1
|
Yan G, Guo M, Yang R, Li X, Gu C, Wang K, Liu Y, Gao M, Huang C, Zou H. Copper Vacancy-Doped Cu 2-xSe Activating Nanozyme Sensor Arrays for Multiprotein Discrimination and Cancer Screening. Anal Chem 2025; 97:9345-9352. [PMID: 40267032 DOI: 10.1021/acs.analchem.5c00078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Given the complexity and interrelated nature of biological activities, acquiring multitarget information is crucial for accurate cancer diagnosis. In this study, we developed a cross-reactive sensor array using peroxidase-like nonstoichiometric copper selenide nanoparticles (Cu2-xSe), which featured varying copper vacancies, for multiplex protein detection and cancer screening. The Cu2-xSe nanoparticles demonstrated outstanding peroxidase-like activity that can be modulated to varying degrees in the presence of multiple proteins with varying isoelectric points and compositions, generating fingerprint outputs. By integrating pattern recognition method principal component analysis with linear discriminant analysis (PCA-LDA), the sensor array effectively discriminated among six proteins and was further applied to cells and clinical samples, attaining 100% accuracy in differentiating cancer patients from healthy individuals, as well as in identifying specific cancer types, namely, liver and prostate cancers. Moreover, as a proof-of-concept, the partial least-squares regression (PLSR) approached the accurate detection of AFP and PSA within the range of 0.1-2.0 μg/L in the complex biological matrix. These results highlighted the practical potential of multitarget sensing and clinical diagnosis by the nanozyme-based chemical nose strategies.
Collapse
Affiliation(s)
- Guojuan Yan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Meihan Guo
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ruiju Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xiaoxiao Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Chenlei Gu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ke Wang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yiming Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Mingxuan Gao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chengzhi Huang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Hongyan Zou
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Key Laboratory of Biomedical Analytics (Southwest University), Chongqing Science and Technology Bureau, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
2
|
Ning J, Shen Y, Gao H, Sun L, Bai X, Jin S, Wu Y, Sun Y, Xu Y, Li X, Pan L. Cathepsin B dependent activatable trigger fluorophore (CAT-Fluor) for in situ functional imaging of antibody-drug conjugates. Biosens Bioelectron 2025; 274:117184. [PMID: 39879789 DOI: 10.1016/j.bios.2025.117184] [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: 09/16/2024] [Revised: 01/18/2025] [Accepted: 01/18/2025] [Indexed: 01/31/2025]
Abstract
Antibody-drug conjugates (ADC) have emerged as an important class of therapeutic agents that combine the target specificity of a monoclonal antibody with the potency of a cytotoxic payload. Despite clinical success, our understanding of receptor endocytosis and ADC toxicity remains limited. Less than 1% of ADCs reach tumors, raising concerns about off-target cytotoxicity. To shed light on these issues, our study introduces a smart antibody-fluorophore conjugate (sAFC) with Cathepsin B dependent Activatable Trigger Fluorophore (CAT-Fluor) to mimic ADC behavior in situ. Using a Cathepsin B-cleavable linker, we linked a Si-rhodamine (SiR) derivative with superior near-infrared emission to antibodies, creating sAFC. Carbamoylation of the primary amino group on SiR is employed to conjugate with the linker and inhibit the electron-push-pull effect of the xanthene skeleton, thus inducing fluorescence quenching. In vitro, the anti-EGFR sAFC emulates ADC metabolism and suggests that specific proteins implicated in endocytosis, like caveolin, significantly influence ADC internalization efficacy, potentially correlating with drug resistance. In vivo studies using sAFC demonstrate that 'passenger ADCs' found in normal tissues release minimal payload, likely elucidating how ADCs mitigate dose-limiting toxicities. Therefore, our sAFC-based strategy, combining CAT-Fluor and targeted interventions, quantitatively and objectively evaluated the impact of various stages and key proteins in the physiological process, spanning from antigen recognition, endocytosis mechanism, to transport and protein hydrolysis, on ADC efficiency. This comprehensive approach lays a mechanistic foundation for advancing ADC research and development, and offers novel insights into tackling ADC efficacy, resistance and potential toxicities from the standpoint of endocytosis mechanisms.
