1
|
Wang F, Yi J, Chen Y, Bai X, Lu C, Feng S, Zhou X. PRSS2 regulates EMT and metastasis via MMP-9 in gastric cancer. Acta Histochem 2023; 125:152071. [PMID: 37331089 DOI: 10.1016/j.acthis.2023.152071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023]
Abstract
Serine protease 2 (PRSS2) is upregulated in gastric cancer tissues, correlates with poor prognosis and promotes migration and invasion of gastric cancer cells. However, the exact mechanism by which PRSS2 promotes metastasis in gastric cancer is unclear. We examined serum PRSS2 levels in healthy controls and gastric cancer patients by enzyme linked immunosorbent assay (ELISA) and analyzed the correlation between PRSS2 serum level with the clinicopathological characteristics of gastric cancer patients and matrix metalloproteinase-9 (MMP-9) expression. A lentiviral MMP-9 overexpression vector was constructed and used to transfect gastric cancer cells with stable silencing of PRSS2, and migration, invasion and epithelial-mesenchymal transition (EMT) of gastric cancer cells were examined. High serum PRSS2 levels were detected in gastric cancer patients and associated with lymphatic metastasis and TNM stage. Serum PRSS2 was positively correlated with serum MMP-9 level. PRSS2 silencing inhibited EMT, and knock-down of PRSS2 partially abrogated cell metastasis and EMT caused by overexpression of MMP-9. These results suggest that PRSS2 promotes the migration and invasion of gastric cancer cells through EMT induction by MMP-9. Our findings suggest that PRSS2 may be a potential early diagnostic marker and therapeutic target of gastric cancer.
Collapse
Affiliation(s)
- Fei Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Department of General Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Jianfeng Yi
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Medical school of Nantong University, Nantong, Jiangsu 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Yu Chen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Medical school of Nantong University, Nantong, Jiangsu 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Xiang Bai
- Department of General Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Chunfeng Lu
- Department of Endocrinology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Shichun Feng
- Department of General Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Xiaojun Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
2
|
Chen Y, Wang B, Zhao Z, Li M, Wang F. PRSS2 overexpression relates to poor prognosis and promotes proliferation, migration and invasion in gastric cancer. Tissue Cell 2022; 79:101949. [DOI: 10.1016/j.tice.2022.101949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/19/2022] [Accepted: 09/28/2022] [Indexed: 12/24/2022]
|
3
|
Abstract
Supramolecular assemblies are essential components of living organisms. Cellular scaffolds, such as the cytoskeleton or the cell membrane, are formed via secondary interactions between proteins or lipids and direct biological processes such as metabolism, proliferation and transport. Inspired by nature’s evolution of function through structure formation, a range of synthetic nanomaterials has been developed in the past decade, with the goal of creating non-natural supramolecular assemblies inside living mammalian cells. Given the intricacy of biological pathways and the compartmentalization of the cell, different strategies can be employed to control the assembly formation within the highly crowded, dynamic cellular environment. In this Review, we highlight emerging molecular design concepts aimed at creating precursors that respond to endogenous stimuli to build nanostructures within the cell. We describe the underlying reaction mechanisms that can provide spatial and temporal control over the subcellular formation of synthetic nanostructures. Showcasing recent advances in the development of bioresponsive nanomaterials for intracellular self-assembly, we also discuss their impact on cellular function and the challenges associated with establishing structure–bioactivity relationships, as well as their relevance for the discovery of novel drugs and imaging agents, to address the shortfall of current solutions to pressing health issues. ![]()
Creating artificial nanostructures inside living cells requires the careful design of molecules that can transform into active monomers within a complex cellular environment. This Review explores the recent development of bioresponsive precursors for the controlled formation of intracellular supramolecular assemblies.
Collapse
|
4
|
Park KC, Dharmasivam M, Richardson DR. The Role of Extracellular Proteases in Tumor Progression and the Development of Innovative Metal Ion Chelators that Inhibit their Activity. Int J Mol Sci 2020; 21:E6805. [PMID: 32948029 PMCID: PMC7555822 DOI: 10.3390/ijms21186805] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022] Open
Abstract
The crucial role of extracellular proteases in cancer progression is well-known, especially in relation to the promotion of cell invasion through extracellular matrix remodeling. This also occurs by the ability of extracellular proteases to induce the shedding of transmembrane proteins at the plasma membrane surface or within extracellular vesicles. This process results in the regulation of key signaling pathways by the modulation of kinases, e.g., the epidermal growth factor receptor (EGFR). Considering their regulatory roles in cancer, therapeutics targeting various extracellular proteases have been discovered. These include the metal-binding agents di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), which increase c-MET degradation by multiple mechanisms. Both the direct and indirect inhibition of protease expression and activity can be achieved through metal ion depletion. Considering direct mechanisms, chelators can bind zinc(II) that plays a catalytic role in enzyme activity. In terms of indirect mechanisms, Dp44mT and DpC potently suppress the expression of the kallikrein-related peptidase-a prostate-specific antigen-in prostate cancer cells. The mechanism of this activity involves promotion of the degradation of the androgen receptor. Additional suppressive mechanisms of Dp44mT and DpC on matrix metalloproteases (MMPs) relate to their ability to up-regulate the metastasis suppressors N-myc downstream regulated gene-1 (NDRG1) and NDRG2, which down-regulate MMPs that are crucial for cancer cell invasion.
