogatto
- 1] Department of Urology, Faculty of Medicine, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, Brazil [2] AC Camargo Cancer Center, Sao Paulo, Brazil
| | - R Szabo
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - L K Vogel
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - W Chen
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - J S Gutkind
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - T H Bugge
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
44
|
HAI-2 suppresses the invasive growth and metastasis of prostate cancer through regulation of matriptase. Oncogene 2013; 33:4643-52. [PMID: 24121274 DOI: 10.1038/onc.2013.412] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 08/19/2013] [Accepted: 09/02/2013] [Indexed: 01/06/2023]
Abstract
Dysregulation of cell surface proteolysis has been strongly implicated in tumorigenicity and metastasis. In this study, we delineated the role of hepatocyte growth factor activator inhibitor-2 (HAI-2) in prostate cancer (PCa) cell migration, invasion, tumorigenicity and metastasis using a human PCa progression model (103E, N1, and N2 cells) and xenograft models. N1 and N2 cells were established through serial intraprostatic propagation of 103E human PCa cells and isolation of the metastatic cells from nearby lymph nodes. The invasion capability of these cells was revealed to gradually increase throughout the serial isolations (103E<N1<N2). In this series of cells, the expression of HAI-2 but not HAI-1 was significantly decreased throughout the progression and occurred in parallel with increased activation of matriptase. The expression level and activity of matriptase increased whereas the HAI-2 protein level decreased over the course of orthotopic tumor growth in mice, which was consistent with the immunohistochemical profiles of matriptase and HAI-2 in archival PCa specimens. Knockdown of matriptase reduced the PCa cell invasion induced by HAI-2 knockdown. HAI-2 overexpression or matriptase silencing in N2 cells downregulated matriptase activity and significantly decreased tumorigenicity and metastatic capability in orthotopically xenografted mice. These results suggest that during the progression of human PCa, matriptase activity is primarily controlled by HAI-2 expression. The imbalance between HAI-2 and matriptase expression led to matriptase activation, thereby increasing cell migration, invasion, tumorigenicity and metastasis.
Collapse
|
45
|
Chou FP, Chen YW, Zhao XF, Xu-Monette ZY, Young KH, Gartenhaus RB, Wang JK, Kataoka H, Zuo AH, Barndt RJ, Johnson M, Lin CY. Imbalanced matriptase pericellular proteolysis contributes to the pathogenesis of malignant B-cell lymphomas. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1306-1317. [PMID: 24070417 PMCID: PMC3791685 DOI: 10.1016/j.ajpath.2013.06.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 05/17/2013] [Accepted: 06/24/2013] [Indexed: 02/08/2023]
Abstract
Membrane-associated serine protease matriptase is widely expressed by epithelial/carcinoma cells in which its proteolytic activity is tightly controlled by the Kunitz-type protease inhibitor, hepatocyte growth factor activator inhibitor (HAI-1). We demonstrate that, although matriptase is not expressed in lymphoid hyperplasia, roughly half of the non-Hodgkin B-cell lymphomas analyzed express significant amounts of matriptase. Furthermore, a significant proportion of these tumors express matriptase in the absence of HAI-1. Aggressive Burkitt lymphoma was more likely than indolent follicular lymphoma to express matriptase alone (86% versus 36%). In the absence of significant HAI-1 expression, the lymphoma cells activate and shed active matriptase when the cells are stimulated with mildly acidic buffer or the hypoxia-mimicking agent, CoCl2. The shed active matriptase can initiate pericellular proteolytic cascades by activating urokinase-type plasminogen activator on the cell surface of monocytes, and it can activate prohepatocyte growth factor. In addition, matriptase knockdown suppressed proliferation and colony-forming ability of neoplastic B cells in culture and growth as tumor xenografts in mice. Furthermore, exogenous expression of HAI-1 significantly suppressed proliferation of neoplastic B cells. These studies suggest that dysregulated pericellular proteolysis as a result of unregulated matriptase expression with limited HAI-1 may contribute to the pathological characteristics of several human B-cell lymphomas through modulation of the tumor microenvironment and enhanced tumor growth.