Collapse
Affiliation(s)
- Jiangtao Ning
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yikai Shen
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Hongfan Gao
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Li Sun
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Xuefei Bai
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Shijie Jin
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yue Wu
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yanping Sun
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yingchun Xu
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Xin Li
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Liqiang Pan
- Deparment of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
3
|
He X, Tian S, Bu L, Zhao X, Zheng L, Zhang P, Guo R, Ma M. Cathepsin D inhibits AGEs-induced phenotypic transformation in vascular smooth muscle cells. Sci Rep 2025; 15:11502. [PMID: 40181129 PMCID: PMC11968932 DOI: 10.1038/s41598-025-96038-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 03/25/2025] [Indexed: 04/05/2025] Open
Abstract
This study investigates the role of Cathepsin D (CTSD) in diabetic vascular complications, particularly its impact on the phenotypic transformation of vascular smooth muscle cells (VSMCs) induced by advanced glycation end-products (AGEs), and explores its potential molecular mechanisms. CTSD was overexpressed in VSMCs using lentiviral vectors. Various methods, including CCK-8, immunofluorescence, SA-β-Gal staining, EdU assay, scratch assay, cell cycle analysis, and Western blotting, were employed to assess VSMC viability, proliferation, migration, senescence, and apoptosis. Additionally, transcriptomic and metabolomic analyses were conducted to investigate the molecular mechanisms underlying CTSD overexpression in VSMCs. AGEs treatment significantly inhibited CTSD expression in VSMCs, leading to reduced cell viability, enhanced proliferation and migration, increased senescence, and apoptosis. In contrast, overexpression of CTSD effectively inhibited AGEs-induced VSMCs proliferation, migration, senescence, and apoptosis. Combined transcriptomic and metabolomic analyses suggested that CTSD may affect VSMCs phenotypic transformation by inhibiting the glycolysis pathway. This study highlights the critical role of CTSD in the phenotypic transformation of VSMCs induced by AGEs and provides a new perspective for cardiovascular and cerebrovascular disease treatment. CTSD may emerge as a novel therapeutic target, though its specific molecular mechanisms and clinical application prospects in VSMCs phenotypic transformation require further investigation.
Collapse
Affiliation(s)
- Xingmin He
- Fenyang College of Shanxi Medical University, Fenyang, 032200, Shanxi, China
| | - Songhao Tian
- Department of Medical Laboratory Science, Fenyang College of Shanxi Medical University, Fenyang, 032200, Shanxi, China
| | - Lixia Bu
- Department of Geratology, Fenyang Hospital of Shanxi Province, Fenyang, 032200, Shanxi, China
| | - Xinna Zhao
- Research Office, Fenyang Hospital of Shanxi Province, Fenyang, 032200, Shanxi, China
| | - Liqiang Zheng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China
| | - Peigang Zhang
- Department of Cardiothoracic Surgery, Lvliang People's Hospital, Li Shi, 033000, Shanxi, China
| | - Renwei Guo
- Department of Cardiology, Fenyang Hospital of Shanxi Province, Fenyang, 032200, Shanxi, China.
| | - Mingfeng Ma
- Department of Cardiology, Fenyang Hospital of Shanxi Province, Fenyang, 032200, Shanxi, China.
- Department of Internal Medicine, Fenyang College of Shanxi Medical University, Fenyang, 032200, Shanxi, China.
| |
Collapse
|
4
|
Conesa-Bakkali R, Morillo-Huesca M, Martínez-Fábregas J. Non-Canonical, Extralysosomal Activities of Lysosomal Peptidases in Physiological and Pathological Conditions: New Clinical Opportunities for Cancer Therapy. Cells 2025; 14:68. [PMID: 39851495 PMCID: PMC11763575 DOI: 10.3390/cells14020068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Lysosomes are subcellular compartments characterised by an acidic pH, containing an ample variety of acid hydrolases involved in the recycling of biopolymers. Among these hydrolases, lysosomal proteases have merely been considered as end-destination proteases responsible for the digestion of waste proteins, trafficked to the lysosomal compartment through autophagy and endocytosis. However, recent reports have started to unravel specific roles for these proteases in the regulation of initially unexpected biological processes, both under physiological and pathological conditions. Furthermore, some lysosomal proteases are no longer restricted to the lysosomal compartment, as more novel non-canonical, extralysosomal targets are being identified. Currently, lysosomal proteases are accepted to play key functions in the extracellular milieu, attached to the plasma membrane and even in the cytosolic and nuclear compartments of the cell. Under physiological conditions, lysosomal proteases, through non-canonical, extralysosomal activities, have been linked to cell differentiation, regulation of gene expression, and cell division. Under pathological conditions, these proteases have been linked to cancer, mostly through their extralysosomal activities in the cytosol and nuclei of cells. In this review, we aim to provide a comprehensive summary of our current knowledge about the extralysosomal, non-canonical functions of lysosomal proteases, both under physiological and pathological conditions, with a particular interest in cancer, that could potentially offer new opportunities for clinical intervention.
Collapse
Affiliation(s)
- Ryan Conesa-Bakkali
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
| | - Macarena Morillo-Huesca
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
| | - Jonathan Martínez-Fábregas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Américo Vespucio 24, 41092 Sevilla, Spain; (R.C.-B.); (M.M.-H.)