Collapse
Affiliation(s)
- Kyung Chan Park
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building, University of Sydney, Sydney 2006, Australia; (K.C.P.); (M.D.)
| | - Mahendiran Dharmasivam
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building, University of Sydney, Sydney 2006, Australia; (K.C.P.); (M.D.)
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute of Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Des R. Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building, University of Sydney, Sydney 2006, Australia; (K.C.P.); (M.D.)
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute of Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
5
|
Søreide K, Roalsø M, Aunan JR. Is There a Trojan Horse to Aggressive Pancreatic Cancer Biology? A Review of the Trypsin-PAR2 Axis to Proliferation, Early Invasion, and Metastasis. J Pancreat Cancer 2020; 6:12-20. [PMID: 32064449 PMCID: PMC7014313 DOI: 10.1089/pancan.2019.0014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Purpose: Pancreatic cancer is one of the most lethal of solid tumors and is associated with aggressive cancer biology. The purpose is to review the role of trypsin and effect on molecular and cellular processes potentially explaining the aggressive biology in pancreatic cancer. Methods: A narrative literature review of studies investigating trypsin and its effect on protease systems in cancer, with special reference to pancreatic cancer biology. Results: Proteases, such as trypsin, provides a significant advantage to developing tumors through the ability to remodel the extracellular matrix, promote cell invasion and migration, and facilitate angiogenesis. Trypsin is a digestive enzyme produced by the exocrine pancreas that is also related to mechanisms of proliferation, invasion and metastasis. Several of these mechanisms may be co-regulated or influenced by activation of proteinase-activated receptor 2 (PAR-2). The current role in pancreatic cancer is not clear but emerging data suggest several potential mechanisms. Trypsin may act as a Trojan horse in the pancreatic gland, facilitating several molecular pathways from the onset, which leads to rapid progression of the disease. Pancreatic cancer cell lines containing PAR-2 proliferate upon exposure to trypsin, whereas cancer cell lines not containing PAR-2 fail to proliferate upon trypsin expression. Several mechanisms of action include a proinflammatory environment, signals inducing proliferation and migration, and direct and indirect evidence for mechanisms promoting invasion and metastasis. Novel techniques (such as organoid models) and increased understanding of mechanisms (such as the microbiome) may yield improved understanding into the role of trypsin in pancreatic carcinogenesis. Conclusion: Trypsin is naturally present in the pancreatic gland and may experience pathological activation intracellularly and in the neoplastic environment, which speeds up molecular mechanisms of proliferation, invasion, and metastasis. Further investigation of these processes will provide important insights into how pancreatic cancer evolves, and suggest new ways for treatment.
Collapse
Affiliation(s)
- Kjetil Søreide
- Gastrointestinal Translational Research Unit, Laboratory for Molecular Medicine, Stavanger University Hospital, Stavanger, Norway.,Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway.,Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Marcus Roalsø
- Gastrointestinal Translational Research Unit, Laboratory for Molecular Medicine, Stavanger University Hospital, Stavanger, Norway.,Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway.,Faculty of Health and Medicine, University of Stavanger, Stavanger, Norway
| | - Jan Rune Aunan
- Gastrointestinal Translational Research Unit, Laboratory for Molecular Medicine, Stavanger University Hospital, Stavanger, Norway.,Department of Gastrointestinal Surgery, HPB Unit, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
6
|
Antithrombin controls tumor migration, invasion and angiogenesis by inhibition of enteropeptidase. Sci Rep 2016; 6:27544. [PMID: 27270881 PMCID: PMC4897635 DOI: 10.1038/srep27544] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/17/2016] [Indexed: 12/18/2022] Open
Abstract
Antithrombin is a key inhibitor of the coagulation cascade, but it may also function as an anti-inflammatory, anti-angiogenic, anti-viral and anti-apoptotic protein. Here, we report a novel function of antithrombin as a modulator of tumor cell migration and invasion. Antithrombin inhibited enteropeptidase on the membrane surface of HT-29, A549 and U-87 MG cells. The inhibitory process required the activation of antithrombin by heparin, and the reactive center loop and the heparin binding domain were essential. Surprisingly, antithrombin non-covalently inhibited enteropeptidase, revealing a novel mechanism of inhibition for this serpin. Moreover, as a consequence of this inhibition, antithrombin was cleaved, resulting in a molecule with anti-angiogenic properties that reduced vessel-like formation of endothelial cells. The addition of antithrombin and heparin to U-87 MG and A549 cells reduced motility in wound healing assays, inhibited the invasion in transwell assays and the degradation of a gelatin matrix mediated by invadopodia. These processes were controlled by enteropeptidase, as demonstrated by RNA interference experiments. Carcinoma cell xenografts in nude mice showed in vivo co-localization of enteropeptidase and antithrombin. Finally, treatment with heparin reduced experimental metastasis induced by HT29 cells in vivo. In conclusion, the inhibition of enteropeptidase by antithrombin may have a double anti-tumor effect through inhibiting a protease involved in metastasis and generating an anti-angiogenic molecule.