Collapse
Affiliation(s)
- Feng-Pai Chou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Ya-Wen Chen
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Xianfeng F. Zhao
- Department of Pathology, University of Maryland, Baltimore, Maryland
| | - Zijun Y. Xu-Monette
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ken H. Young
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ronald B. Gartenhaus
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Maryland
| | - Jehng-Kang Wang
- Department of Biochemistry, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Hiroaki Kataoka
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Annie H. Zuo
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Robert J. Barndt
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Michael Johnson
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| | - Chen-Yong Lin
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia
| |
Collapse
|
46
|
Hepatocyte growth factor activator inhibitor-1 is induced by bone morphogenetic proteins and regulates proliferation and cell fate of neural progenitor cells. PLoS One 2013; 8:e56117. [PMID: 23409135 PMCID: PMC3567048 DOI: 10.1371/journal.pone.0056117] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 01/04/2013] [Indexed: 12/20/2022] Open
Abstract
Background Neural progenitor cells (NPCs) in the developing neuroepithelium are regulated by intrinsic and extrinsic factors. There is evidence that NPCs form a self-supporting niche for cell maintenance and proliferation. However, molecular interactions and cell-cell contacts and the microenvironment within the neuroepithelium are largely unknown. We hypothesized that cellular proteases especially those associated with the cell surface of NPCs play a role in regulation of progenitor cells in the brain. Methodology/Principal Findings In this work, we show that NPCs, isolated from striatal anlage of developing rat brain, express hepatocyte growth factor activator inhibitor-1 and -2 (HAI-1 and HAI-2) that are cell surface-linked serine protease inhibitors. In addition, radial glia cells derived from mouse embryonic stem cells also express HAI-1 and HAI-2. To study the functional significance of HAI-1 and HAI-2 in progenitor cells, we modulated their levels using expression plasmids or silencing RNA (siRNA) transfected into the NPCs. Data showed that overexpression of HAI-1 or HAI-2 decreased cell proliferation of cultured NPCs, whilst their siRNAs had opposite effects. HAI-1 also influenced NPC differentiation by increasing the number of glial fibrillary acidic protein (GFAP) expressing cells in the culture. Expression of HAI-1 in vivo decreased cell proliferation in developing neuroepithelium in E15 old animals and promoted astrocyte cell differentiation in neonatal animals. Studying the regulation of HAI-1, we observed that Bone morphogenetic protein-2 (BMP-2) and BMP-4 increased HAI-1 levels in the NPCs. Experiments using HAI-1-siRNA showed that these BMPs act on the NPCs partly in a HAI-1-dependent manner. Conclusions This study shows that the cell-surface serine protease inhibitors, HAI-1 and HAI-2 influence proliferation and cell fate of NPCs and their expression levels are linked to BMP signaling. Modulation of the levels and actions of HAI-1 in NPCs may be of a potential value in stem cell therapies in various brain diseases.
