1
|
Tanwar S, Zheng P, Wu L, Barman I. Stimuli-responsive 'On-Off' SERS-darkfield bimodal plasmonic nanoprobes for selective cancer cell illumination. Biosens Bioelectron 2025; 286:117615. [PMID: 40449262 DOI: 10.1016/j.bios.2025.117615] [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: 03/24/2025] [Revised: 04/29/2025] [Accepted: 05/20/2025] [Indexed: 06/03/2025]
Abstract
Early detection is essential for improving cancer treatment outcomes. Surface-enhanced Raman scattering (SERS) offers high sensitivity and molecular specificity but suffers from slow imaging speed when used independently. Here, we introduce stimuli-responsive bimodal SERS-dark-field (DF) nanoprobes for rapid and selective imaging of metastatic prostate cancer cells. These nanoprobes utilize legumain enzyme-responsive peptide coatings on plasmonic gold nanocubes and a bio-orthogonal self-condensation mechanism to facilitate enzyme-triggered nanoprobe aggregation. Upon internalization, the nanoprobes selectively aggregate in DU145 cells that overexpress legumain, while remaining uniformly dispersed in LNCaP cells with minimal legumain activity. DF microscopy quickly identifies these aggregates, significantly expediting subsequent SERS imaging. The engineered design of these nanoprobes ensures distinct vibrational signatures specifically correlated to enzyme activity, establishing a direct link between enzymatic presence and SERS signal generation. By combining the rapid localization capabilities of DF imaging with the molecular precision of SERS, our bimodal imaging strategy significantly surpasses conventional single-modal SERS techniques, providing a versatile platform for fast and accurate detection of various diseases through their distinctive enzymatic profiles.
Collapse
Affiliation(s)
- Swati Tanwar
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Peng Zheng
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lintong Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ishan Barman
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA.
| |
Collapse
|
2
|
Yang KF, Zhang JY, Feng M, Yao K, Liu YY, Zhou MS, Jia H. Secretase promotes AD progression: simultaneously cleave Notch and APP. Front Aging Neurosci 2024; 16:1445470. [PMID: 39634655 PMCID: PMC11615878 DOI: 10.3389/fnagi.2024.1445470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Alzheimer's disease (AD) involves complex pathological mechanisms. Secretases include membrane protein extracellular structural domain proteases and intramembrane proteases that cleave the topology to type I or type II. Secretases can effectively regulate the activation of Notch and amyloid precursor protein (APP), key factors in the progression of AD and cancer. This article systematically summarizes the intracellular localization, cleavage sites and products, and biological functions of six subtypes of secretases (α-secretase, β-secretase, γ-secretase, δ-secretase, ε-secretase, and η-secretase), and for the first time, elucidates the commonalities and differences between these subtypes of secretases. We found that each subtype of secretase primarily cleaves APP and Notch as substrates, regulating Aβ levels through APP cleavage to impact the progression of AD, while also cleaving Notch receptors to affect cancer progression. Finally, we review the chemical structures, indications, and research stages of various secretase inhibitors, emphasizing the promising development of secretase inhibitors in the fields of cancer and AD.
Collapse
Affiliation(s)
- Ke-Fan Yang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Shenyang Medical College, Shenyang, China
| | - Jing-Yi Zhang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Mei Feng
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Shenyang Medical College, Shenyang, China
| | - Kuo Yao
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Shenyang Medical College, Shenyang, China
| | - Yue-Yang Liu
- Science and Experimental Research Center of Shenyang Medical College, Shenyang, Liaoning, China
| | - Ming-Sheng Zhou
- Science and Experimental Research Center of Shenyang Medical College, Shenyang, Liaoning, China
| | - Hui Jia
- Science and Experimental Research Center of Shenyang Medical College, Shenyang, Liaoning, China
- School of Traditional Chinese Medicine, Shenyang Medical College, Shenyang, Liaoning, China
| |
Collapse
|
3
|
Lustenberger SK, Castro Jaramillo CA, Bärtschi LA, Williams R, Schibli R, Mu L, Krämer SD. Towards imaging the immune state of cancer by PET: Targeting legumain with 11C-labeled P1-Asn peptidomimetics carrying a cyano-warhead. Nucl Med Biol 2024; 138-139:108951. [PMID: 39303441 DOI: 10.1016/j.nucmedbio.2024.108951] [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: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE M2-type tumor-associated macrophages (TAM) residing in the tumor microenvironment (TME) have been linked to tumor invasiveness, metastasis and poor prognosis. M2 TAMs suppress T cell activation, silencing the recognition of the cancer by the immune system. Targeting TAMs in anti-cancer therapy may support the immune system and immune-checkpoint inhibitor therapies to fight the cancer cells. We aimed to develop a PET tracer for the imaging of M2 TAM infiltration of cancer, using activated legumain as the imaging target. BASIC PROCEDURES Two P1-mimicking inhibitors with a cyano-warhead were labeled with carbon-11 and evaluated in vitro and in vivo with a CT26 tumor mouse model. Target expression and activity were quantified from RT-qPCR and in vitro substrate conversion, respectively. The co-localization of legumain and TAMs was assessed by fluorescence microscopy. The two tracers were evaluated by PET with subsequent biodistribution analysis with the dissected tissues. Parent-to-total radioactivity in plasma was determined at several time points after i.v. tracer injection, using reverse phase radio-UPLC. MAIN FINDINGS Legumain displayed a target density of 40.7 ± 19.1 pmol per mg total protein in tumor lysate (n = 4) with high substrate conversion and colocalization with M2 macrophages in the tumor periphery. [11C]1 and [11C]2 were synthesized with >95 % radiochemical purity and 12.9-382.2 GBq/μmol molar activity at the end of synthesis. We observed heterogeneous tumor accumulation in in vitro autoradiography and PET for both tracers. However, excess unlabeled 1 or 2 did not compete with tracer accumulation. Both [11C]1 and [11C]2 were rapidly metabolized to a polar radiometabolite in vivo. PRINCIPAL CONCLUSIONS The legumain tracers [11C]1 and [11C]2, synthesized with high radiochemical purity and molar activity, accumulate in the legumain-positive CT26 tumor in vivo. However, the lack of competition by excess compound questions their specificity. Both tracers are rapidly metabolized in vivo, requiring structural modifications towards more stable tracers for further investigations.
Collapse
Affiliation(s)
- Severin K Lustenberger
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Claudia A Castro Jaramillo
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Lena A Bärtschi
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Rich Williams
- Queens University Belfast, BT7 1NN Belfast, United Kingdom of Great Britain and Northern Ireland
| | - Roger Schibli
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Linjing Mu
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Stefanie D Krämer
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland.
| |
Collapse
|
4
|
Wu L, Tanwar S, Kaur G, Date S, Goel L, Chatterjee A, McGuiggan P, Barman I. DNA Origami-Engineered Plasmonic Nanoprobes for Targeted Cancer Imaging. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2309929. [PMID: 39131199 PMCID: PMC11309351 DOI: 10.1002/adfm.202309929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Indexed: 08/13/2024]
Abstract
Plasmonic nanomaterials bearing targeting ligands are of great interest for surface-enhanced Raman scattering (SERS)-based bioimaging applications. However, the practical utility of SERS-based imaging strategies has been hindered by the lack of a straightforward method to synthesize highly sensitive SERS-active nanostructures with high yield and efficiency. In this work, leveraging DNA origami principles, we report the first-in-class design of a SERS-based plasmonically coupled nanoprobe for targeted cancer imaging (SPECTRA). The nanoprobe harnesses a cancer cell targeting DNA aptamer sequence and vibrational tag with stretching frequency in the cell-silent Raman window. Through the integration of aptamer sequence specific for DU145 cells, we show the unique capabilities of SPECTRA for targeted imaging of DU145 cells. Our results demonstrate that the scalability, cost-effectiveness, and reproducibility of this method of fabrication of SERS nanoprobes can serve as a versatile platform for creating nanoprobes with broad applications in the fields of cancer biology and biomedical imaging.
Collapse
Affiliation(s)
- Lintong Wu
- Department of Mechanical Engineering, Johns Hopkins University, Maryland 21218, USA
| | - Swati Tanwar
- Department of Mechanical Engineering, Johns Hopkins University, Maryland 21218, USA
| | - Gagandeep Kaur
- Department of Mechanical Engineering, Johns Hopkins University, Maryland 21218, USA
| | - Siddhi Date
- Department of Biomedical Engineering, Johns Hopkins University, Maryland 21218, USA
| | - Linika Goel
- Department of Biomedical Engineering, Johns Hopkins University, Maryland 21218, USA
| | - Arnab Chatterjee
- Department of Mechanical Engineering, Johns Hopkins University, Maryland 21218, USA
| | - Patty McGuiggan
- Department of Material Science and Engineering, Johns Hopkins University, Maryland 21218, USA
- Department of Chemistry, Johns Hopkins University, Maryland 21218, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Maryland 21218, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Maryland 21205, USA
- Department of Oncology, Johns Hopkins University, Maryland 21231, USA
| |
Collapse
|
5
|
Xu H, Xu D, Zheng Y, Wang H, Li A, Zheng X. Investigation of prognostic values of immune infiltration and LGMN expression in the microenvironment of osteosarcoma. Discov Oncol 2024; 15:275. [PMID: 38980440 PMCID: PMC11233489 DOI: 10.1007/s12672-024-01123-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 06/25/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Osteosarcoma (OS), the most common primary malignant bone tumor, predominantly affects children and young adults and is characterized by high invasiveness and poor prognosis. Despite therapeutic advancements, the survival rate remains suboptimal, indicating an urgent need for novel biomarkers and therapeutic targets. This study aimed to investigate the prognostic significance of LGMN expression and immune cell infiltration in the tumor microenvironment of OS. METHODS We performed an integrative bioinformatics analysis utilizing the GEO and TARGET-OS databases to identify differentially expressed genes (DEGs) associated with LGMN in OS. We conducted Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) to explore the biological pathways and functions. Additionally, we constructed protein-protein interaction (PPI) networks, a competing endogenous RNA (ceRNA) network, and applied the CIBERSORT algorithm to quantify immune cell infiltration. The diagnostic and prognostic values of LGMN were evaluated using the area under the receiver operating characteristic (ROC) curve and Cox regression analysis. Furthermore, we employed Consensus Clustering Analysis to explore the heterogeneity within OS samples based on LGMN expression. RESULTS The analysis revealed significant upregulation of LGMN in OS tissues. DEGs were enriched in immune response and antigen processing pathways, suggesting LGMN's role in immune modulation within the TME. The PPI and ceRNA network analyses provided insights into the regulatory mechanisms involving LGMN. Immune cell infiltration analysis indicated a correlation between high LGMN expression and increased abundance of M2 macrophages, implicating an immunosuppressive role. The diagnostic AUC for LGMN was 0.799, demonstrating its potential as a diagnostic biomarker. High LGMN expression correlated with reduced overall survival (OS) and progression-free survival (PFS). Importantly, Consensus Clustering Analysis identified two distinct subtypes of OS, highlighting the heterogeneity and potential for personalized medicine approaches. CONCLUSIONS Our study underscores the prognostic value of LGMN in osteosarcoma and its potential as a therapeutic target. The identification of LGMN-associated immune cell subsets and the discovery of distinct OS subtypes through Consensus Clustering Analysis provide new avenues for understanding the immunosuppressive TME of OS and may aid in the development of personalized treatment strategies. Further validation in larger cohorts is warranted to confirm these findings.
Collapse
Affiliation(s)
- Hualiang Xu
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, No. 396, Tongfu Middle Road, Haizhu District, Guangzhou, Guangdong, People's Republic of China
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, People's Republic of China
| | - Dawei Xu
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, No. 396, Tongfu Middle Road, Haizhu District, Guangzhou, Guangdong, People's Republic of China
| | - Yinfeng Zheng
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, No. 396, Tongfu Middle Road, Haizhu District, Guangzhou, Guangdong, People's Republic of China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, People's Republic of China
| | - Aiguo Li
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, No. 396, Tongfu Middle Road, Haizhu District, Guangzhou, Guangdong, People's Republic of China.
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, Jinan University, No. 613, Huangpu Avenue West, Tianhe District, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
6
|
Wang Y, Peng Y, Zi G, Chen J, Peng B. Co-delivery of Cas9 mRNA and guide RNAs for editing of LGMN gene represses breast cancer cell metastasis. Sci Rep 2024; 14:8095. [PMID: 38582932 PMCID: PMC10998893 DOI: 10.1038/s41598-024-58765-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 04/03/2024] [Indexed: 04/08/2024] Open
Abstract
Legumain (or asparagine endopeptidase/AEP) is a lysosomal cysteine endopeptidase associated with increased invasive and migratory behavior in a variety of cancers. In this study, co-delivery of Cas9 mRNA and guide RNA (gRNA) by lipid nanoparticles (LNP) for editing of LGMN gene was performed. For in-vitro transcription (IVT) of gRNA, two templates were designed: linearized pUC57-T7-gRNA and T7-gRNA oligos, and the effectiveness of gRNA was verified in multiple ways. Cas9 plasmid was modified and optimized for IVT of Cas9 mRNA. The effects of LGMN gene editing on lysosomal/autophagic function and cancer cell metastasis were investigated. Co-delivery of Cas9 mRNA and gRNA resulted in impaired lysosomal/autophagic degradation, clone formation, migration, and invasion capacity of cancer cells in-vitro. Experimental lung metastasis experiment indicates co-delivery of Cas9 mRNA and gRNA by LNP reduced the migration and invasion capacity of cancer cells in-vivo. These results indicate that co-delivery of Cas9 mRNA and gRNA can enhance the efficiency of CRISPR/Cas9-mediated gene editing in-vitro and in-vivo, and suggest that Cas9 mRNA and gRNA gene editing of LGMN may be a potential treatment for breast tumor metastasis.