Collapse
|
7
|
Intracellular Cleavage of the Cx43 C-Terminal Domain by Matrix-Metalloproteases: A Novel Contributor to Inflammation? Mediators Inflamm 2015; 2015:257471. [PMID: 26424967 PMCID: PMC4573893 DOI: 10.1155/2015/257471] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/13/2015] [Indexed: 01/11/2023] Open
Abstract
The coordination of tissue function is mediated by gap junctions (GJs) that enable direct cell-cell transfer of metabolic and electric signals. GJs are formed by connexin (Cx) proteins of which Cx43 is most widespread in the human body. Beyond its role in direct intercellular communication, Cx43 also forms nonjunctional hemichannels (HCs) in the plasma membrane that mediate the release of paracrine signaling molecules in the extracellular environment. Both HC and GJ channel function are regulated by protein-protein interactions and posttranslational modifications that predominantly take place in the C-terminal domain of Cx43. Matrix metalloproteases (MMPs) are a major group of zinc-dependent proteases, known to regulate not only extracellular matrix remodeling, but also processing of intracellular proteins. Together with Cx43 channels, both GJs and HCs, MMPs contribute to acute inflammation and a small number of studies reports on an MMP-Cx43 link. Here, we build further on these reports and present a novel hypothesis that describes proteolytic cleavage of the Cx43 C-terminal domain by MMPs and explores possibilities of how such cleavage events may affect Cx43 channel function. Finally, we set out how aberrant channel function resulting from cleavage can contribute to the acute inflammatory response during tissue injury.
Collapse
|
8
|
Abstract
This review describes studies performed by our group and other laboratories in the field aimed at development of biomarkers not only for cancer but also for other diseases. The markers covered include tumor-associated trypsin inhibitor (TATI), tumor-associated trypsin (TAT), human chorionic gonadotropin (hCG), prostate-specific antigen (PSA) and their various molecular forms, their biology and diagnostic use. The discovery of TATI was the result of a hypothesis-driven project aimed at finding new biomarkers for ovarian cancer among urinary peptides. TATI has since proved to be a useful prognostic marker for several cancers. Recently, it has been named Serine Peptidase Inhibitor Kazal Type 1 (SPINK1) after being rediscovered by several groups as a tumor-associated peptide by gene expression profiling and proteomic techniques and shown to promote tumor development by stimulating the EGF receptor. To explain why a trypsin inhibitor is strongly expressed in some cancers, research focused on the protease that it inhibited led to the finding of tumor-associated trypsin (TAT). Elevated serum concentrations of TAT-2 were found in some cancer types, but fairly high background levels of pancreatic trypsinogen-2 limited the use of TAT-2 for cancer diagnostics. However, trypsinogen-2 and its complex with α1-protease inhibitor proved to be very sensitive and specific markers for pancreatitis. Studies on hCG were initiated by the need to develop more rapid and sensitive pregnancy tests. These studies showed that serum from men and non-pregnant women contains measurable concentrations of hCG derived from the pituitary. Subsequent development of assays for the subunits of hCG showed that the β subunit of hCG (hCGβ) is expressed at low concentrations by most cancers and that it is a strong prognostic marker. These studies led to the formation of a working group for standardization of hCG determinations and the development of new reference reagents for several molecular forms of hCG. The preparation of intact hCG has been adopted as the fifth international standard by WHO. Availability of several well-defined forms of hCG made it possible to characterize the epitopes of nearly 100 monoclonal antibodies. This will facilitate design of immunoassays with pre-defined specificity. Finally, the discovery of different forms of immunoreactive PSA in serum from a prostate cancer patient led to identification of the complex between PSA and α1-antichymotrypsin, and the use of assays for free and total PSA in serum for improved diagnosis of prostate cancer. Epitope mapping of PSA antibodies and establishment of PSA standards has facilitated establishment well-standardized assays for the various forms of PSA.