Collapse
|
47
|
Larsen BR, Steffensen SDR, Nielsen NVL, Friis S, Godiksen S, Bornholdt J, Soendergaard C, Nonboe AW, Andersen MN, Poulsen SS, Szabo R, Bugge TH, Lin CY, Skovbjerg H, Jensen JK, Vogel LK. Hepatocyte growth factor activator inhibitor-2 prevents shedding of matriptase. Exp Cell Res 2013; 319:918-29. [PMID: 23333561 DOI: 10.1016/j.yexcr.2013.01.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 01/02/2013] [Accepted: 01/07/2013] [Indexed: 12/31/2022]
Abstract
Hepatocyte growth factor activator inhibitor-2 (HAI-2) is an inhibitor of many proteases in vitro, including the membrane-bound serine protease, matriptase. Studies of knock-out mice have shown that HAI-2 is essential for placental development only in mice expressing matriptase, suggesting that HAI-2 is important for regulation of matriptase. Previous studies have shown that recombinant expression of matriptase was unsuccessful unless co-expressed with another HAI, HAI-1. In the present study we show that when human matriptase is recombinantly expressed alone in the canine cell line MDCK, then human matriptase mRNA can be detected and the human matriptase ectodomain is shed to the media, suggesting that matriptase expressed alone is rapidly transported through the secretory pathway and shed. Whereas matriptase expressed together with HAI-1 or HAI-2 accumulates on the plasma membrane where it is activated, as judged by cleavage at Arg614 and increased peptidolytic activity of the cell extracts. Mutagenesis of Kunitz domain 1 but not Kunitz domain 2 abolished this function of HAI-2. HAI-2 seems to carry out its function intracellularly as this is where the vast majority of HAI-2 is located and since HAI-2 could not be detected on the basolateral plasma membrane where matriptase resides. However, minor amounts of HAI-2 not undergoing endocytosis could be detected on the apical plasma membrane. Our results suggest that Kunitz domain 1 of HAI-2 cause matriptase to accumulate in a membrane-bound form on the basolateral plasma membrane.
Collapse
Affiliation(s)
- Brian R Larsen
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
From prediction to experimental validation: desmoglein 2 is a functionally relevant substrate of matriptase in epithelial cells and their reciprocal relationship is important for cell adhesion. Biochem J 2012; 447:61-70. [PMID: 22783993 DOI: 10.1042/bj20111432] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Accurate identification of substrates of a protease is critical in defining its physiological functions. We previously predicted that Dsg-2 (desmoglein-2), a desmosomal protein, is a candidate substrate of the transmembrane serine protease matriptase. The present study is an experimental validation of this prediction. As demanded by our published method PNSAS [Prediction of Natural Substrates from Artificial Substrate of Proteases; Venkatraman, Balakrishnan, Rao, Hooda and Pol (2009) PLoS ONE 4, e5700], this enzyme-substrate pair shares a common subcellular distribution and the predicted cleavage site is accessible to the protease. Matriptase knock-down cells showed enhanced immunoreactive Dsg-2 at the cell surface and formed larger cell clusters. When matriptase was mobilized from intracellular storage deposits to the cell surface there was a decrease in the band intensity of Dsg-2 in the plasma membrane fractions with a concomitant accumulation of a cleaved product in the conditioned medium. The exogenous addition of pure active recombinant matriptase decreased the surface levels of immunoreactive Dsg-2, whereas the levels of CD44 and E-cadherin were unaltered. Dsg-2 with a mutation at the predicted cleavage site is resistant to cleavage by matriptase. Thus Dsg-2 seems to be a functionally relevant physiological substrate of matriptase. Since breakdown of cell-cell contact is the first major event in invasion, this reciprocal relationship is likely to have a profound role in cancers of epithelial origin. Our algorithm has the potential to become an integral tool for discovering new protease-substrate pairs.