Collapse
Affiliation(s)
- Yue Wang
- College of Pharmacy, Dali University, 2 HongShen Road, Dali, 671003, Yunnan, China
| | - Yatu Peng
- JinCai High School, 2788 Yang Gao Middle Road, Pudong New District, Shanghai, 200135, China
| | - Guanghui Zi
- College of Pharmacy, Dali University, 2 HongShen Road, Dali, 671003, Yunnan, China
| | - Jin Chen
- College of Pharmacy, Dali University, 2 HongShen Road, Dali, 671003, Yunnan, China
| | - Baowei Peng
- College of Pharmacy, Dali University, 2 HongShen Road, Dali, 671003, Yunnan, China.
| |
Collapse
|
7
|
Sharma A, Verwilst P, Li M, Ma D, Singh N, Yoo J, Kim Y, Yang Y, Zhu JH, Huang H, Hu XL, He XP, Zeng L, James TD, Peng X, Sessler JL, Kim JS. Theranostic Fluorescent Probes. Chem Rev 2024; 124:2699-2804. [PMID: 38422393 PMCID: PMC11132561 DOI: 10.1021/acs.chemrev.3c00778] [Citation(s) in RCA: 86] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.
Collapse
Affiliation(s)
- Amit Sharma
- Amity
School of Chemical Sciences, Amity University
Punjab, Sector 82A, Mohali 140 306, India
| | - Peter Verwilst
- Rega
Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49, Box 1041, 3000 Leuven, Belgium
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Dandan Ma
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Nem Singh
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Jiyoung Yoo
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Ying Yang
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Jing-Hui Zhu
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Haiqiao Huang
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xi-Le Hu
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiao-Peng He
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- National
Center for Liver Cancer, the International Cooperation Laboratory
on Signal Transduction, Eastern Hepatobiliary
Surgery Hospital, Shanghai 200438, China
| | - Lintao Zeng
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- School
of Chemistry and Chemical Engineering, Henan
Normal University, Xinxiang 453007, China
| | - Xiaojun Peng
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- State
Key Laboratory of Fine Chemicals, Dalian
University of Technology, Dalian 116024, China
| | - Jonathan L. Sessler
- Department
of Chemistry, The University of Texas at
Austin, Texas 78712-1224, United
States
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- TheranoChem Incorporation, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
8
|
Ghaemi B, Tanwar S, Singh A, Arifin DR, McMahon MT, Barman I, Bulte JWM. Cell-Penetrating and Enzyme-Responsive Peptides for Targeted Cancer Therapy: Role of Arginine Residue Length on Cell Penetration and In Vivo Systemic Toxicity. ACS APPLIED MATERIALS & INTERFACES 2024; 16:11159-11171. [PMID: 38385360 PMCID: PMC11362383 DOI: 10.1021/acsami.3c14908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
For the improved delivery of cancer therapeutics and imaging agents, the conjugation of cell-penetrating peptides (CPPs) increases the cellular uptake and water solubility of agents. Among the various CPPs, arginine-rich peptides have been the most widely used. Combining CPPs with enzyme-responsive peptides presents an innovative strategy to target specific intracellular enzymes in cancer cells and when combined with the appropriate click chemistry can enhance theranostic drug delivery through the formation of intracellular self-assembled nanostructures. However, one drawback of CPPs is their high positive charge which can cause nonspecific binding, leading to off-target accumulation and potential toxicity. Hence, balancing cell-specific penetration, toxicity, and biocompatibility is essential for future clinical efficacy. We synthesized six cancer-specific, legumain-responsive RnAANCK peptides containing one to six arginine residues, with legumain being an asparaginyl endopeptidase that is overexpressed in aggressive prostate tumors. When conjugated to Alexa Fluor 488, R1-R6AANCK peptides exhibited a concentration- and time-dependent cell penetration in prostate cancer cells, which was higher for peptides with higher R values, reaching a plateau after approximately 120 min. Highly aggressive DU145 prostate tumor cells, but not less aggressive LNCaP cells, self-assembled nanoparticles in the cytosol after the cleavage of the legumain-specific peptide. The in vivo biocompatibility was assessed in mice after the intravenous injection of R1-R6AANCK peptides, with concentrations ranging from 0.0125 to 0.4 mmol/kg. The higher arginine content in R4-6 peptides showed blood and urine indicators for the impairment of bone marrow, liver, and kidney function in a dose-dependent manner, with instant hemolysis and morbidity in extreme cases. These findings underscore the importance of designing peptides with the optimal arginine residue length for a proper balance of cell-specific penetration, toxicity, and in vivo biocompatibility.
Collapse
Affiliation(s)
- Behnaz Ghaemi
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Swati Tanwar
- Department of Mechanical Engineering, The Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland 21218, United States
| | - Aruna Singh
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Inc., Baltimore, Maryland 21205, United States
| | - Dian R Arifin
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Michael T McMahon
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Inc., Baltimore, Maryland 21205, United States
| | - Ishan Barman
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Mechanical Engineering, The Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland 21218, United States
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Inc., Baltimore, Maryland 21205, United States
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland 21218, United States
| |
Collapse
|
9
|
Bandi DR, Chitturi CMK, Aswathanarayan JB, Veeresh PKM, Bovilla VR, Sukocheva OA, Devi PS, Natraj SM, Madhunapantula SV. Pigmented Microbial Extract (PMB) from Exiguobacterium Species MB2 Strain (PMB1) and Bacillus subtilis Strain MB1 (PMB2) Inhibited Breast Cancer Cells Growth In Vivo and In Vitro. Int J Mol Sci 2023; 24:17412. [PMID: 38139241 PMCID: PMC10743659 DOI: 10.3390/ijms242417412] [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: 10/20/2023] [Revised: 12/01/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Breast cancer (BC) continues to be one of the major causes of cancer deaths in women. Progress has been made in targeting hormone and growth factor receptor-positive BCs with clinical efficacy and success. However, little progress has been made to develop a clinically viable treatment for the triple-negative BC cases (TNBCs). The current study aims to identify potent agents that can target TNBCs. Extracts from microbial sources have been reported to contain pharmacological agents that can selectively inhibit cancer cell growth. We have screened and identified pigmented microbial extracts (PMBs) that can inhibit BC cell proliferation by targeting legumain (LGMN). LGMN is an oncogenic protein expressed not only in malignant cells but also in tumor microenvironment cells, including tumor-associated macrophages. An LGMN inhibition assay was performed, and microbial extracts were evaluated for in vitro anticancer activity in BC cell lines, angiogenesis assay with chick chorioallantoic membrane (CAM), and tumor xenograft models in Swiss albino mice. We have identified that PMB from the Exiguobacterium (PMB1), inhibits BC growth more potently than PMB2, from the Bacillus subtilis strain. The analysis of PMB1 by GC-MS showed the presence of a variety of fatty acids and fatty-acid derivatives, small molecule phenolics, and aldehydes. PMB1 inhibited the activity of oncogenic legumain in BC cells and induced cell cycle arrest and apoptosis. PMB1 reduced the angiogenesis and inhibited BC cell migration. In mice, intraperitoneal administration of PMB1 retarded the growth of xenografted Ehrlich ascites mammary tumors and mitigated the proliferation of tumor cells in the peritoneal cavity in vivo. In summary, our findings demonstrate the high antitumor potential of PMB1.
Collapse
Affiliation(s)
- Deepa R. Bandi
- Department of Applied Microbiology, Sri Padmavathi Mahila Viswavidyalayam, Tirupati 517502, Andhra Pradesh, India; (D.R.B.); (P.S.D.)
| | - Ch M. Kumari Chitturi
- Department of Applied Microbiology, Sri Padmavathi Mahila Viswavidyalayam, Tirupati 517502, Andhra Pradesh, India; (D.R.B.); (P.S.D.)
| | - Jamuna Bai Aswathanarayan
- Department of Microbiology, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India;
| | - Prashant Kumar M. Veeresh
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India; (P.K.M.V.); (V.R.B.); (S.M.N.)
| | - Venugopal R. Bovilla
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India; (P.K.M.V.); (V.R.B.); (S.M.N.)
| | - Olga A. Sukocheva
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia;
| | - Potireddy Suvarnalatha Devi
- Department of Applied Microbiology, Sri Padmavathi Mahila Viswavidyalayam, Tirupati 517502, Andhra Pradesh, India; (D.R.B.); (P.S.D.)
| | - Suma M. Natraj
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India; (P.K.M.V.); (V.R.B.); (S.M.N.)
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory, Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India; (P.K.M.V.); (V.R.B.); (S.M.N.)
- Special Interest Group (SIG) in Cancer Biology and Cancer Stem Cells (CBCSC), JSS Academy of Higher Education & Research (JSS AHER), Mysore 570015, Karnataka, India
| |
Collapse
|
10
|
Tanwar S, Wu L, Zahn N, Raj P, Ghaemi B, Chatterjee A, Bulte JWM, Barman I. Targeted Enzyme Activity Imaging with Quantitative Phase Microscopy. NANO LETTERS 2023; 23:4602-4608. [PMID: 37154678 PMCID: PMC10798004 DOI: 10.1021/acs.nanolett.3c01090] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Quantitative phase imaging (QPI) is a powerful optical imaging modality for label-free, rapid, and three-dimensional (3D) monitoring of cells and tissues. However, molecular imaging of important intracellular biomolecules such as enzymes remains a largely unexplored area for QPI. Herein, we introduce a fundamentally new approach by designing QPI contrast agents that allow sensitive detection of intracellular biomolecules. We report a new class of bio-orthogonal QPI-nanoprobes for in situ high-contrast refractive index (RI) imaging of enzyme activity. The nanoprobes feature silica nanoparticles (SiO2 NPs) having higher RI than endogenous cellular components and surface-anchored cyanobenzothiazole-cysteine (CBT-Cys) conjugated enzyme-responsive peptide sequences. The nanoprobes specifically aggregated in cells with target enzyme activity, increasing intracellular RI and enabling precise visualization of intracellular enzyme activity. We envision that this general design of QPI-nanoprobes could open doors for spatial-temporal mapping of enzyme activity with direct implications for disease diagnosis and evaluating the therapeutic efficacy.
Collapse
Affiliation(s)
- Swati Tanwar
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Lintong Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Noah Zahn
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Piyush Raj
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Behnaz Ghaemi
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Arnab Chatterjee
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Jeff W M Bulte
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Inc., Baltimore, Maryland 21205, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University, School of Medicine, Baltimore, Maryland 21205, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| |
Collapse
|
11
|
Chu Y, Shi D, Wang N, Ren L, Liu N, Hu F, Meng W, Hong SJ, Bai X. Clonorchis sinensis legumain promotes migration and invasion of cholangiocarcinoma cells via regulating tumor-related molecules. Parasit Vectors 2023; 16:71. [PMID: 36797792 PMCID: PMC9933405 DOI: 10.1186/s13071-023-05694-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Clonorchis sinensis infection causes serious pathological changes in the bile duct and is highly correlated with cholangiocarcinoma. The excretory-secretory products (ESP) of C. sinensis play a critical role in the oncogenesis and progression of cholangiocarcinoma, while the components and precise mechanism remain unclear. Here, we evaluated the function of C. sinensis legumain (Cslegumain) in promoting the invasion and migration of cholangiocarcinoma cells and the mechanism involved. METHODS The structural and molecular characteristics of Cslegumain were predicted and analyzed using the online program Phyre2. Quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemical staining were performed to test the transcriptional level of Cslegumain and its localization in the adult. Native Cslegumain was detected by western blotting assay. The effects of Cslegumain on the proliferation, invasion and migration of cholangiocarcinoma cells were checked using CCK-8 assay, Matrigel transwell assay and scratch wound healing assay. Expression levels of tumor-related molecules regulated by Cslegumain were evaluated by qRT-PCR and western blotting assay. RESULTS Cslegumain showed high similarity with human legumain in the secondary and tertiary structures and displayed higher transcriptional levels in the adult worm than in the metacercariae. Native Cslegumain was detected in a catalytic form and was localized mainly in the intestine of the C. sinensis adult and epithelial cells of the intrahepatic bile duct. After transfection into RBE cells, Cslegumain showed high ability in promoting the invasion and migration but not the proliferation of cholangiocarcinoma RBE cells. Furthermore, the expression levels of some molecules including E-cadherin and N-cadherin were downregulated, while the levels of α-actinin 4, β-catenin and inducible nitric oxide synthase (iNOS) were upregulated. CONCLUSIONS Our findings indicated that Cslegumain showed very similar structures as those of human legumain and could promote the invasion and migration of cholangiocarcinoma cells by regulating some tumor-related molecules.