Collapse
Affiliation(s)
- Ulf-Håkan Stenman
- a Department of Clinical Chemistry , Biomedicum, Helsinki University and Helsinki University Central Hospital (HUCH) , Helsinki , Finland
| |
Collapse
|
9
|
Mehrian-Shai R, Freedman S, Shams S, Doherty J, Slattery W, Hsu NYH, Reichardt JKV, Andalibi A, Toren A. Schwannomas exhibit distinct size-dependent gene-expression patterns. Future Oncol 2015; 11:1751-8. [PMID: 26075443 DOI: 10.2217/fon.15.72] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Neurofibromatosis type 2 (NF2)-associated vestibular schwannomas have variable size at presentation which presents a unique challenge in NF2 patient management. Therefore, we investigated the molecular signature characteristic of the differences in size for improved individualized precise therapy. MATERIALS & METHODS RNA expression analysis was performed on 15 small and 27 large NF2-associated vestibular schwannoma tumors using a microarray analyzing over 47,000 transcripts. RESULTS A signature of 11 genes was found to be correlated with NF2 tumor size. CONCLUSION We have identified the genetic hallmark that differentiates large NF2-associated tumors from smaller tumors. This is the first time that these genes have been shown to be the hallmark for NF2 tumor size.
Collapse
Affiliation(s)
- Ruty Mehrian-Shai
- Department of Pediatric Hemato-Oncology, The Cancer Research Center, Sheba Medical Center, 2 Sheba Road, Ramat Gan, 52621, Israel
| | - Shany Freedman
- Department of Pediatric Hemato-Oncology, The Cancer Research Center, Sheba Medical Center, 2 Sheba Road, Ramat Gan, 52621, Israel
| | - Soheil Shams
- BioDiscovery, 5155 W Rosecrans Ave # 310, Hawthorne, CA 90250, USA
| | - Joni Doherty
- Head & Neck Surgery, University of California, San Diego School of Medicine, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - William Slattery
- Department of Clinical Studies, House Ear Institute, 2100 W 3rd St #500, Los Angeles, CA 90057, USA
| | | | - Juergen K V Reichardt
- Division of Tropical Health & Medicine, James Cook University, Townsville, QLD, Australia
| | - Ali Andalibi
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Amos Toren
- Department of Pediatric Hemato-Oncology, The Cancer Research Center, Sheba Medical Center, 2 Sheba Road, Ramat Gan, 52621, Israel
| |
Collapse
|
10
|
Lin J, Zhao D, Wang J, Wang Y, Li H, Yin X, Yang L, Zhou X. Transcriptome changes upon in vitro challenge with Mycobacterium bovis in monocyte-derived macrophages from bovine tuberculosis-infected and healthy cows. Vet Immunol Immunopathol 2014; 163:146-56. [PMID: 25550244 DOI: 10.1016/j.vetimm.2014.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 11/19/2014] [Accepted: 12/05/2014] [Indexed: 02/06/2023]
Abstract
As innate immune cells, macrophages are expected to respond to mycobacterial infection equally in both Mycobacterium bovis-infected cows and healthy cows. We previously found that monocyte-derived macrophages (MDMs) from M. bovis-infected cows respond differently than MDMs from healthy cows when exposed to in vitro M. bovis challenge. We have now used the Agilent™ Bovine Gene Expression Microarray to examine transcriptional differences between these MDMs. At a high multiplicity of infection (10), in vitro challenge led to changes in several thousands of genes, with dysregulation at multiple orders of magnitude. For example, significant changes were seen for colony stimulating factor 3 (granulocyte) (CSF3), colony stimulating factor 2 (granulocyte-macrophage) (CSF2), and chemokine (C-C motif) ligand 20 (CCL20). Classical macrophage activation was also observed, although to a lesser degree in interleukin 12 (IL12) expression. For macrophages, kallikrein-related peptidase 12 (KLK12) and protease, serine, 2 (trypsin 2) (PRSS2), as well as a secreted protein, acidic, cysteine-rich (osteonectin) (SPARC)-centered matricellular gene network, were differentially expressed in infected animals. Finally, global transcriptome fold-changes caused by in vitro challenge were higher in healthy cows than in tuberculosis-positive cows, suggesting that healthy macrophages responded marginally better to in vitro infection. Macrophages from healthy and already infected animals can both be fully activated during M. bovis infection, yet there are differences between these macrophages: distinct expression pattern in matricellular proteins, and their different responses to in vitro infection.
Collapse
Affiliation(s)
- Jingjun Lin
- The State Key Lab of Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National TSE Lab, College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Deming Zhao
- The State Key Lab of Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National TSE Lab, College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Jin Wang
- The State Key Lab of Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National TSE Lab, College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Yang Wang
- Institut de Génétique et Microbiologie, Université Paris-Sud 11, 91405 Orsay, France
| | - Hua Li
- The State Key Lab of Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National TSE Lab, College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Xiaomin Yin
- The State Key Lab of Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National TSE Lab, College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Lifeng Yang
- The State Key Lab of Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National TSE Lab, College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Xiangmei Zhou
- The State Key Lab of Agrobiotechnology, Key Lab of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National TSE Lab, College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing 100193, China.