Collapse
|
49
|
Netzel-Arnett S, Buzza MS, Shea-Donohue T, Désilets A, Leduc R, Fasano A, Bugge TH, Antalis TM. Matriptase protects against experimental colitis and promotes intestinal barrier recovery. Inflamm Bowel Dis 2012; 18:1303-14. [PMID: 22081509 PMCID: PMC3288858 DOI: 10.1002/ibd.21930] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 09/28/2011] [Indexed: 12/24/2022]
Abstract
BACKGROUND Matriptase is a membrane-anchored serine protease encoded by suppression of tumorigenicity-14 (ST14) that is required for epithelial barrier homeostasis. However, its functional role in inflammatory bowel disease (IBD) is unexplored. METHODS Matriptase expression in control, Crohn's disease, and ulcerative colitis tissue specimens was studied by quantitative polymerase chain reaction (qPCR) and immunostaining. Matriptase function was investigated by subjecting St14 hypomorphic and control littermates to dextran sodium sulfate (DSS)-induced colitis and by siRNA silencing in cultured monolayers. Mice were analyzed for clinical, histological, molecular, and cellular effects. RESULTS Matriptase protein and ST14 mRNA levels are significantly downregulated in inflamed colonic tissues from Crohn's disease and ulcerative colitis patients. Matriptase-deficient St14 hypomorphic mice administered DSS for 7 days followed by water without DSS for 3 days develop a severe colitis, with only 30% of the St14 hypomorphic mice surviving to day 14, compared with 100% of control littermates. Persistent colitis in surviving St14 hypomorphic mice was associated with sustained cytokine production, an inability to recover barrier integrity, and enhanced claudin-2 expression. Cytokines implicated in barrier disruption during IBD suppress matriptase expression in T84 epithelial monolayers and restoration of matriptase improves barrier integrity in the cytokine-perturbed monolayers. CONCLUSIONS These data demonstrate a critical role for matriptase in restoring barrier function to injured intestinal mucosa during colitis, which is suppressed by excessive activation of the immune system. Strategies to enhance matriptase-mediated barrier recovery could be important for intervening in the cycle of inflammation associated with IBD.
Collapse
Affiliation(s)
- Sarah Netzel-Arnett
- Center for Vascular and Inflammatory Diseases and Department of Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Québec, Canada
| | - Marguerite S. Buzza
- Center for Vascular and Inflammatory Diseases and Department of Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Québec, Canada
| | - Terez Shea-Donohue
- Mucosal Biology Research Center, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Québec, Canada
| | - Antoine Désilets
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Québec, Canada
| | - Richard Leduc
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Québec, Canada
| | - Alessio Fasano
- Mucosal Biology Research Center, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Québec, Canada
| | - Thomas H. Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Cranofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Toni M. Antalis
- Center for Vascular and Inflammatory Diseases and Department of Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Québec, Canada,Corresponding author: Toni M. Antalis Ph.D., The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, 800 West Baltimore St, Baltimore MD 21201 USA. Ph: 410 706 8222; Fax: 410 706 8121;
| |
Collapse
|
50
|
Platelet-derived growth factor-C (PDGF-C) activation by serine proteases: implications for breast cancer progression. Biochem J 2012; 441:909-18. [PMID: 22035541 DOI: 10.1042/bj20111020] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The PDGF (platelet-derived growth factor) family members are potent mitogens for cells of mesenchymal origin and serve as important regulators of cell migration, survival, apoptosis and transformation. Tumour-derived PDGF ligands are thought to function in both autocrine and paracrine manners, activating receptors on tumour and surrounding stromal cells. PDGF-C and -D are secreted as latent dimers, unlike PDGF-A and -B. Cleavage of the CUB domain from the PDGF-C and -D dimers is required for their biological activity. At present, little is known about the proteolytic processing of PDGF-C, the rate-limiting step in the regulation of PDGF-C activity. In the present study we show that the breast carcinoma cell line MCF7, engineered to overexpress PDGF-C, produces proteases capable of cleaving PDGF-C to its active form. Increased PDGF-C expression enhances cell proliferation, anchorage-independent cell growth and tumour cell motility by autocrine signalling. In addition, MCF7-produced PDGF-C induces fibroblast cell migration in a paracrine manner. Interestingly, PDGF-C enhances tumour cell invasion in the presence of fibroblasts, suggesting a role for tumour-derived PDGF-C in tumour-stromal interactions. In the present study, we identify tPA (tissue plasminogen activator) and matriptase as major proteases for processing of PDGF-C in MCF7 cells. In in vitro studies, we also show that uPA (urokinase-type plasminogen activator) is able to process PDGF-C. Furthermore, by site-directed mutagenesis, we identify the cleavage site for these proteases in PDGF-C. Lastly, we provide evidence suggesting a two-step proteolytic processing of PDGF-C involving creation of a hemidimer, followed by GFD-D (growth factor domain dimer) generation.
Collapse
|