Collapse
Affiliation(s)
- Yanfei Chu
- grid.452240.50000 0004 8342 6962Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, 256603 Shandong People’s Republic of China
| | - Doufei Shi
- grid.452240.50000 0004 8342 6962Department of Geriatric Medicine, Binzhou Medical University Hospital, Binzhou, 256603 Shandong People’s Republic of China
| | - Nan Wang
- grid.452240.50000 0004 8342 6962Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, 256603 Shandong People’s Republic of China
| | - Lebin Ren
- grid.452240.50000 0004 8342 6962Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, 256603 Shandong People’s Republic of China
| | - Naiguo Liu
- grid.452240.50000 0004 8342 6962Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, 256603 Shandong People’s Republic of China
| | - Fengai Hu
- grid.452240.50000 0004 8342 6962Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, 256603 Shandong People’s Republic of China
| | - Wei Meng
- grid.452240.50000 0004 8342 6962Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, 256603 Shandong People’s Republic of China
| | - Sung-Jong Hong
- grid.254224.70000 0001 0789 9563Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Dongjak-Gu, Seoul, 156-756 Republic of Korea
| | - Xuelian Bai
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, 256603, Shandong, People's Republic of China.
| |
Collapse
|
12
|
Tu W, Qin M, Li Y, Wu W, Tong X. Metformin regulates autophagy via LGMN to inhibit choriocarcinoma. Gene X 2023; 853:147090. [PMID: 36464174 DOI: 10.1016/j.gene.2022.147090] [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: 07/22/2022] [Revised: 11/16/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Choriocarcinoma has the problem of chemotherapy insensitivity and recurrence. Metformin may be a promising candidate to restrict choriocarcinoma progress because of its indirect and direct beneficial role on inhabitations of cancer cells without severe adverse side effects. In this study, metformin pressed the proliferation and invasion of choriocarcinoma JAR cells in vitro and the growth of the JAR subcutaneous xenografts in vivo. The high throughput sequencing and bioinformatics technology identified the low expression of legumain (LGMN) in lysosomal pathway caused by metformin, which was upregulated in human choriocarcinoma tissues compared with the early pregnancy tissues. As elevating metformin concentration and treatment time, the mRNA and protein expression of LGMN both depressed in two choriocarcinoma cell lines (JAR and JEG-3). LGMN was involved in metformin-mediated inhibition of cell proliferation and invasion. Furthermore, metformin induced autophagy via inhibiting LGMN through AKT/mTOR/LC3II signaling pathway of choriocarcinoma. Autophagy inhibitor could depress metformin-induced autophagy and improve cell proliferation and invasion ability dropped by metformin, while autophagy inducer could partially reverse the change of cell proliferation and invasion modulated by combination of metformin and LGMN overexpression. These results indicated that metformin inhibited cell proliferation and invasion ability by inducing autophagy in a LGMN-dependent manner so as to play a role in the treatment of choriocarcinoma.
Collapse
Affiliation(s)
- Weiyan Tu
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Menglu Qin
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Li
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Weimin Wu
- Department of Gynecology, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaowen Tong
- Department of Gynecology and Obstetrics, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
13
|
Khan SU, Khan IM, Khan MU, Ud Din MA, Khan MZ, Khan NM, Liu Y. Role of LGMN in tumor development and its progression and connection with the tumor microenvironment. Front Mol Biosci 2023; 10:1121964. [PMID: 36825203 PMCID: PMC9942682 DOI: 10.3389/fmolb.2023.1121964] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
Legumain (LGMN) has been demonstrated to be overexpressed not just in breast, prostatic, and liver tumor cells, but also in the macrophages that compose the tumor microenvironment. This supports the idea that LGMN is a pivotal protein in regulating tumor development, invasion, and dissemination. Targeting LGMN with siRNA or chemotherapeutic medicines and peptides can suppress cancer cell proliferation in culture and reduce tumor growth in vivo. Furthermore, legumain can be used as a marker for cancer detection and targeting due to its expression being significantly lower in normal cells compared to tumors or tumor-associated macrophages (TAMs). Tumor formation is influenced by aberrant expression of proteins and alterations in cellular architecture, but the tumor microenvironment is a crucial deciding factor. Legumain (LGMN) is an in vivo-active cysteine protease that catalyzes the degradation of numerous proteins. Its precise biological mechanism encompasses a number of routes, including effects on tumor-associated macrophage and neovascular endothelium in the tumor microenvironment. The purpose of this work is to establish a rationale for thoroughly investigating the function of LGMN in the tumor microenvironment and discovering novel tumor early diagnosis markers and therapeutic targets by reviewing the function of LGMN in tumor genesis and progression and its relationship with tumor milieu.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China,Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China,*Correspondence: Ibrar Muhammad Khan, ; Yong Liu,
| | - Munir Ullah Khan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, International Research Center for X Polymers, Zhejiang University, Hangzhou, China
| | - Muhammad Azhar Ud Din
- Faculty of Pharmacy, Gomal University Dera Ismail Khan KPK, Dera IsmailKhan, Pakistan
| | - Muhammad Zahoor Khan
- Department of Animal Breeding and Genetics, Faculty of Veterinary and Animal Sciences, University of Agriculture, Dera IsmailKhan, Pakistan
| | - Nazir Muhammad Khan
- Department of Zoology, University of Science and Technology, Bannu, Pakistan
| | - Yong Liu
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China,*Correspondence: Ibrar Muhammad Khan, ; Yong Liu,
| |
Collapse
|
14
|
Kanathasan JS, Palanisamy UD, Radhakrishnan AK, Chakravarthi S, Thong TB, Swamy V. Protease-targeting peptide-functionalized porous silicon nanoparticles for cancer fluorescence imaging. Nanomedicine (Lond) 2022; 17:1511-1528. [PMID: 36382634 DOI: 10.2217/nnm-2022-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Porous silicon (pSi) nanoparticles (NPs) functionalized with suitable targeting ligands are now established cancer bioimaging agents and drug-delivery platforms. With growing interest in peptides as tumor-targeting ligands, much work has focused on the use of various peptides in combination with pSi NPs for cancer theranostics. Here, the authors investigated the targeting potential of pSi NPs functionalized with two types of peptide, a linear 10-mer peptide and its branched (Y-shaped) equivalent, that respond to legumain activity in tumor cells. Results: In vitro experiments established that the linear peptide-pSi NP conjugate had better aqueous stability under tumor conditions and higher binding efficiency (p < 0.001) toward legumain-expressing cells such as RAW 264.7 cells compared with that of its branched equivalent. In vivo studies (analyzed using ex vivo fluorescence) with the linear peptide-pSi NP formulation using a syngeneic mouse model of breast cancer showed a higher accumulation (p > 0.05) of linear peptide-conjugated pSi NPs in the tumor site within 4 h compared with nonconjugated pSi NPs. These results suggest that the linear peptide-pSi NP formulation is a nontoxic, stable and efficient fluorescence bioimaging agent and potential drug-delivery platform.
Collapse
Affiliation(s)
- Jayasree S Kanathasan
- Mechanical Engineering Discipline, School of Engineering, Monash University Malaysia, Bandar Sunway, Subang Jaya, Selangor, 47500, Malaysia
| | - Uma Devi Palanisamy
- Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Bandar Sunway, Subang Jaya, Selangor, 47500, Malaysia
| | - Ammu K Radhakrishnan
- Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Bandar Sunway, Subang Jaya, Selangor, 47500, Malaysia
| | - Srikumar Chakravarthi
- MAHSA University, Jalan SP 2, Bandar Saujana Putra, Jenjarom, Selangor, 42610, Malaysia
| | - Tan Boon Thong
- Mechanical Engineering Discipline, School of Engineering, Monash University Malaysia, Bandar Sunway, Subang Jaya, Selangor, 47500, Malaysia
| | - Varghese Swamy
- Mechanical Engineering Discipline, School of Engineering, Monash University Malaysia, Bandar Sunway, Subang Jaya, Selangor, 47500, Malaysia
| |
Collapse
|
15
|
The Role of Perineural Invasion in Prostate Cancer and Its Prognostic Significance. Cancers (Basel) 2022; 14:cancers14174065. [PMID: 36077602 PMCID: PMC9454778 DOI: 10.3390/cancers14174065] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Prostate cancer is one of the most frequently diagnosed cancers in men worldwide. Perineural invasion (PNI), the movement of cancer cells along nerves, is a commonly observed approach to tumor spread and is important in both research and clinical practice of prostate cancer. However, despite many studies reporting on molecules and pathways involved in PNI, understanding its clinical relevance remains insufficient. In this review, we aim to summarize the current knowledge of mechanisms and prognostic significance of PNI in prostate cancer, which may provide new perspectives for future studies and improved treatment. Abstract Perineural invasion (PNI) is a common indication of tumor metastasis that can be detected in multiple malignancies, including prostate cancer. In the development of PNI, tumor cells closely interact with the nerve components in the tumor microenvironment and create the perineural niche, which provides a supportive surrounding for their survival and invasion and benefits the nerve cells. Various transcription factors, cytokines, chemokines, and their related signaling pathways have been reported to be important in the progress of PNI. Nevertheless, the current understanding of the molecular mechanism of PNI is still very limited. Clinically, PNI is commonly associated with adverse clinicopathological parameters and poor outcomes for prostate cancer patients. However, whether PNI could act as an independent prognostic predictor remains controversial among studies due to inconsistent research aim and endpoint, sample type, statistical methods, and, most importantly, the definition and inclusion criteria. In this review, we provide a summary and comparison of the prognostic significance of PNI in prostate cancer based on existing literature and propose that a more standardized description of PNI would be helpful for a better understanding of its clinical relevance.
Collapse
|
16
|
Liu C, Wang J, Zheng Y, Zhu Y, Zhou Z, Liu Z, Lin C, Wan Y, Wen Y, Liu C, Yuan M, Zeng YA, Yan Z, Ge G, Chen J. Autocrine pro-legumain promotes breast cancer metastasis via binding to integrin αvβ3. Oncogene 2022; 41:4091-4103. [PMID: 35854065 DOI: 10.1038/s41388-022-02409-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 11/09/2022]
Abstract
Tumor metastasis is the leading cause of cancer-associated mortality. Unfortunately, the underlying mechanism of metastasis is poorly understood. Expression of legumain (LGMN), an endo-lysosomal cysteine protease, positively correlates with breast cancer metastatic progression and poor prognosis. Here, we report that LGMN is secreted in the zymogen form by motile breast cancer cells. Through binding to cell surface integrin αvβ3 via an RGD motif, the autocrine pro-LGMN activates FAK-Src-RhoA signaling in cancer cells and promotes cancer cell migration and invasion independent of LGMN protease activity. Either silencing LGMN expression or mutationally abolishing pro-LGMN‒αvβ3 interaction significantly inhibits cancer cell migration and invasion in vitro and breast cancer metastasis in vivo. Finally, we developed a monoclonal antibody against LGMN RGD motif, which blocks pro-LGMN‒αvβ3 binding, and effectively suppresses cancer cell migration and invasion in vitro and breast cancer metastasis in vivo. Thus, disruption of pro-LGMN‒integrin αvβ3 interaction may be a potentially promising strategy for treating breast cancer metastasis.
Collapse
Affiliation(s)
- Cui Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - JunLei Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - YaJuan Zheng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue Zhu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - ZhengHang Zhou
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - ZhaoYuan Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - ChangDong Lin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - YaoYing Wan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - YaTing Wen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - ChunYe Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - MengYa Yuan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - ZhanJun Yan
- Department of Orthopedics, Suzhou Ninth People's Hospital, Soochow University, Suzhou, 215000, China.
| | - GaoXiang Ge
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - JianFeng Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
17
|
The High Expression of Legumain in Canine Neoplasms: A Retrospective Analysis of 100 Cases. Animals (Basel) 2022; 12:ani12040504. [PMID: 35203212 PMCID: PMC8868329 DOI: 10.3390/ani12040504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/16/2022] [Accepted: 02/01/2022] [Indexed: 11/28/2022] Open
Abstract
Simple Summary Cancer is the leading cause of death in humans and is one of the most common canine diseases. The similarities in pathological features and tumor behaviors between spontaneous canine tumors and their human counterparts make dogs ideal models for comparative cancer research. Legumain is a novel asparaginyl endopeptidase that is overexpressed in numerous types of human tumors. Furthermore, legumain-targeted cancer therapy has been proposed, and the treatment efficacy is well-tolerated. Previous studies have shown that legumain regulates extracellular matrix degradation and triggers the invasion and the metastasis of tumors. However, in dogs, the role of legumain in the progression of tumors remains largely unknown, and few investigations have described the expression levels of this protein in canine tumors. The present study was carried out to evaluate whether legumain is expressed in ten different types of canine neoplasms. We found that heightened signals of legumain were expressed in all canine tumor samples in the study, and, notably, the non-mesenchymal types of tumors harbored relatively high expression levels. This study is the first to describe the legumain distribution pattern in a series of canine tumors. Though further investigation is needed, the current study has provided large-scale pan-screening data on legumain as a potential biomarker, or a therapeutic target, in veterinary oncology. Abstract Legumain, a novel asparaginyl endopeptidase, has been observed to be overexpressed in several types of human solid tumors. Elevated levels of legumain are found in human cancers, and this oncoprotein may facilitate tumor invasion and metastasis when overexpressed. These findings suggest that legumain plays a malignant role in cancer biology. However, currently, no publications have identified the role of legumain in the development of canine cancers. The present study first compared the expression patterns of legumain in paraffin-embedded canine tumor tissues, with those of normal tissues, by immunohistochemistry. A total of 100 canine tumor samples, including mast cell tumors, soft tissue sarcoma, hemangiosarcoma, lymphoma, mammary gland carcinoma, hepatoid gland tumor, squamous cell carcinoma, trichoblastoma, and melanoma were evaluated. Compared with the normal tissues, all tumor samples displayed high intensities of legumain expression. Mesenchymal-type tumors displayed immunoreactivity for legumain, with an average expression of 40.07% ± 1.70%, which was significantly lower than those of epithelial tumors and other types of tumors, which had median expressions of 49.12% ± 1.75% and 47.35% ± 2.71%, respectively (p < 0.05). These findings indicate that legumain has a high potential to be a candidate for distinguishing tumors from normal tissues. Although further studies on a larger number of cases are necessary to clarify the clinical application of legumain, the overexpression patterns of legumain in canine tumor tissues are reported, for the first time, in this study.