| |
Collapse
|
11
|
Rakashanda S, Qazi AK, Majeed R, Andrabi SM, Hamid A, Sharma PR, Amin S. Plant-derived protease inhibitors LC-pi (Lavatera cashmeriana) inhibit human lung cancer cell proliferation in vitro. Nutr Cancer 2014; 67:156-66. [PMID: 25412192 DOI: 10.1080/01635581.2015.967876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The objective of this study was to check the anticancer activity of purified protease inhibitors of Lavatera cashmeriana viz LC-pi I, II, III, and IV (Lavatera cashmeriana protease inhibitors) on A549 (lung) cell. It was found that LC-pi I and II significantly inhibited the proliferation of A549 cells with IC₅₀ value of 54 μg/ml and 38 μg/ml, respectively, whereas inhibition by LC-pi III and IV was negligible. LC-pi I and II were further found to inhibit formation of colonies in a dose-dependent manner. Also, both inhibitors were found to induce apoptosis causing chromatin condensation and DNA fragmentation, without loss of mitochondrial membrane potential. Cell cycle revealed a significant increase of subG₀/G₁ phase cells that are apoptotic cells. We also demonstrated a dose-dependent decrease in migration of A549 cells on cell migration assay by both inhibitors. Taken together, we demonstrate that LC-pi I and II inhibited proliferation through arresting cells before apoptosis, inducing apoptosis and inhibiting cell migration in human lung cancer cells, but the study warrants further investigation. Our results support the notion that plant protease inhibitors may have the potential to advance as chemopreventive agents.
Collapse
Affiliation(s)
- Syed Rakashanda
- a Department of Biochemistry , The University of Kashmir , Srinagar , India
| | | | | | | | | | | | | |
Collapse
|
12
|
Pessoa JIC, Guimarães GN, Viola NV, da Silva WJ, de Souza AP, Tjäderhane L, Line SR, Marques MR. In situ study of the gelatinase activity in demineralized dentin from rat molar teeth. Acta Histochem 2013; 115:245-51. [PMID: 22897943 DOI: 10.1016/j.acthis.2012.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Revised: 07/10/2012] [Accepted: 07/11/2012] [Indexed: 11/29/2022]
Abstract
Matrix metalloproteinases (MMPs) in dentin are believed to participate in various physiological and pathological events in coronal dentin, but their exact source and location is not clear. The purpose of this study was to evaluate the activity of gelatinases in decalcified rat molars crowns by in situ zymography. Hemi-mandibles of five male Wistar rats were fixed in paraformaldehyde, decalcified in EDTA and glycerol solution and embedded in paraffin. Sections from the region of molar teeth were incubated with or without DQ gelatin in 50mM Tris-CaCl2 at 37°C for 2h and observed by means of confocal microscopy. Gelatinolytic activity was observed throughout the coronal dentin with varying intensities in different locations. High gelatinase activity was observed in the dentinal tubules, dentin-enamel junction (DEJ) and predentin, and it was weaker and less uniform in the intertubular dentin. This study shows that the location of gelatinase and relative activity can be detected by means of in situ zymography and confocal microcopy, and this methodology may provide a useful tool in studies on the role of gelatinases in tooth development, maturation and in pathological conditions.
Collapse
Affiliation(s)
- Juliana Isabelita Cyrino Pessoa
- Department of Morphology, Division of Histology, Piracicaba Dental School, State University of Campinas, Piracicaba, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Solli AI, Fadnes B, Winberg JO, Uhlin-Hansen L, Hadler-Olsen E. Tissue- and cell-specific co-localization of intracellular gelatinolytic activity and matrix metalloproteinase 2. J Histochem Cytochem 2013; 61:444-61. [PMID: 23482328 DOI: 10.1369/0022155413484765] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Matrix metalloproteinase 2 (MMP-2) is a proteolytic enzyme that degrades extracellular matrix proteins. Recent studies indicate that MMP-2 also has a role in intracellular proteolysis during various pathological conditions, such as ischemic injuries in heart and brain and in tumor growth. The present study was performed to map the distribution of intracellular MMP-2 activity in various mouse tissues and cells under physiological conditions. Samples from normal brain, heart, lung, liver, spleen, pancreas, kidney, adrenal gland, thyroid gland, gonads, oral mucosa, salivary glands, esophagus, intestines, and skin were subjected to high-resolution in situ gelatin zymography and immunohistochemical staining. In hepatocytes, cardiac myocytes, kidney tubuli cells, epithelial cells in the oral mucosa as well as in excretory ducts of salivary glands, and adrenal cortical cells, we found strong intracellular gelatinolytic activity that was significantly reduced by the metalloprotease inhibitor EDTA but not by the cysteine protease inhibitor E-64. Furthermore, the gelatinolytic activity was co-localized with MMP-2. Western blotting and electron microscopy combined with immunogold labeling revealed the presence of MMP-2 in different intracellular compartments of isolated hepatocytes. Our results indicate that MMP-2 takes part in intracellular proteolysis in specific tissues and cells during physiological conditions.