Collapse
|
18
|
Dall E, Stanojlovic V, Demir F, Briza P, Dahms SO, Huesgen PF, Cabrele C, Brandstetter H. The Peptide Ligase Activity of Human Legumain Depends on Fold Stabilization and Balanced Substrate Affinities. ACS Catal 2021; 11:11885-11896. [PMID: 34621593 PMCID: PMC8491156 DOI: 10.1021/acscatal.1c02057] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/30/2021] [Indexed: 12/11/2022]
Abstract
Protein modification by enzymatic breaking and forming of peptide bonds significantly expands the repertoire of genetically encoded protein sequences. The dual protease-ligase legumain exerts the two opposing activities within a single protein scaffold. Primarily localized to the endolysosomal system, legumain represents a key enzyme in the generation of antigenic peptides for subsequent presentation on the MHCII complex. Here we show that human legumain catalyzes the ligation and cyclization of linear peptides at near-neutral pH conditions, where legumain is intrinsically unstable. Conformational stabilization significantly enhanced legumain's ligase activity, which further benefited from engineering the prime substrate recognition sites for improved affinity. Additionally, we provide evidence that specific legumain activation states allow for differential regulation of its activities. Together these results set the basis for engineering legumain proteases and ligases with applications in biotechnology and drug development.
Collapse
Affiliation(s)
- Elfriede Dall
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Vesna Stanojlovic
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Fatih Demir
- Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Peter Briza
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Sven O. Dahms
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Pitter F. Huesgen
- Central Institute for Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, 52428 Jülich, Germany
- CECAD, Medical Faculty and University Hospital, University of Cologne, 50931 Cologne, Germany
- Institute for Biochemistry, Faculty of Mathematics and Natural Sciences, University of Cologne, 50674 Cologne, Germany
| | - Chiara Cabrele
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Hans Brandstetter
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| |
Collapse
|
19
|
Protease-triggered bioresponsive drug delivery for the targeted theranostics of malignancy. Acta Pharm Sin B 2021; 11:2220-2242. [PMID: 34522585 PMCID: PMC8424222 DOI: 10.1016/j.apsb.2021.01.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Proteases have a fundamental role in maintaining physiological homeostasis, but their dysregulation results in severe activity imbalance and pathological conditions, including cancer onset, progression, invasion, and metastasis. This striking importance plus superior biological recognition and catalytic performance of proteases, combining with the excellent physicochemical characteristics of nanomaterials, results in enzyme-activated nano-drug delivery systems (nanoDDS) that perform theranostic functions in highly specific response to the tumor phenotype stimulus. In the tutorial review, the key advances of protease-responsive nanoDDS in the specific diagnosis and targeted treatment for malignancies are emphatically classified according to the effector biomolecule types, on the premise of summarizing the structure and function of each protease. Subsequently, the incomplete matching and recognition between enzyme and substrate, structural design complexity, volume production, and toxicological issues related to the nanocomposites are highlighted to clarify the direction of efforts in nanotheranostics. This will facilitate the promotion of nanotechnology in the management of malignant tumors.
Collapse
|
20
|
Zhang W, Lin Y. The Mechanism of Asparagine Endopeptidase in the Progression of Malignant Tumors: A Review. Cells 2021; 10:cells10051153. [PMID: 34068767 PMCID: PMC8151911 DOI: 10.3390/cells10051153] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/24/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022] Open
Abstract
Asparagine endopeptidase (AEP), also called legumain, is currently the only known cysteine protease that specifically cleaves peptide bonds in asparaginyl residue in the mammalian genome. Since 2003, AEP has been reported to be widely expressed in a variety of carcinomas and is considered a potential therapeutic target. In the following years, researchers intensively investigated the substrates of AEP and the mechanism of AEP in partial tumors. With the identification of substrate proteins such as P53, integrin αvβ3, MMP-2, and MMP-9, the biochemical mechanism of AEP in carcinomas is also more precise. This review will clarify the probable mechanisms of AEP in the progression of breast carcinoma, glioblastoma, gastric carcinoma, and epithelial ovarian carcinoma. This review will also discuss the feasibility of targeted therapy with AEP inhibitor (AEPI) in these carcinomas.
Collapse
|
21
|
Vizovisek M, Ristanovic D, Menghini S, Christiansen MG, Schuerle S. The Tumor Proteolytic Landscape: A Challenging Frontier in Cancer Diagnosis and Therapy. Int J Mol Sci 2021; 22:ijms22052514. [PMID: 33802262 PMCID: PMC7958950 DOI: 10.3390/ijms22052514] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
In recent decades, dysregulation of proteases and atypical proteolysis have become increasingly recognized as important hallmarks of cancer, driving community-wide efforts to explore the proteolytic landscape of oncologic disease. With more than 100 proteases currently associated with different aspects of cancer development and progression, there is a clear impetus to harness their potential in the context of oncology. Advances in the protease field have yielded technologies enabling sensitive protease detection in various settings, paving the way towards diagnostic profiling of disease-related protease activity patterns. Methods including activity-based probes and substrates, antibodies, and various nanosystems that generate reporter signals, i.e., for PET or MRI, after interaction with the target protease have shown potential for clinical translation. Nevertheless, these technologies are costly, not easily multiplexed, and require advanced imaging technologies. While the current clinical applications of protease-responsive technologies in oncologic settings are still limited, emerging technologies and protease sensors are poised to enable comprehensive exploration of the tumor proteolytic landscape as a diagnostic and therapeutic frontier. This review aims to give an overview of the most relevant classes of proteases as indicators for tumor diagnosis, current approaches to detect and monitor their activity in vivo, and associated therapeutic applications.
Collapse
|
22
|
Menezes TDA, Bustamante-Filho IC, Paschoal AFL, Dalberto PF, Bizarro CV, Bernardi ML, Ulguim RDR, Bortolozzo FP, Mellagi APG. Differential seminal plasma proteome signatures of boars with high and low resistance to hypothermic semen preservation at 5°C. Andrology 2021; 8:1907-1922. [PMID: 33460278 DOI: 10.1111/andr.12869] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/20/2020] [Accepted: 07/15/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hypothermic storage at 5°C has been investigated as an alternative to promote the prudent use of antibiotics for boar artificial insemination doses. However, this temperature is challenging for some ejaculates or boars. OBJECTIVE The present study aimed to identify putative biomarkers for semen resistance to hypothermic storage at 5°C by comparing the seminal plasma proteomes of boars with high and low seminal resistance to preservation at 5°C. MATERIALS AND METHODS From an initial group of 34 boars, 15 were selected based on the following criteria: ejaculate with ≤20% abnormal spermatozoa and at least 70% progressive motility at 120 hours of storage at 17°C. Then, based on the response to semen hypothermic storage at 5°C, boars were classified into two categories: high resistance-progressive motility of >75% in the three collections (n = 3); and low resistance-progressive motility of <75% in the three collections (n = 3). Seminal plasma proteins were analyzed in pools, and differential proteomics was performed using Multidimensional Protein Identification Technology. RESULTS Progressive motility was lower at 120 hours of storage in low resistance, compared to high resistance boars (P < .05). Acrosome and plasma membrane integrity were not affected by the boar category, storage time, or their interaction (P ≥ .104). Sixty-five proteins were considered for differential proteomics. Among the differentially expressed and exclusive proteins, the identification of proteins such cathepsin B, legumain, and cystatin B suggests significant changes in key enzymes (eg, metalloproteinases) involved in spermatogenesis, sperm integrity, and fertilizing potential. DISCUSSION AND CONCLUSION Differences in the seminal plasma suggest that proteins involved in the proteolytic activation of metalloproteinases and proteins related to immune response modulation could disrupt key cellular pathways during spermatogenesis and epididymal maturation, resulting in altered resistance to chilling injury. Further in vivo studies focusing on the immunological crosstalk between epithelial cells and gametes might explain how the immune regulators influence sperm resistance to hipothermic storage.
Collapse
Affiliation(s)
- Tila de Alcantara Menezes
- Setor de Suínos, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | - Pedro Ferrari Dalberto
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristiano Valim Bizarro
- Centro de Pesquisas em Biologia Molecular e Funcional, Instituto Nacional de Ciência e Tecnologia em Tuberculose, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-Graduação em Biologia Celular e Molecular, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mari Lourdes Bernardi
- Departamento de Zootecnia, Faculdade de Agronomia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafael da Rosa Ulguim
- Setor de Suínos, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | |
Collapse
|
23
|
Legumain Induces Oral Cancer Pain by Biased Agonism of Protease-Activated Receptor-2. J Neurosci 2020; 41:193-210. [PMID: 33172978 DOI: 10.1523/jneurosci.1211-20.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most painful cancers, which interferes with orofacial function including talking and eating. We report that legumain (Lgmn) cleaves protease-activated receptor-2 (PAR2) in the acidic OSCC microenvironment to cause pain. Lgmn is a cysteine protease of late endosomes and lysosomes that can be secreted; it exhibits maximal activity in acidic environments. The role of Lgmn in PAR2-dependent cancer pain is unknown. We studied Lgmn activation in human oral cancers and oral cancer mouse models. Lgmn was activated in OSCC patient tumors, compared with matched normal oral tissue. After intraplantar, facial or lingual injection, Lgmn evoked nociception in wild-type (WT) female mice but not in female mice lacking PAR2 in NaV1.8-positive neurons (Par2Nav1.8), nor in female mice treated with a Lgmn inhibitor, LI-1. Inoculation of an OSCC cell line caused mechanical and thermal hyperalgesia that was reversed by LI-1. Par2Nav1.8 and Lgmn deletion attenuated mechanical allodynia in female mice with carcinogen-induced OSCC. Lgmn caused PAR2-dependent hyperexcitability of trigeminal neurons from WT female mice. Par2 deletion, LI-1, and inhibitors of adenylyl cyclase or protein kinase A (PKA) prevented the effects of Lgmn. Under acidified conditions, Lgmn cleaved within the extracellular N terminus of PAR2 at Asn30↓Arg31, proximal to the canonical trypsin activation site. Lgmn activated PAR2 by biased mechanisms in HEK293 cells to induce Ca2+ mobilization, cAMP formation, and PKA/protein kinase D (PKD) activation, but not β-arrestin recruitment or PAR2 endocytosis. Thus, in the acidified OSCC microenvironment, Lgmn activates PAR2 by biased mechanisms that evoke cancer pain.SIGNIFICANCE STATEMENT Oral squamous cell carcinoma (OSCC) is one of the most painful cancers. We report that legumain (Lgmn), which exhibits maximal activity in acidic environments, cleaves protease-activated receptor-2 (PAR2) on neurons to produce OSCC pain. Active Lgmn was elevated in OSCC patient tumors, compared with matched normal oral tissue. Lgmn evokes pain-like behavior through PAR2 Exposure of pain-sensing neurons to Lgmn decreased the current required to generate an action potential through PAR2 Inhibitors of adenylyl cyclase and protein kinase A (PKA) prevented the effects of Lgmn. Lgmn activated PAR2 to induce calcium mobilization, cAMP formation, and activation of protein kinase D (PKD) and PKA, but not β-arrestin recruitment or PAR2 endocytosis. Thus, Lgmn is a biased agonist of PAR2 that evokes cancer pain.
Collapse
|
24
|
Wang Y, Zhang S, Wang H, Cui Y, Wang Z, Cheng X, Li W, Hou J, Ji Y, Liu T. High Level of Legumain Was Correlated With Worse Prognosis and Peritoneal Metastasis in Gastric Cancer Patients. Front Oncol 2020; 10:966. [PMID: 32766126 PMCID: PMC7378441 DOI: 10.3389/fonc.2020.00966] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/15/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Accumulating evidence has demonstrated that legumain (LGMN) is abnormally expressed in several malignancies and functions as an oncogene. However, the association between LGMN and gastric cancer (GC) has not yet been fully elucidated. In this study, we performed a comprehensive analysis of the role of LGMN in clinicopathologic characteristics and survival of GC patients. Methods: The study had two patient cohorts, The Cancer Genome Atlas (TCGA) cohort and the Zhongshan Hospital cohort, both of which were used to analyze the role of LGMN in GC samples. The relationship between LGMN and clinicopathologic characteristics was determined by the Chi-square test and logistic regression analysis. The Kaplan–Meier method and Cox proportional hazards regression analysis were conducted to investigate the prognostic role of LGMN in GC patients. Moreover, a nomogram was constructed based on the factors that were independently associated with peritoneal metastasis. Finally, the gene set enrichment analysis (GSEA) was conducted to explore the underlying pathways through which LGMN was involved in GC progression. Results: The mRNA and protein levels of LGMN were significantly upregulated in GC tissues, especially for diffuse-type GC. High level of LGMN was independently associated with poor prognosis in both TCGA and Zhongshan cohorts. Further analysis showed that increased protein level of LGMN was related to peritoneal metastasis in GC patients. In a nomogram model, the LGMN expression could help predict the possibility of peritoneal metastasis in GC patients. LGMN was a strong determinant for prediction of peritoneal metastasis. GC patients with high LGMN expression tended to have worse survival together with more frequent diffuse-type tumors and increased risk of peritoneal metastasis. The GSEA results showed that focal adhesion, ecm receptor interaction, cell adhesion molecules cams, TGF-β signaling pathway, JAK-STAT signaling pathway, gap junction, etc. were differentially enriched in the phenotype with high LGMN expression. Conclusion: LGMN was an independent prognostic factor for OS in GC patients. Increased expression of LGMN was significantly associated with peritoneal metastasis. The nomogram based on LGMN might guide the clinical decisions for patients with GC.
Collapse
Affiliation(s)
- Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | - Haiwei Wang
- Maternity and Children's Hospital of Fujian Province, Fujian Medical University, Fuzhou, China
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiming Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xi Cheng
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.,Center of Evidence-Based Medicine, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Chen HC, Rui W, You SY, Liu XW, Huang J, Chen HY. Evaluation of the anti-cervical cancer effect of a prodrug :CBZ-AAN-DOX with hypoxic cell culture and tumor-bearing zebrafish models. Exp Cell Res 2020; 391:111980. [PMID: 32229193 DOI: 10.1016/j.yexcr.2020.111980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/20/2020] [Accepted: 03/26/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Prodrugs are medications or compounds that, after administration, can be converted into pharmacologically active drugs through metabolism. Unlike conventional drugs, prodrugs have reduced adverse or unintended effects, which could become critical limitations in treatments such as chemotherapy. Previously through computer-aided drug design and chemical synthesis, we have obtained and examined a prodrug N-benzyloxycarbonyl-Ala-Asn-Doxorubicin (CBZ-AAN-DOX). CBZ-AAN-DOX is essentially Doxorubicin that is chemically-modified with tripeptides to target Legumain, a highly expressed protein in cancer cells and is involved in tumor metastasis and tumor microvessel formation. The difficulty to test the safety and efficacy of the prodrug (including the pharmacodynamic parameters of CBZ-AAN-DOX on metastasis and invasion of tumors, as well as cardiac and vascular toxicity) primarily comes from the lack of appropriate experimental models. METHODS Human cervical cancer cell lines CaSki under hypoxic conditions were used to evaluate the cell viability by CCK-8 assay after the prodrug treatment. Western blotting method was performed for Legumain protein determination in the cell culture. Wound healing and transwell invasion assays were performed to determine the effects of the prodrug on tumor metastasis and invasion, respectively. Zebrafish models were constructed for toxicity and angiogenesis visual analysis after in vivo treatment with the prodrug. RESULTS The CCK-8 results showed that CBZ-AAN-DOX exhibits an IC50 of 28.7 μM in 48 h on CaSki cells that had a lower cell inhibition rate than DOX 80.3 μM for 24 h. Legumain expression was significantly increased in a time-dependent manner in 48 h under hypoxia conditions. The results also showed that 13.9 μM of the prodrug significantly inhibited the migration and invasion of cells and the effects were significantly stronger than that of 41.8 μM of DOX under hypoxia conditions after 48 h. The effects of 160 μM of the prodrug on the survival rate of zebrafish after 72 h and heart-toxicity showed no obvious abnormalities. Cell metastasis and angiogenesis were also inhibited in tumor-bearing zebrafish model. CONCLUSION The findings in this study demonstrated that CBZ-AAN-DOX is a promising chemotherapy candidate with low toxicity and high efficiency for cervical cancer. Remarkably, the hypoxic culture model together with the zebrafish model serve as a good system for the evaluation of the toxicity, targeting and impact of the prodrug on tumor invasion and metastasis.
Collapse
Affiliation(s)
- Hong-Ce Chen
- Department of Pathogenic Biology and Immunology, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510632, Guangdong Province, PR China
| | - Wen Rui
- Centre for Novel Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong Province, PR China; Guangdong Engineering & Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong Province, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangzhou, 510006, Guangdong Province, PR China
| | - Si-Yuan You
- Department of Pathogenic Biology and Immunology, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510632, Guangdong Province, PR China
| | - Xia-Wan Liu
- Department of Pathogenic Biology and Immunology, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510632, Guangdong Province, PR China
| | - Jun Huang
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, Guangdong Province, PR China
| | - Hong-Yuan Chen
- Department of Pathogenic Biology and Immunology, School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510632, Guangdong Province, PR China; Guangdong Engineering & Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong Province, PR China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Guangzhou, 510006, Guangdong Province, PR China.
| |
Collapse
|
26
|
Li N, Liu C, Ma G, Tseng Y, Pan D, Chen J, Li F, Zeng X, Luo T, Chen S. Asparaginyl endopeptidase may promote liver sinusoidal endothelial cell angiogenesis via PI3K/Akt pathway. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2020; 111:214-222. [PMID: 30507245 DOI: 10.17235/reed.2018.5709/2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS pathological angiogenesis plays an important role in the progression of chronic liver diseases. Asparaginyl endopeptidase (AEP) participates in tumor angiogenesis and was recently shown to be associated with liver fibrosis. This study aimed to explore the effect of AEP on liver sinusoidal endothelial cell (LSECs) angiogenesis and determine the underlying mechanism. METHODS cultured LSECs were infected with lentiviruses in order to suppress AEP expression (AEP-KD1, AEP-KD2). The effect of AEP on LSECs proliferation, apoptosis and migration were subsequently determined by a CCK8 assay, flow cytometry and wound-healing and Transwell assays, respectively, in AEP knocked-down and control LSECs. The expression of the endothelial cell surface markers CD31, CD34 and von Willebrand factor (vWF) were detected by immunofluorescence assay and western blot. The angiogenic factors, vascular endothelial growth factor receptor 2 (VEGFR2) and interleukin 8 (IL 8) were detected by real-time PCR and western blot. The effect of AEP on vessel tube formation by LSECs was examined by Matrigel™ tube-formation assay. Phosphoinositide 3-kinase (PI3K)/Akt expression and phosphorylation were detected by western blot. RESULTS AEP was effectively knocked down by lentivirus infection in LSECs. Down-regulation of AEP expression significantly decreased proliferation and migration and increased apoptosis of LSECs. Moreover, expression levels of the endothelial cell surface markers CD31, CD34 and vWF, as well as angiogenic factors VEGFR2 and IL 8, were also reduced after AEP was knocked-down. The vessel tube formation abilities of AEP-KD1 and AEP-KD2 LSECs were significantly inhibited compared with LSECs without AEP knocked-down. Down-regulation of AEP also inhibited the phosphorylation of PI3K and Akt. CONCLUSION AEP promotes LSECs angiogenesis in vitro, possibly via the PI3K/Akt pathway. AEP may therefore be a potential therapeutic target for preventing the progression of liver fibrosis.
Collapse
Affiliation(s)
- Na Li
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Chu Liu
- Department of Gastroenterology, Zhongshan Hospital, Fudan Ubiversity, China
| | - Guifen Ma
- Department of Radiotherapy, Zhongshan Hospital, Fudan University, China
| | - Yujen Tseng
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Duyi Pan
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Jie Chen
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Feng Li
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Xiaoqing Zeng
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Tiancheng Luo
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| | - Shiyao Chen
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, China
| |
Collapse
|
27
|
The exosomal integrin α5β1/AEP complex derived from epithelial ovarian cancer cells promotes peritoneal metastasis through regulating mesothelial cell proliferation and migration. Cell Oncol (Dordr) 2020; 43:263-277. [PMID: 32080801 DOI: 10.1007/s13402-019-00486-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2019] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Epithelial ovarian cancer (EOC) is one of the most malignant cancers in the gynecologic system. Many patients are diagnosed at an advanced stage with disseminated intra-peritoneal metastases. EOC spreads via both direct extension and trans-coelomic spread. However, the interplay between human peritoneal mesothelial cells (HPMCs) and EOC cells is still ambiguous. We hypothesize that integrins (ITG) in HPMCs may play important roles in EOC metastasis. METHODS The expression of different integrin subtypes from HPMCs was assessed using Western blotting. The expression of integrin α5β1 (ITGA5B1) and its co-localization with asparaginyl endopeptidase (AEP) in HPMCs derived from EOC patients (EOC-HPMCs) were assessed using immunofluorescence. The role and mechanism of the exosomal ITGA5B1/AEP complex in HPMCs was assessed using both in vitro and in vivo assays. A retrospective study involving 234 cases was carried out to assess ITGA5B1 and AEP levels in circulating sera and ascites of EOC patients, as well as associations between ITGA5B1/AEP expression and overall survival. RESULTS We found that ITGA5B1was highly expressed and co-localized with AEP in EOC cells, and that the exosomal ITGA5B1/AEP complex secreted by EOC cells played an important role in the proliferation and migration of HPMCs. High levels of exosomal ITGA5B1/AEP were also found in circulating sera and ascites of EOC patients, and the expression of ITGA5B1/AEP in EOC tissues was found to be negatively associated with overall survival. CONCLUSIONS Our data indicate that EOCs may regulate the function of HPMCs through exosomal ITGA5B1/AEP, which may be crucial for peritoneal metastasis.
Collapse
|
28
|
Design of a New Peptide Substrate Probe of the Putative Biomarker Legumain with Potential Application in Prostate Cancer Diagnosis Ex Vivo. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09994-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
AbstractThe lysosomal endoprotease legumain (asparaginyl endoprotease) has been proposed as a putative biomarker in prostate tumours, in which the enzyme is markedly overexpressed. Overexpression, coupled with highly selective specificity for cleavage of substrates at the C-terminus of asparagine (Asn) residues, make legumain an attractive biochemical target for potential diagnosis, prognosis and treatment. We report the design, synthesis, characterisation and preliminary evaluation of a new rhodamine-B (Rho-B)-labelled legumain peptide substrate probe5[Rho-Pro-Ala-Asn-PEG-AQ(4-OH)] and its selective targeting to lysosomes in PC3 prostate cancer cells. Probe5was efficiently activated by recombinant human legumain to afford the high quantum yield reporter fluorophore tripeptide4b(Rho-Pro-Ala-Asn-OH) with concomitant release of intense fluorescence. Furthermore, probe5was activated upon incubation with homogenates derived from fresh-frozen tissue material of prostatectomy specimens. Probe5represents a new viable biochemical tool for probing the activity of legumain with the potential to be used in ex vivo diagnostics in the cancer pathology laboratory.
Collapse
|
29
|
Fuchigami T, Itagaki K, Ishikawa N, Yoshida S, Nakayama M. Synthesis and evaluation of radioactive/fluorescent peptide probes for imaging of legumain activity. Bioorg Med Chem Lett 2019; 29:126629. [PMID: 31445852 DOI: 10.1016/j.bmcl.2019.126629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/22/2019] [Accepted: 08/17/2019] [Indexed: 11/29/2022]
Abstract
Legumain or asparaginyl endopeptidase is an enzyme overexpressed in some cancers and involved in cancer migration, invasion, and metastasis. We have developed radioiodine- ([125I]I-LCP) or fluorescein-labeled peptides (FL-LCP) with a cell-permeable d-Arg nonamer fused to an anionic d-Glu nonamer via a legumain-cleavable linker, to function as peptide probes that measure and monitor legumain activity. Non-cleavable probes of FL-NCP and [125I]I-NCP were similarly prepared and evaluated as negative control probes by altering their non-cleavable sequence. Model peptides with the legumain-cleavable or non-cleavable sequence (LCP and NCP, respectively) reacted with recombinant human legumain, and only LCP was digested by this enzyme. [125I]I-LCP uptake in legumain-positive HCT116 cells was significantly higher than that of [125I]I-NCP (11.2 ± 0.44% vs 1.75 ± 0.06% dose/mg). The accumulation of FL-LCP in the HCT116 cells was rather low (4.75 ± 0.29% dose/mg protein), but not significantly different from the levels of FL-NCP. It is possible that low concentrations of [125I]I-LCP (40 pM) can be effectively internalized after legumain cleavage. On the other hand, the cellular uptake of much higher concentrations of the FL-LCP derivative (1 mM) may be restricted by high concentrations of polyanions. The in vivo biodistribution studies in tumor-bearing mice demonstrated that the tumor uptake of [125I]I-LCP was 1.34% injected dose per gram (% ID/g) at 30 min. The tumor/blood and tumor/muscle ratios at 30 min were 0.63 and 1.77, respectively, indicating that the [125I]I-LCP accumulation in tumors was inadequate for in vivo imaging. Although further structural modifications are necessary to improve pharmacokinetic properties, [125I]I-LCP has been demonstrated to be an effective scaffold for the development of nuclear medicine imaging probes to monitor legumain activity in living subjects.
Collapse
Affiliation(s)
- Takeshi Fuchigami
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| | - Kohnosuke Itagaki
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Natsumi Ishikawa
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Sakura Yoshida
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Morio Nakayama
- Department of Hygienic Chemistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| |
Collapse
|
30
|
Eddie SL, Gregson A, Graham E, Burton S, Harrison T, Burden R, Scott CJ, Mullan PB, Williams R. Identification and SAR exploration of a novel series of Legumain inhibitors. Bioorg Med Chem Lett 2019; 29:1546-1548. [DOI: 10.1016/j.bmcl.2019.03.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/25/2019] [Accepted: 03/15/2019] [Indexed: 10/27/2022]
|
31
|
Li X, Liu Q, Ye S, Wang S, Li K, Lv G, Peng Y, Qiu L, Lin J. A protease-responsive fluorescent probe for sensitive imaging of legumain activity in living tumor cells. Chem Biol Drug Des 2019; 94:1494-1503. [PMID: 31002467 DOI: 10.1111/cbdd.13530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/06/2019] [Accepted: 04/06/2019] [Indexed: 01/25/2023]
Abstract
Legumain, a lysosomal cysteine protease, is critical for pathological progression and has been found to play an important role in the occurrence and development of several cancers. However, its biological functions remain few recognized. To further understand the role of legumain activity in tumor progression, a legumain protease-responsive fluorescent probe was developed in the present study. The probe 1 was synthesized by conjugating an aminoluciferin fluorophore with an alanine-alanine-asparagine (AAN) peptide sequence. The successful synthesis of probe 1 was validated by NMR and MS spectra as well as HPLC analysis. The probe 1 was non-toxic and exhibited great stability in the physiological solutions. More importantly, compared with the aminoluciferin fluorophore, the peptide conjugation may dramatically increase the targeting specificity. Probe 1 was able to effectively detect the legumain activity in living HCT116 cells through fluorescence imaging. All these results implied that probe 1 could act as a promising fluorescent probe specialized for the monitoring of legumain activity in living cells.