Collapse
Affiliation(s)
- Ann Iren Solli
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| | | | | | | | | |
Collapse
|
14
|
Fluctuating roles of matrix metalloproteinase-9 in oral squamous cell carcinoma. ScientificWorldJournal 2013; 2013:920595. [PMID: 23365550 PMCID: PMC3556887 DOI: 10.1155/2013/920595] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 12/10/2012] [Indexed: 12/12/2022] Open
Abstract
One hallmark of cancer is the degradation of the extracellular matrix (ECM), which is caused by proteinases. In oral cancers, matrix metalloproteinases (MMPs), especially MMP-9, are associated with this degradation. MMPs break down the ECM allowing cancer to spread; they also release various factors from their cryptic sites, including cytokines. These factors modulate cell behavior and enhance cancer progression by regulating angiogenesis, migration, proliferation, and invasion. The development of early metastases is typical for oral cancer, and increased MMP-9 expression is associated with a poor disease prognosis. However, many studies fail to relate MMP-9 expression with metastasis formation. Contrary to earlier models, recent studies show that MMP-9 plays a protective role in oral cancers. Therefore, the role of MMP-9 is complicated and may fluctuate throughout the different types and stages of oral cancers.
Collapse
|
15
|
Han SJ, Hawkins SM, Begum K, Jung SY, Kovanci E, Qin J, Lydon JP, DeMayo FJ, O'Malley BW. A new isoform of steroid receptor coactivator-1 is crucial for pathogenic progression of endometriosis. Nat Med 2012; 18:1102-11. [PMID: 22660634 DOI: 10.1038/nm.2826] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 05/03/2012] [Indexed: 01/07/2023]
Abstract
Endometriosis is considered to be an estrogen-dependent inflammatory disease, but its etiology is unclear. Thus far, a mechanistic role for steroid receptor coactivators (SRCs) in the progression of endometriosis has not been elucidated. An SRC-1-null mouse model reveals that the mouse SRC-1 gene has an essential role in endometriosis progression. Notably, a previously unidentified 70-kDa SRC-1 proteolytic isoform is highly elevated both in the endometriotic tissue of mice with surgically induced endometriosis and in endometriotic stromal cells biopsied from patients with endometriosis compared to normal endometrium. Tnf⁻/⁻ and Mmp9⁻/⁻ mice with surgically induced endometriosis showed that activation of tumor necrosis factor a (TNF-α)-induced matrix metallopeptidase 9 (MMP9) activity mediates formation of the 70-kDa SRC-1 C-terminal isoform in endometriotic mouse tissue. In contrast to full-length SRC-1, the endometriotic 70-kDa SRC-1 C-terminal fragment prevents TNF-α-mediated apoptosis in human endometrial epithelial cells and causes the epithelial-mesenchymal transition and the invasion of human endometrial cells that are hallmarks of progressive endometriosis. Collectively, the newly identified TNF-α-MMP9-SRC-1 isoform functional axis promotes pathogenic progression of endometriosis.
Collapse
Affiliation(s)
- Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Vilen ST, Suojanen J, Salas F, Risteli J, Ylipalosaari M, Itkonen O, Koistinen H, Baumann M, Stenman UH, Sorsa T, Salo T, Nyberg P. Trypsin-2 enhances carcinoma invasion by processing tight junctions and activating ProMT1-MMP. Cancer Invest 2012; 30:583-92. [PMID: 22909050 DOI: 10.3109/07357907.2012.716467] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Enhanced proteolysis and altered tight junction (TJ) proteins associate with carcinoma invasion. We hypothesized that trypsin-2, a tumor-associated serine proteinase, induces tongue carcinoma invasion by activating pro-membrane type-1 matrix metalloproteinase (MT1-MMP) and disturbing the TJs. The effects of invasion were analyzed using trypsin-2 over-expressing human tongue squamous cell carcinoma cells (Try2-HSC-3) in vitro and in vivo. The invasion of Try2-HSC-3 cells was increased in mouse xenografts and human organotypic model. Trypsin-2 activated proMT1-MMP, as well as altered the expression of TJ protein claudin-7. In conclusion, trypsin-2 over-expression enhanced tongue carcinoma cell invasion by various genetic and proteolytic mechanisms.
Collapse
Affiliation(s)
- Suvi-Tuuli Vilen
- Institute of Dentistry, University of Helsinki, and Department of Oral and Maxillofacial Diseases, Helsinki University Hospital, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Prohaska TA, Wahlmüller FC, Furtmüller M, Geiger M. Interaction of protein C inhibitor with the type II transmembrane serine protease enteropeptidase. PLoS One 2012; 7:e39262. [PMID: 22723979 PMCID: PMC3378520 DOI: 10.1371/journal.pone.0039262] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 05/22/2012] [Indexed: 11/18/2022] Open
Abstract
The serine protease inhibitor protein C inhibitor (PCI) is expressed in many human tissues and exhibits broad protease reactivity. PCI binds glycosaminoglycans and certain phospholipids, which modulate its inhibitory activity. Enteropeptidase (EP) is a type II transmembrane serine protease mainly found on the brush border membrane of epithelial cells in the duodenum, where it activates trypsinogen to initiate the digestion of food proteins. Some active EP is also present in duodenal fluid and has been made responsible for causing pancreatitis in case of duodeno-pancreatic reflux. Together with its substrate trypsinogen, EP is furthermore present in the epidermis and in some cancer cells. In this report, we show that PCI inhibited EP with an apparent 2nd order rate constant of 4.48 × 10(4) M(-1) s(-1). Low molecular weight (LMWH) and unfractionated heparin (UFH) slightly reduced the inhibitory effect of PCI. The SI (stoichiometry of inhibition) value for the inhibition of EP by PCI was 10.8 in the absence and 17.9 in the presence of UFH (10 U/ml). By inhibiting trypsin, chymotrypsin, and additionally EP, PCI might play a role in the protection of the pancreas from autodigestion. Furthermore the interaction of PCI with EP may influence the regulation of epithelial differentiation.