Collapse
Affiliation(s)
- Xi Li
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China.,Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Qingzhu Liu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Siqin Ye
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China.,School of Pharmaceutical Science, Jiangnan University, Wuxi, China
| | - Shijie Wang
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China.,Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Ke Li
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Gaochao Lv
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Ying Peng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Ling Qiu
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, China.,Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Jianguo Lin
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| |
Collapse
|
32
|
Kang L, Shen L, Lu L, Wang D, Zhao Y, Chen C, Du L, Gong J, Zhang Y, Mi X, Xiang R, Zhang M, Tan X. Asparaginyl endopeptidase induces endothelial permeability and tumor metastasis via downregulating zonula occludens protein ZO-1. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2267-2275. [PMID: 31096007 DOI: 10.1016/j.bbadis.2019.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Zona occludens-1 (ZO-1) is a key component of tight junctions that govern the function of the endothelial barrier against tumor metastasis. Factors secreted by tumor cells contribute to the maintenance of tumor vascular networks. How tumor cell-derived protein signals regulate ZO-1 expression is unclear. Here, we explored the effect of tumor cell-secreted asparaginyl endopeptidase (AEP) on the permeability of endothelial cells in the tumor microenvironment. First, we confirmed the existence of AEP in conditioned medium (CM) from AEP-overexpressing MDA-MB-231 and 4T1 cells. Treatment with CM from AEP-overexpressing tumor cells increased the permeability and tumor cell transversal of an endothelial monolayer. Furthermore, CM from AEP-overexpressing tumor cells suppressed endothelial ZO-1 expression, as well as ZO-1-associated nucleic acid binding protein ZONAB. In addition, the level of phosphorylated STAT3 was increased by treatment with AEP-containing CM. A mutation of RGD or blocking integrin αvβ3 with antibody recovered the ZO-1 downregulation induced by AEP. In vivo, a lung metastatic mouse model showed increased endothelial permeability in the AEP-overexpressing group compared with the control group. An orthotopic tumor transplantation model was established using AEP-overexpression and compared with mice receiving control 4T1 cells. Compared with controls, overexpression of AEP increased lung metastatic foci and area, as well as vascular instability in primary tumors or lung metastatic sites. Moreover, endothelial ZO-1 was decreased in the AEP-overexpressing group. Taken together, our data show that tumor cell-derived AEP increases the permeability of endothelial barriers. Interactions between RGD and endothelial integrin αvβ3 mediate this effect by downregulating ZO-1.
Collapse
Affiliation(s)
- Lichun Kang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Long Shen
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Liqing Lu
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Dekun Wang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Yong Zhao
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Chuan'ai Chen
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Lingfang Du
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Junbo Gong
- Tianjin Key Laboratory of Modern Drug Delivery and High Efficiency, Tianjin University, Tianjin 300072, China
| | - Yuying Zhang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Xue Mi
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Rong Xiang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Mianzhi Zhang
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing 100078, China.
| | - Xiaoyue Tan
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.
| |
Collapse
|
33
|
Toss MS, Miligy IM, Gorringe KL, McCaffrey L, AlKawaz A, Abidi A, Ellis IO, Green AR, Rakha EA. Legumain is an independent predictor for invasive recurrence in breast ductal carcinoma in situ. Mod Pathol 2019; 32:639-649. [PMID: 30429518 DOI: 10.1038/s41379-018-0180-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 12/21/2022]
Abstract
Legumain is a proteolytic enzyme that plays a role in the regulation of cell proliferation in invasive breast cancer. Studies evaluating its role in ductal carcinoma in situ (DCIS) are lacking. Here, we aimed to characterize legumain protein expression in DCIS and evaluate its prognostic significance. Legumain was assessed immunohistochemically in a tissue microarray of a well-characterized cohort of DCIS (n = 776 pure DCIS and n = 239 DCIS associated with invasive breast cancer (DCIS-mixed)). Legumain immunoreactivity was scored in tumor cells and surrounding stroma and related to clinicopathological parameters and patient outcome. High legumain expression was observed in 23% of pure DCIS and was associated with features of high-risk DCIS including higher nuclear grade, comedo necrosis, hormone receptor negativity, HER2 positivity, and higher proliferation index. Legumain expression was higher in DCIS associated with invasive breast cancer than in pure DCIS (p < 0.0001). In the DCIS-mixed cohort, the invasive component showed higher legumain expression than the DCIS component (p < 0.0001). Legumain was an independent predictor of shorter local recurrencefree interval for all recurrences (p = 0.0003) and for invasive recurrences (p = 0.002). When incorporated with other risk factors, legumain provided better patient risk stratification. High legumain expression is associated with poor prognosis in DCIS and could be a potential marker to predict DCIS progression to invasive disease.
Collapse
Affiliation(s)
- Michael S Toss
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK.,Histopathology department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Islam M Miligy
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK.,Histopathology department, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt
| | - Kylie L Gorringe
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - L McCaffrey
- Department of Oncology, Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Abdulbaqi AlKawaz
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK.,College of dentistry, Al Mustansiriya University, Baghdad, Iraq
| | - Asima Abidi
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Ian O Ellis
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Andrew R Green
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK
| | - Emad A Rakha
- Department of Histopathology, Nottingham Breast Cancer Research Center, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham University Hospital NHS Trust, Nottingham City Hospital, Nottingham, UK. .,Histopathology department, Faculty of Medicine, Menoufia University, Shebeen El-Kom, Egypt.
| |
Collapse
|
34
|
Cogo F, Williams R, Burden RE, Scott CJ. Application of nanotechnology to target and exploit tumour associated proteases. Biochimie 2019; 166:112-131. [PMID: 31029743 DOI: 10.1016/j.biochi.2019.04.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023]
Abstract
Proteases are hydrolytic enzymes fundamental for a variety of physiological processes, but the loss of their regulation leads to aberrant functions that promote onset and progression of many diseases including cancer. Proteases have been implicated in almost every hallmark of cancer and whilst widely investigated for tumour therapy, clinical adoption of protease inhibitors as drugs remains a challenge due to issues such as off-target toxicity and inability to achieve therapeutic doses at the disease site. Now, nanotechnology-based solutions and strategies are emerging to circumvent these issues. In this review, preclinical advances in approaches to enhance the delivery of protease drugs and the exploitation of tumour-derived protease activities to promote targeting of nanomedicine formulations is examined. Whilst this field is still in its infancy, innovations to date suggest that nanomedicine approaches to protease targeting or inhibition may hold much therapeutic and diagnostic potential.
Collapse
Affiliation(s)
- Francesco Cogo
- Centre for Cancer Research and Cell Biology, 97 Lisburn Road, BT9 7AE, UK
| | - Rich Williams
- Centre for Cancer Research and Cell Biology, 97 Lisburn Road, BT9 7AE, UK
| | - Roberta E Burden
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | | |
Collapse
|
35
|
Bosnjak T, Solberg R, Hemati PD, Jafari A, Kassem M, Johansen HT. Lansoprazole inhibits the cysteine protease legumain by binding to the active site. Basic Clin Pharmacol Toxicol 2019; 125:89-99. [PMID: 30916878 DOI: 10.1111/bcpt.13230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/20/2019] [Indexed: 12/13/2022]
Abstract
Proton pump inhibitors (PPIs) are prodrugs used in the treatment of peptic ulcer diseases. Once activated by acidic pH, the PPIs subsequently inhibit the secretion of gastric acid by covalently forming disulphide bonds with the SH groups of the parietal proton pump, that is the H+ /K+ -ATPase. Long-term use of PPIs has been associated with numerous adverse effects, including bone fractures. Considering the mechanism of activation, PPIs could also be active in acidic micro-environments such as in lysosomes, tumours and bone resorption sites. We suggested that the SH group in the active site of cysteine proteases could be susceptible for inhibition by PPIs. In this study, the inhibition by lansoprazole was shown on the cysteine proteases legumain and cathepsin B by incubating purified proteases or cell lysates with lansoprazole at different concentrations and pH conditions. The mechanism of legumain inhibition was shown to be a direct interaction of lansoprazole with the SH group in the active site, and thus blocking binding of the legumain-selective activity-based probe MP-L01. Lansoprazole was also shown to inhibit both legumain and cathepsin B in various cell models like HEK293, monoclonal legumain over-expressing HEK293 cells (M38L) and RAW264.7 macrophages, but not in human bone marrow-derived skeletal (mesenchymal) stem cells (hBMSC-TERT). During hBMSC-TERT differentiation to osteoblasts, lansoprazole inhibited legumain secretion, alkaline phosphatase activity, but had no effects on in vitro mineralization capacity. In conclusion, lansoprazole acts as a direct covalent inhibitor of cysteine proteases via disulphide bonds with the SH group in the protease active site. Such inhibition of cysteine proteases could explain some of the off-target effects of PPIs.
Collapse
Affiliation(s)
- Tatjana Bosnjak
- Section for Pharmacology and Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Rigmor Solberg
- Section for Pharmacology and Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Paya Diana Hemati
- Section for Pharmacology and Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark.,Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Harald Thidemann Johansen
- Section for Pharmacology and Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| |
Collapse
|
36
|
Chen X, Wang C, Liao K, Zhou S, Cao L, Chen J, Xu C, Lin Y. USP17 Suppresses Tumorigenesis and Tumor Growth through Deubiquitinating AEP. Int J Biol Sci 2019; 15:738-748. [PMID: 30906206 PMCID: PMC6429017 DOI: 10.7150/ijbs.30106] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
Ubiquitin-specific protease 17 (USP17), a novel member of deubiquitinase, is reported to play essential roles in several solid tumors. However, the expression and function of USP17 in breast cancer tumorigenesis remains ambiguity. Here we found that the mRNA level of USP17 was lower in breast cancer tissues than normal tissues. Meanwhile, higher USP17 level was detected in normal epithelial cell MCF-10A and a less-malignant cell MCF-7 than malignant cell line MDA-MB-231. Inhibition of USP17 in MCF7 cells enhanced tumorigenesis and tumor growth while overexpression of USP17 in malignant MDA-MB-231 cells reduced its tumorigenesis and growth ability in vitro and in vivo. Further study revealed that USP17 interacted with and deubiquitinated Asparaginyl endopeptidase (AEP), resulting in decreased protein levels of AEP. Moreover, knockdown of AEP inhibited breast cancer tumorigenesis and growth in vitro and in vivo through the inactivation of ERK signaling. Taken together, our works indicate that USP17 deubiquitinates AEP, down-regulates its protein level, and inhibits breast cancer tumorigenesis through disturbing ERK signaling. Thus, our data suggests that USP17 is a potential tumor suppressor in breast cancer and AEP is a promising target in breast cancer therapy.
Collapse
Affiliation(s)
- Xi Chen
- CAS key laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Chen Wang
- Shanghai Institute of Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Keman Liao
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai, 200127, China
| | - Sunhai Zhou
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai, 200127, China
| | - Lu Cao
- Department of Radiation Oncology, Ruijin Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai 200025, China
| | - Jiayi Chen
- Department of Radiation Oncology, Ruijin Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai 200025, China
| | - Cheng Xu
- Department of Radiation Oncology, Ruijin Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai 200025, China
| | - Yingying Lin
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao-tong University, Shanghai, 200127, China
| |
Collapse
|
37
|
Dutta A, Potier DN, Walker MJ, Gray OJ, Parker C, Holland M, Williamson AJK, Pierce A, Unwin RD, Krishnan S, Saha V, Whetton AD. Development of a selected reaction monitoring mass spectrometry-based assay to detect asparaginyl endopeptidase activity in biological fluids. Oncotarget 2018; 7:70822-70831. [PMID: 27683124 PMCID: PMC5342591 DOI: 10.18632/oncotarget.12224] [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: 05/16/2016] [Accepted: 09/15/2016] [Indexed: 12/22/2022] Open
Abstract
Cancer Biomarkers have the capability to improve patient outcomes. They have potential applications in diagnosis, prognosis, monitoring of disease progression and measuring response to treatment. This type of information is particularly useful in the individualisation of treatment regimens. Biomarkers may take many forms but considerable effort has been made to identify and quantify proteins in biological fluids. However, a major challenge in measuring protein in biological fluids, such as plasma, is the sensitivity of the assay and the complex matrix of proteins present. Furthermore, determining the effect of proteases in disease requires measurement of their activity in biological fluids as quantification of the protein itself may not provide sufficient information. To date little progress has been made towards monitoring activity of proteases in plasma. The protease asparaginyl endopeptidase has been implicated in diseases such as breast cancer, leukaemia and dementia. Here we describe a new approach to sensitively and in a targeted fashion quantify asparaginyl endopeptidase activity in plasma using a synthetic substrate peptide protected from nonspecific hydrolysis using D-amino acids within the structure. Our selected reaction monitoring approach enabled asparaginyl endopeptidase activity to be measured in human plasma with both a high dynamic range and sensitivity. This manuscript describes a paradigm for future development of assays to measure protease activities in biological fluids as biomarkers of disease.