Collapse
Affiliation(s)
- Thomas A. Prohaska
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Felix C. Wahlmüller
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Margareta Furtmüller
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Margarethe Geiger
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
18
|
Ostapchenko VG, Gasparian ME, Kosinsky YA, Efremov RG, Dolgikh DA, Kirpichnikov MP. Dissecting structural basis of the unique substrate selectivity of human enteropeptidase catalytic subunit. J Biomol Struct Dyn 2012; 30:62-73. [DOI: 10.1080/07391102.2012.674249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
19
|
Characterization of dsRNA-induced pancreatitis model reveals the regulatory role of IFN regulatory factor 2 (Irf2) in trypsinogen5 gene transcription. Proc Natl Acad Sci U S A 2011; 108:18766-71. [PMID: 22042864 DOI: 10.1073/pnas.1116273108] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mice deficient for interferon regulatory factor (Irf)2 (Irf2(-/-) mice) exhibit immunological abnormalities and cannot survive lymphocytic choriomeningitis virus infection. The pancreas of these animals is highly inflamed, a phenotype replicated by treatment with poly(I:C), a synthetic double-stranded RNA. Trypsinogen5 mRNA was constitutively up-regulated about 1,000-fold in Irf2(-/-) mice compared with controls as assessed by quantitative RT-PCR. Further knockout of IFNα/β receptor 1(Ifnar1) abolished poly(I:C)-induced pancreatitis but had no effect on the constitutive up-regulation of trypsinogen5 gene, indicating crucial type I IFN signaling to elicit the inflammation. Analysis of Ifnar1(-/-) mice confirmed type I IFN-dependent transcriptional activation of dsRNA-sensing pattern recognition receptor genes MDA5, RIG-I, and TLR3, which induced poly(I:C)-dependent cell death in acinar cells in the absence of IRF2. We speculate that Trypsin5, the trypsinogen5 gene product, leaking from dead acinar cells triggers a chain reaction leading to lethal pancreatitis in Irf2(-/-) mice because it is resistant to a major endogenous trypsin inhibitor, Spink3.
Collapse
|
20
|
Petzold A, Tisdall MM, Girbes AR, Martinian L, Thom M, Kitchen N, Smith M. In vivo monitoring of neuronal loss in traumatic brain injury: a microdialysis study. ACTA ACUST UNITED AC 2011; 134:464-83. [PMID: 21278408 PMCID: PMC3030768 DOI: 10.1093/brain/awq360] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Traumatic brain injury causes diffuse axonal injury and loss of cortical neurons. These features are well recognized histologically, but their in vivo monitoring remains challenging. In vivo cortical microdialysis samples the extracellular fluid adjacent to neurons and axons. Here, we describe a novel neuronal proteolytic pathway and demonstrate the exclusive neuro-axonal expression of Pavlov’s enterokinase. Enterokinase is membrane bound and cleaves the neurofilament heavy chain at positions 476 and 986. Using a 100 kDa microdialysis cut-off membrane the two proteolytic breakdown products, extracellular fluid neurofilament heavy chains NfH476−986 and NfH476−1026, can be quantified with a relative recovery of 20%. In a prospective clinical in vivo study, we included 10 patients with traumatic brain injury with a median Glasgow Coma Score of 9, providing 640 cortical extracellular fluid samples for longitudinal data analysis. Following high-velocity impact traumatic brain injury, microdialysate extracellular fluid neurofilament heavy chain levels were significantly higher (6.18 ± 2.94 ng/ml) and detectable for longer (>4 days) compared with traumatic brain injury secondary to falls (0.84 ± 1.77 ng/ml, <2 days). During the initial 16 h following traumatic brain injury, strong correlations were found between extracellular fluid neurofilament heavy chain levels and physiological parameters (systemic blood pressure, anaerobic cerebral metabolism, excessive brain tissue oxygenation, elevated brain temperature). Finally, extracellular fluid neurofilament heavy chain levels were of prognostic value, predicting mortality with an odds ratio of 7.68 (confidence interval 2.15–27.46, P = 0.001). In conclusion, this study describes the discovery of Pavlov’s enterokinase in the human brain, a novel neuronal proteolytic pathway that gives rise to specific protein biomarkers (NfH476−986 and NfH476−1026) applicable to in vivo monitoring of diffuse axonal injury and neuronal loss in traumatic brain injury.
Collapse
Affiliation(s)
- Axel Petzold
- Department of Neuroimmunology, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK.
| | | | | | | | | | | | | |
Collapse
|
21
|
Hadler-Olsen E, Wetting HL, Ravuri C, Omair A, Rikardsen O, Svineng G, Kanapathippillai P, Winberg JO, Uhlin-Hansen L. Organ specific regulation of tumour invasiveness and gelatinolytic activity at the invasive front. Eur J Cancer 2011; 47:305-15. [DOI: 10.1016/j.ejca.2010.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Revised: 08/19/2010] [Accepted: 09/02/2010] [Indexed: 10/19/2022]
|
22
|
Hadler-Olsen E, Fadnes B, Sylte I, Uhlin-Hansen L, Winberg JO. Regulation of matrix metalloproteinase activity in health and disease. FEBS J 2010; 278:28-45. [PMID: 21087458 DOI: 10.1111/j.1742-4658.2010.07920.x] [Citation(s) in RCA: 271] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The activity of matrix metalloproteinases (MMPs) is regulated at several levels, including enzyme activation, inhibition, complex formation and compartmentalization. Regulation at the transcriptional level is also important, although this is not a subject of the present minireview. Most MMPs are secreted and have their function in the extracellular environment. This is also the case for the membrane-type MMPs (MT-MMPs). MMPs are also found inside cells, both in the nucleus, cytosol and organelles. The role of intracellular located MMPs is still poorly understood, although recent studies have unraveled some of their functions. The localization, activation and activity of MMPs are regulated by their interactions with other proteins, proteoglycan core proteins and/or their glycosaminoglycan chains, as well as other molecules. Complexes formed between MMPs and various molecules may also include interactions with noncatalytic sites. Such exosites are regions involved in substrate processing, localized outside the active site, and are potential binding sites of specific MMP inhibitors. Knowledge about regulation of MMP activity is essential for understanding various physiological processes and pathogenesis of diseases, as well as for the development of new MMP targeting drugs.
Collapse
Affiliation(s)
- Elin Hadler-Olsen
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Norway
| | | | | | | | | |
Collapse
|
23
|
Fracalossi ACC, Miranda SR, Oshima CTF, Franco M, Ribeiro DA. The role of matrix metalloproteinases 2 and 9 during rat tongue carcinogenesis induced by 4-nitroquinoline 1-oxide. J Mol Histol 2010; 41:19-25. [DOI: 10.1007/s10735-010-9258-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Accepted: 02/16/2010] [Indexed: 12/19/2022]
|
24
|
Sterz CM, Kulle C, Dakic B, Makarova G, Böttcher MC, Bette M, Werner JA, Mandic R. A basal-cell-like compartment in head and neck squamous cell carcinomas represents the invasive front of the tumor and is expressing MMP-9. Oral Oncol 2009; 46:116-22. [PMID: 20036607 DOI: 10.1016/j.oraloncology.2009.11.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 11/25/2009] [Accepted: 11/25/2009] [Indexed: 01/06/2023]
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are the most frequent malignancies of the upper aerodigestive tract. The cancer stem cell (CSC) hypothesis concludes that CSCs constitute the dangerous tumor cell population due to their ability of self-renewal and being associated with relapse of tumor disease, invasiveness and resistance to chemo(radio)therapy. The aim of this study was to look for CSC candidates and expression of MMP-9 that previously was implicated in HNSCC invasiveness. Immunohistochemical, immunofluorescence and Western blot analysis were performed on HNSCC tumor specimens using antibodies specific for MMP-9, CD44, ALDH1 and CK14. Gelatinolytic activity was assessed by zymography. Pearson correlation analysis was used for statistical comparison. Immunohistochemical analysis found CD44 and MMP-9 to co-localize in tumor cells at the invasive front. Western blot analysis demonstrated a significant correlation (p=0.0047) between CD44 and MMP-9 in the tested tissues. In addition gelatinolytic activity of HNSCC tissues was found to significantly correlate (p=0.0010) with MMP-9 expression. The CD44(+) invasive front of the tumor was also positive for ALDH1 and CK14, all of them being typically expressed by cells in the basal cell layer of normal stratified squamous epithelia that also harbors the epithelial stem cells. The observations point to a role of a MMP-9 positive basal-cell-like cell layer in the process of HNSCC invasiveness. This compartment likely contains CSCs since it is expressing the putative CSC markers CD44, ALDH1 and CK14. This cell layer therefore should be considered a major therapeutic target in the treatment of head and neck cancer.
Collapse
Affiliation(s)
- Carolina M Sterz
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Giessen and Marburg, Campus Marburg, Deutschhausstrasse 3, D-35037 Marburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|