Collapse
Affiliation(s)
- Anindita Dutta
- Stem Cell and Leukaemia Proteomics Laboratory, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Tata Translational Cancer Research Centre, Kolkata, India
| | - David N Potier
- Stem Cell and Leukaemia Proteomics Laboratory, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Michael J Walker
- Stem Cell and Leukaemia Proteomics Laboratory, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Oliver J Gray
- Stoller Biomarker Discovery Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catriona Parker
- Children's Cancer Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Mark Holland
- Children's Cancer Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew J K Williamson
- Stem Cell and Leukaemia Proteomics Laboratory, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew Pierce
- Stem Cell and Leukaemia Proteomics Laboratory, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Richard D Unwin
- Stem Cell and Leukaemia Proteomics Laboratory, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Current address: Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust and Institute of Human Development, University of Manchester, Manchester, UK
| | | | - Vaskar Saha
- Tata Translational Cancer Research Centre, Kolkata, India.,Children's Cancer Group, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Anthony D Whetton
- Stem Cell and Leukaemia Proteomics Laboratory, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Stoller Biomarker Discovery Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
38
|
Asparaginyl endopeptidase promotes the invasion and metastasis of gastric cancer through modulating epithelial-to-mesenchymal transition and analysis of their phosphorylation signaling pathways. Oncotarget 2018; 7:34356-70. [PMID: 27102302 PMCID: PMC5085161 DOI: 10.18632/oncotarget.8879] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/28/2016] [Indexed: 02/06/2023] Open
Abstract
Asparaginyl endopeptidase (AEP) is a lysosomal protease often overexpressed in gastric cancer. AEP was expressed higher in peritoneal metastatic loci than in primary gastric cancer. Then we overexpressed AEP or knocked it down with a lentiviral vector in gastric cancer cell lines and detected the cell cycle arrest and the changes of the invasive and metastatic ability in vitro and in vivo. When AEP was knocked-down, the proliferative, invasive and metastatic capacity of gastric cancer cells were inhibited, and the population of sub-G1 cells increased. AEP knockdown led to significant decrease of expression of transcription factor Twist and the mesenchymal markers N-cadherin, ß-catenin and Vimentin and to increased expression of epithelial marker E-cadherin. These results showed that AEP could promote invasion and metastasis by modulating EMT. We used phosphorylation-specific antibody microarrays to investigate the mechanism how AEP promotes gastric cancer invasion and metastasis, and found that the phosphorylation level of AKT and MAPK signaling pathways was decreased significantly if AEP was knocked-down. Therefore, AKT and MAPK signaling pathways took part in the modulation.
Collapse
|
39
|
Liu X, Wang Z, Zhang G, Zhu Q, Zeng H, Wang T, Gao F, Qi Z, Zhang J, Wang R. Overexpression of asparaginyl endopeptidase is significant for esophageal carcinoma metastasis and predicts poor patient prognosis. Oncol Lett 2017; 15:1229-1235. [PMID: 29399177 DOI: 10.3892/ol.2017.7433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 09/22/2017] [Indexed: 12/20/2022] Open
Abstract
Esophageal cancer is one of the most common types of cancer with poor prognosis. The molecular mechanisms of esophageal cancer progression remain unknown. In the present study, the aim was to investigate the clinical significance and biological function of protease asparaginyl endopeptidase (AEP) in esophageal cancer. The expression of AEP in esophageal cancer was examined, and its association with clinicopathological factors and patient prognosis was analyzed. A series of functional and mechanistic assays were performed to further investigate the underlying molecular mechanisms, and functions in esophageal cancer. The expression of AEP was elevated in esophageal cancer tissues, and patients with high AEP expression displayed a significantly shorter survival time compared with those with low AEP expression. In addition, loss of function experiments demonstrated that knockdown of AEP significantly reduced the migration and invasion ability of esophageal cancer cells. Furthermore, the pro-oncogenic effects of AEP in esophageal cancer were mediated by the upregulation of matrix-metalloproteinase 2 and 3. Taken together, the data from the present study indicates that high AEP expression is associated with esophageal cancer progression and AEP is an indicator of poor prognosis in patients with esophageal cancer. AEP therefore, may be considered as a novel prognostic biomarker or potential therapeutic target in esophageal cancer.
Collapse
Affiliation(s)
- Xinyang Liu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital Fudan University, Shanghai 200032, P.R. China
| | - Zhichao Wang
- Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai 200032, P.R. China
| | - Guoliang Zhang
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Qikun Zhu
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Hui Zeng
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Tao Wang
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Feng Gao
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Zhan Qi
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jinwen Zhang
- Department of Medical Affairs, Hebei Chest Hospital, Shijiazhuang, Hebei 050041, P.R. China
| | - Rui Wang
- Department of Thoracic Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
40
|
Qi Q, Obianyo O, Du Y, Fu H, Li S, Ye K. Blockade of Asparagine Endopeptidase Inhibits Cancer Metastasis. J Med Chem 2017; 60:7244-7255. [PMID: 28820254 DOI: 10.1021/acs.jmedchem.7b00228] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Asparagine endopeptidase (AEP), also called legumain, is highly expressed in various solid tumors, promoting cancer cell invasion, migration, and metastasis. It has been proposed to be a prognostic marker and therapeutic target for cancer treatment. However, an effective nonpeptide, small-molecule inhibitor against this protease has not yet been identified. Here we show that a family of xanthine derivatives selectively inhibit AEP and suppress matrix metalloproteinase (MMP) cleavage, leading to the inhibition of cancer metastasis. Through structure-activity relationship (SAR) analysis, we obtained an optimized lead compound (38u) that represses breast cancer invasion and migration. Chronic treatment of nude mice, which had been inoculated with MDA-MB-231 cells, with inhibitor 38u via oral administration robustly inhibits breast cancer lung metastasis in a dose-dependent manner, associated with blockade of MMP-2 by AEP. Therefore, our study supports that 38u might act as a potent and specific AEP inhibitor useful for cancer treatment.
Collapse
Affiliation(s)
- Qi Qi
- Department of Pathology and Laboratory Medicine ‡Department of Pharmacology, Emory Chemical Biology Discovery Center Emory University School of Medicine Atlanta, Georgia 30322, United States
| | - Obiamaka Obianyo
- Department of Pathology and Laboratory Medicine ‡Department of Pharmacology, Emory Chemical Biology Discovery Center Emory University School of Medicine Atlanta, Georgia 30322, United States
| | - Yuhong Du
- Department of Pathology and Laboratory Medicine ‡Department of Pharmacology, Emory Chemical Biology Discovery Center Emory University School of Medicine Atlanta, Georgia 30322, United States
| | - Haian Fu
- Department of Pathology and Laboratory Medicine ‡Department of Pharmacology, Emory Chemical Biology Discovery Center Emory University School of Medicine Atlanta, Georgia 30322, United States
| | - Shiyong Li
- Department of Pathology and Laboratory Medicine ‡Department of Pharmacology, Emory Chemical Biology Discovery Center Emory University School of Medicine Atlanta, Georgia 30322, United States
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine ‡Department of Pharmacology, Emory Chemical Biology Discovery Center Emory University School of Medicine Atlanta, Georgia 30322, United States
| |
Collapse
|
41
|
Wang ZH, Liu P, Liu X, Manfredsson FP, Sandoval IM, Yu SP, Wang JZ, Ye K. Delta-Secretase Phosphorylation by SRPK2 Enhances Its Enzymatic Activity, Provoking Pathogenesis in Alzheimer's Disease. Mol Cell 2017; 67:812-825.e5. [PMID: 28826672 DOI: 10.1016/j.molcel.2017.07.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/28/2017] [Accepted: 07/19/2017] [Indexed: 01/06/2023]
Abstract
Delta-secretase, a lysosomal asparagine endopeptidase (AEP), simultaneously cleaves both APP and tau, controlling the onset of pathogenesis of Alzheimer's disease (AD). However, how this protease is post-translationally regulated remains unclear. Here we report that serine-arginine protein kinase 2 (SRPK2) phosphorylates delta-secretase and enhances its enzymatic activity. SRPK2 phosphorylates serine 226 on delta-secretase and accelerates its autocatalytic cleavage, leading to its cytoplasmic translocation and escalated enzymatic activities. Delta-secretase is highly phosphorylated in human AD brains, tightly correlated with SRPK2 activity. Overexpression of a phosphorylation mimetic (S226D) in young 3xTg mice strongly promotes APP and tau fragmentation and facilitates amyloid plaque deposits and neurofibrillary tangle (NFT) formation, resulting in cognitive impairment. Conversely, viral injection of the non-phosphorylatable mutant (S226A) into 5XFAD mice decreases APP and tau proteolytic cleavage, attenuates AD pathologies, and reverses cognitive defects. Our findings support that delta-secretase phosphorylation by SRPK2 plays a critical role in aggravating AD pathogenesis.
Collapse
Affiliation(s)
- Zhi-Hao Wang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pathophysiology, Key Laboratory of Ministry of Education of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pai Liu
- Emory College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Fredric P Manfredsson
- Department of Translational Science and Molecular Medicine, Michigan State University, 333 Bostwick Avenue NE, Grand Rapids, MI 49503, USA
| | - Ivette M Sandoval
- Department of Translational Science and Molecular Medicine, Michigan State University, 333 Bostwick Avenue NE, Grand Rapids, MI 49503, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Co-innovation Center of Neuroregeneration, Nantong 226001, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.
| |
Collapse
|
42
|
Spiciarich DR, Nolley R, Maund SL, Purcell SC, Herschel J, Iavarone AT, Peehl DM, Bertozzi CR. Bioorthogonal Labeling of Human Prostate Cancer Tissue Slice Cultures for Glycoproteomics. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201701424] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- David R. Spiciarich
- College of Chemistry; University of California, Berkeley; Berkeley CA 94720 USA
| | - Rosalie Nolley
- Department of Urology; Stanford University School of Medicine; Stanford CA 94305 USA
| | - Sophia L. Maund
- Department of Urology; Stanford University School of Medicine; Stanford CA 94305 USA
| | - Sean C. Purcell
- College of Chemistry; University of California, Berkeley; Berkeley CA 94720 USA
| | - Jason Herschel
- Department of Mathematics; California State University; East Bay Hayward CA 94542 USA
| | - Anthony T. Iavarone
- QB3/Chemistry Mass Spectrometry Facility; UC Berkeley; Berkeley CA 94720 USA
| | - Donna M. Peehl
- Department of Urology; Stanford University School of Medicine; Stanford CA 94305 USA
| | - Carolyn R. Bertozzi
- Department of Chemistry; Stanford University; Stanford CA 94305-4401 USA
- Howard Hughes Medical Institute; USA
| |
Collapse
|
43
|
Spiciarich DR, Nolley R, Maund SL, Purcell SC, Herschel J, Iavarone AT, Peehl DM, Bertozzi CR. Bioorthogonal Labeling of Human Prostate Cancer Tissue Slice Cultures for Glycoproteomics. Angew Chem Int Ed Engl 2017. [PMID: 28649697 DOI: 10.1002/anie.201701424] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sialylated glycans are found at elevated levels in many types of cancer and have been implicated in disease progression. However, the specific glycoproteins that contribute to the cancer cell-surface sialylation are not well characterized, specifically in bona fide human disease tissue. Metabolic and bioorthogonal labeling methods have previously enabled the enrichment and identification of sialoglycoproteins from cultured cells and model organisms. Herein, we report the first application of this glycoproteomic platform to human tissues cultured ex vivo. Both normal and cancerous prostate tissues were sliced and cultured in the presence of the azide-functionalized sialic acid biosynthetic precursor Ac4 ManNAz. The compound was metabolized to the azidosialic acid and incorporated into cell surface and secreted sialoglycoproteins. Chemical biotinylation followed by enrichment and mass spectrometry led to the identification of glycoproteins that were found at elevated levels or uniquely in cancerous prostate tissue. This work therefore extends the use of bioorthogonal labeling strategies to problems of clinical relevance.
Collapse
Affiliation(s)
- David R Spiciarich
- College of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Rosalie Nolley
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sophia L Maund
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sean C Purcell
- College of Chemistry, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Jason Herschel
- Department of Mathematics, California State University, East Bay Hayward, CA, 94542, USA
| | - Anthony T Iavarone
- QB3/Chemistry Mass Spectrometry Facility, UC Berkeley, Berkeley, CA, 94720, USA
| | - Donna M Peehl
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, 94305-4401, USA.,Howard Hughes Medical Institute, USA
| |
Collapse
|
44
|
Zhou H, Sun H, Lv S, Zhang D, Zhang X, Tang Z, Chen X. Legumain-cleavable 4-arm poly(ethylene glycol)-doxorubicin conjugate for tumor specific delivery and release. Acta Biomater 2017; 54:227-238. [PMID: 28315495 DOI: 10.1016/j.actbio.2017.03.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/03/2017] [Accepted: 03/13/2017] [Indexed: 10/20/2022]
Abstract
Traditional chemotherapy strategy exists undesirable toxic side-effects to normal tissues due to the low selectively to cancer cells of micromolecule cytotoxic drugs. One considered method to realizing the targeted delivery and increasing the specificity to tumor tissues of the cytotoxic drug is to transporting and discharging it through an environment-sensitive mechanism. In this study, a novel enzyme-sensitive polymer-doxorubicin conjugate was designed to delivery chemotherapeutic drug in a tumor-specific behavior and selectively activated in tumor tissue. Briefly, doxorubicin (DOX) was conjugated to carboxyl-terminated 4-arm poly(ethylene glycol) through a tetrapeptide linker, alanine-alanine-asparagine-leucine (AANL), which was one of the substrates of legumain, an asparaginyl endopeptidase that was found presented in plants, mammals and also highly expressed in human tumor tissues. Hereinafter, the polymer-DOX conjugate was termed as 4-arm PEG-AANL-DOX. Dynamic laser scattering (DLS) and transmission electron microscopy (TEM) measurements indicated that the 4-arm PEG-AANL-DOX could self-assemble into micelles in aqueous solution. Drug release and in vitro cytotoxicity studies revealed that the 4-arm PEG-AANL-DOX could be cleaved by legumain. Ex vivo DOX fluorescence imaging measurements demonstrated that the 4-arm PEG-AANL-DOX had an improved tumor-targeting delivery as compared with the free DOX·HCl. In vivo studies on nude mice bearing MDA-MB-435 tumors revealed that the 4-arm PEG-AANL-DOX had a comparable anticancer efficacy with the free DOX·HCl but without DOX-related toxicities to normal tissues as measured by body weight change and histological assessments, indicating that the 4-arm PEG-AANL-DOX had an improved therapeutic index for cancer therapy. STATEMENT OF SIGNIFICANCE Herein we describe the construction of a novel tumor environment-sensitive delivery system through the instruction of a legumain-cleavable linkage to a polymer-DOX conjugate (4-arm PEG-AANL-DOX). This particular design strategy allows for polymer-DOX conjugates to be delivered in a tumor-specific manner and selectively activable in tumor microenvironment so that it can combine the advantages of tumor-specific delivery and tumor intracellular microenvironment-triggered release systems.
Collapse
|
45
|
Cui Y, Li Q, Li H, Wang Y, Wang H, Chen W, Zhang S, Cao J, Liu T. Asparaginyl endopeptidase improves the resistance of microtubule-targeting drugs in gastric cancer through IQGAP1 modulating the EGFR/JNK/ERK signaling pathway. Onco Targets Ther 2017; 10:627-643. [PMID: 28223821 PMCID: PMC5304996 DOI: 10.2147/ott.s125579] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
PURPOSE In recent years, understanding of the role of asparaginyl endopeptidase (AEP) in tumorigenesis has steadily increased. In this study, we investigated whether AEP expression correlates with sensitivity to chemotherapeutic drugs in gastric cancer and explored the mechanism. PATIENTS AND METHODS AEP expression in the serum of patients' peripheral blood was measured by enzyme-linked immunosorbent assay. Patient survival time was evaluated using univariate and multivariate analyses. Mass spectrometry and co-immunoprecipitation assays were utilized to discover proteins that interact with AEP. Gastric cancer cell lines were established, in which AEP was overexpressed or knocked out using lentiviral CRISPR. The proliferative abilities of these cell lines in response to chemotherapy agents were evaluated using the Cell Counting Kit-8 method. Gene expression changes in these lines were assessed by real-time polymerase chain reaction and Western blot. RESULTS Patients with low expression of AEP were significantly more likely to have a good prognosis and experience complete response or partial response after treatment with docetaxel/S-1 regimen. Mass spectrum analysis showed that several proteins in the focal adhesion and mitogen-activated protein kinase signaling pathways interacted with AEP. IQGAP1 was confirmed to be one of the proteins interacting with AEP, and its protein level increased when AEP was knocked out. AEP knockout decreased resistance to microtubule inhibitors, including paclitaxel, docetaxel, and T-DM1. The expression levels of MDR1, p-EGFR, p-JNK, p-ERK, and p-Rac1/cdc42 were decreased in AEP knockout gastric cancer cell lines, and inhibitors of both JNK and ERK could block AEP-induced expression of MDR1. CONCLUSION AEP was not only a prognostic factor but also a predictive marker. AEP knockout could inhibit the activity of the EGFR/JNK/ERK signaling pathway and improve sensitivity to microtubule inhibitors through interacting with IQGAP1.
Collapse
Affiliation(s)
| | | | | | | | - Hongshan Wang
- General Surgery Department, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Weidong Chen
- General Surgery Department, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Shangmin Zhang
- Pathology Department, Yale School of Medicine, New Haven, CT, USA
| | - Jian Cao
- Pathology Department, Yale School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
46
|
Yamane T, Kozuka M, Yamamoto Y, Nakano Y, Nakagaki T, Ohkubo I, Ariga H. Protease activity of legumain is inhibited by an increase of cystatin E/M in the DJ-1-knockout mouse spleen, cerebrum and heart. Biochem Biophys Rep 2017; 9:187-192. [PMID: 28956004 PMCID: PMC5614579 DOI: 10.1016/j.bbrep.2016.12.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 11/28/2016] [Accepted: 12/19/2016] [Indexed: 11/15/2022] Open
Abstract
Legumain (EC 3.4.22.34) is an asparaginyl endopeptidase. Legumain activity has been detected in various mouse tissues including the kidney, spleen and epididymis. Legumain is overexpressed in the majority of human solid tumors and transcription of the legumain gene is regulated by the p53 tumor suppressor in HCT116 cells. The legumain activity is also increased under acid conditions in Alzheimer's disease brains. DJ-1/PARK7, a cancer- and Parkinson's disease-associated protein, works as a coactivator to various transcription factors, including the androgen receptor, p53, PSF, Nrf2, SREBP and RREB1. Recently, we found that legumain expression, activation and cleavage of annexin A2 are regulated by DJ-1 through p53. In this study, we found that the expression levels of legumain mRNA were increased in the cerebrum, kidney, spleen, heart, lung, epididymis, stomach, small intestine and pancreas from DJ-1-knockout mice, although legumain activity levels were decreased in the cerebrum, spleen and heart from DJ-1-knockout mice. Furthermore, we found that cystatin E/M expression was increased in the spleen, cerebrum and heart from DJ-1-knockout mice. These results suggest that reduction of legumain activity is caused by an increase of cystatin E/M expression in the spleen, cerebrum and heart from DJ-1-knockout mice. Legumain is strongly activated in the epididymis from DJ-1-knockout mice. Expression level of legumain mRNA is increased but activity is decreased in the spleen, cerebrum and heart from DJ-1-knockout mice. Expression level of cystatin E/M is increased in the spleen, cerebrum and heart from DJ-1-knockout mice.
Collapse
Affiliation(s)
- Takuya Yamane
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Miyuki Kozuka
- Department of Health and Nutrition, Faculty of Human Science, Hokkaido Bunkyo University, Eniwa 061-1449, Japan
| | - Yoshio Yamamoto
- Laboratory of Environmental Chemistry, Mie University Iga Research Institute, Yumegaoka, Iga 518-0131, Japan
| | - Yoshihisa Nakano
- Center for Research and Development Bioresources, Research Organization for University-Community Collaborations, Osaka Prefecture University, Sakai, Osaka 599-8570, Japan
| | - Takenori Nakagaki
- Institute of Food Sciences, Nakagaki Consulting Engineer and Co., Ltd, Nishi-ku, Sakai 593-8328, Japan
| | - Iwao Ohkubo
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Higashi-ku, Sapporo 065-0013, Japan
| | - Hiroyoshi Ariga
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
47
|
Hong JA, Choi NE, La YK, Nam HY, Seo J, Lee J. Development of a smart activity-based probe to detect subcellular activity of asparaginyl endopeptidase in living cells. Org Biomol Chem 2017; 15:8018-8022. [DOI: 10.1039/c7ob01467h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A smart activity-based probe that generates a turn-on fluorescence signal in response to enzyme activity was developed, allowing dynamic imaging of subcellular enzyme activity in living cells.
Collapse
Affiliation(s)
- Jong-Ah Hong
- Department of Global Medical Science
- Sungshin University
- Seoul 01133
- Republic of Korea
| | - Na-Eun Choi
- Department of Global Medical Science
- Sungshin University
- Seoul 01133
- Republic of Korea
| | - Yeo-Kyoung La
- Department of Global Medical Science
- Sungshin University
- Seoul 01133
- Republic of Korea
| | - Ho Yeon Nam
- Department of Chemistry
- Gwangju Institute of Science and Technology
- Gwangju 61005
- Republic of Korea
| | - Jiwon Seo
- Department of Chemistry
- Gwangju Institute of Science and Technology
- Gwangju 61005
- Republic of Korea
| | - Jiyoun Lee
- Department of Global Medical Science
- Sungshin University
- Seoul 01133
- Republic of Korea
| |
Collapse
|
48
|
Zhang ZQ, Cao Z, Liu C, Li R, Wang WD, Wang XY. MiRNA-Embedded ShRNAs for Radiation-Inducible LGMN Knockdown and the Antitumor Effects on Breast Cancer. PLoS One 2016; 11:e0163446. [PMID: 27656894 PMCID: PMC5033420 DOI: 10.1371/journal.pone.0163446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 09/08/2016] [Indexed: 11/18/2022] Open
Abstract
Legumain (LGMN) is highly expressed in breast cancer (BC) and other solid tumors and is a potential anticancer target. Here we investigate the anti-tumor effects of short hairpin RNAs (shRNAs) targeting LGMN embedded in a microRNA-155 (miR-155) architecture, which is driven by a radiation-inducible chimeric RNA polymerase II (Pol II) promoter. Lentiviral vectors were generated with the chimeric promoter which controlled the expression of downstream shRNA-miR-155 cassette. Fluorescence was observed by using confocal microscopy. Real-time quantitative PCR and Western blotting were used to determine the expression level of LGMN, MMP2, and MMP9. Furthermore, the proliferation and invasive ability of BC cells was analyzed via plate colony formation and invasion assays. Here we demonstrated that the chimeric promoter could be effectively induced by radiation treatment. Furthermore, the shRNA-miR-155 cassette targeting LGMN could be effectively activated by the chimeric promoter. Radiation plus knockdown of LGMN impairs colony formation and dampens cell migration and invasion in BC cells. Inhibition of LGMN downregulates MMP2 and MMP9 expression in BC cells. Pol II-driven shRNA-miR-155 could effectively suppress the growth and invasiveness of BC cells, and that the interference effects could be regulated by radiation doses. Moreover, knockdown of LGMN alleviates the aggressive phenotype of BC cells through modulating MMPs expression.
Collapse
Affiliation(s)
- Zhi-Qiang Zhang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
- Department of Critical Care Medicine, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Zhi Cao
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu, 610041, China
| | - Cong Liu
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Rong Li
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Wei-Dong Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu, 610041, China
- * E-mail: (WW); (XW)
| | - Xing-Yong Wang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China
- Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China
- Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
- Department of Critical Care Medicine, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
- * E-mail: (WW); (XW)
| |
Collapse
|
49
|
Zhu W, Shao Y, Yang M, Jia M, Peng Y. Asparaginyl endopeptidase promotes proliferation and invasiveness of prostate cancer cells via PI3K/AKT signaling pathway. Gene 2016; 594:176-182. [PMID: 27590439 DOI: 10.1016/j.gene.2016.08.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 08/01/2016] [Accepted: 08/29/2016] [Indexed: 12/27/2022]
Abstract
Recurrence and metastasis are the major lethal causes of prostate cancer. It is urgent to find out the mechanisms and key factors governing prostate cancer progression and metastasis for developing new therapeutic strategies. Asparaginyl endopeptidase (AEP) overexpression has been found in a number of solid tumors. In prostate cancer, AEP has also been shown to exhibit a vesicular staining pattern and significantly associated with advanced tumor stage, high Gleason score, perineural invasion, and larger tumor. Here, we found that AEP was differentially expressed in prostate cancer cells with higher expression in 22RV1 cells and lower expression in PC-3 cells. AEP knockdown in 22RV1 cells significantly inhibited cell proliferation and invasion abilities while overexpression of AEP in PC-3 cells prompted cell proliferation and invasion abilities. Meanwhile, AEP knockdown upregulated cell apoptosis and vice versa. Further, we firstly identified that AEP promotes activation of the PI3K-AKT signaling pathway in prostate cancer cells. Taken together, our results suggest that AEP may be an attractive target for prostate cancer therapy.
Collapse
Affiliation(s)
- Wenjing Zhu
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqun Shao
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming Yang
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Moran Jia
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Peng
- Department of Urology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
50
|
Counter Selection Substrate Library Strategy for Developing Specific Protease Substrates and Probes. Cell Chem Biol 2016; 23:1023-35. [PMID: 27478158 DOI: 10.1016/j.chembiol.2016.05.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/11/2016] [Accepted: 05/30/2016] [Indexed: 01/29/2023]
Abstract
Legumain (AEP) is a lysosomal cysteine protease that was first characterized in leguminous seeds and later discovered in higher eukaryotes. AEP upregulation is linked to a number of diseases including inflammation, arteriosclerosis, and tumorigenesis. Thus this protease is an excellent molecular target for the development of new chemical markers. We deployed a hybrid combinatorial substrate library (HyCoSuL) approach to obtain P1-Asp fluorogenic substrates and biotin-labeled inhibitors that targeted legumain. Since this approach led to probes that were also recognized by caspases, we introduced a Counter Selection Substrate Library (CoSeSuL) approach that biases the peptidic scaffold against caspases, thus delivering highly selective legumain probes. The selectivity of these tools was validated using M38L and HEK293 cells. We also propose that the CoSeSuL methodology can be considered as a general principle in the design of selective probes for other protease families where selectivity is difficult to achieve by conventional sequence-based profiling.
Collapse
|