- Departamento de Bioquímica Vegetal y Biología Molecular, Facultad de Biología, Universidad de Sevilla, Avenida Reina Mercedes, 41012 Sevilla, Spain
| |
Collapse
|
5
|
Stewart MR, Quentel A, Manalo E, Montoya Mira J, Ranganathan S, Branchaud BP, Fischer JM, Tu E, Civitci F, Chiu YJ, Yildirim A. Profiling protease cleavage patterns in plasma for pancreatic cancer detection. Sci Rep 2024; 14:31809. [PMID: 39738320 PMCID: PMC11686259 DOI: 10.1038/s41598-024-83077-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025] Open
Abstract
Proteases are promising biomarkers for cancer early detection. Their enzymatic activity against peptide substrates allows for their straightforward detection using low-cost tests. However, the complexity of the human proteome makes it challenging to develop sensitive and selective tests against a specific protease biomarker. Here, we report a different approach by utilizing the total protease activity in plasma samples to detect pancreatic cancer. Instead of targeting a specific protease using a specific peptide substrate, we utilized an array of 360 FRET substrates to screen for cleavage patterns in plasma samples collected from screen negatives and pancreatitis or pancreatic ductal adenocarcinoma cancer (PDAC) patients. In this proof of concept study, we first screened all 360 substrates using a small cohort (n = 13) to identify the top 5 substrates that best separate different conditions. Then, we performed a validation study using a larger cohort (n = 86) and the selected substrates. There was a statistically significant increase in the total protease activity in PDAC samples compared to screen negative and pancreatitis samples. The selected substrates detected PDAC with an area under the curve (AUC) of 0.8. This work represents a novel strategy for identifying peptide substrates for the detection of PDAC and other cancers.
Collapse
Affiliation(s)
- Morgan R Stewart
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Arnaud Quentel
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Elise Manalo
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Jose Montoya Mira
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Srivathsan Ranganathan
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Bruce P Branchaud
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Jared M Fischer
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Eugene Tu
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Fehmi Civitci
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Yu-Jui Chiu
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA
| | - Adem Yildirim
- CEDAR, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA.
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR, 97239, USA.
- Division of Oncological Sciences, Knight Cancer Institute, School of Medicine, Oregon Health and Science University, Portland, OR, 97201, USA.
| |
Collapse
|
6
|
Nolan-Stevaux O, Smith R. Logic-gated and contextual control of immunotherapy for solid tumors: contrasting multi-specific T cell engagers and CAR-T cell therapies. Front Immunol 2024; 15:1490911. [PMID: 39606234 PMCID: PMC11599190 DOI: 10.3389/fimmu.2024.1490911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
CAR-T cell and T cell engager therapies have demonstrated transformational efficacy against hematological malignancies, but achieving efficacy in solid tumors has been more challenging, in large part because of on-target/off-tumor toxicities and sub-optimal T cell anti-tumor cytotoxic functions. Here, we discuss engineering solutions that exploit biological properties of solid tumors to overcome these challenges. Using logic gates as a framework, we categorize the numerous approaches that leverage two inputs instead of one to achieve better cancer selectivity or efficacy in solid tumors with dual-input CAR-Ts or multi-specific TCEs. In addition to the "OR gate" and "AND gate" approaches that leverage dual tumor antigen targeting, we also review "contextual AND gate" technologies whereby continuous cancer-selective inputs such a pH, hypoxia, target density, tumor proteases, and immune-suppressive cytokine gradients can be creatively incorporated in therapy designs. We also introduce the notion of "output directionality" to distinguish dual-input strategies that mechanistically impact cancer cell killing or T cell fitness. Finally, we contrast the feasibility and potential benefits of the various approaches using CAR-T and TCE therapeutics and discuss why the promising "IF/THEN" and "NOT" gate types pertain more specifically to CAR-T therapies, but can also succeed by integrating both technologies.
Collapse
Affiliation(s)
| | - Richard Smith
- Cell Biology Research, Kite Pharma, Foster City, CA, United States
| |
Collapse
|
7
|
Li QQ, Guo M, He GH, Xi KH, Zhou MY, Shi RY, Chen GQ. VEGF-induced Nrdp1 deficiency in vascular endothelial cells promotes cancer metastasis by degrading vascular basement membrane. Oncogene 2024; 43:1836-1851. [PMID: 38654108 DOI: 10.1038/s41388-024-03038-9] [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: 08/04/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Vascular endothelial cells (VECs) are key players in the formation of neovessels and tumor metastasis, the ultimate cause of the majority of cancer-related human death. However, the crosstalk between VECs and metastasis remain greatly elusive. Based on our finding that tumor-associated VECs present significant decrease of Nrdp1 protein which is closely correlated with higher metastatic probability, herein we show that the conditional medium from hypoxia-incubated cancer cells induces extensive Nrdp1 downregulation in human and mouse VECs by vascular endothelial growth factor (VEGF), which activates CHIP, followed by Nrdp1 degradation in ubiquitin-proteasome-dependent way. More importantly, lung metastases of cancer cells significantly increase in conditional VECs Nrdp1 knockout mice. Mechanically, Nrdp1 promotes degradation of Fam20C, a secretory kinase involved in phosphorylating numerous secreted proteins. Reciprocally, deficiency of Nrdp1 in VECs (ecNrdp1) results in increased secretion of Fam20C, which induces degradation of extracellular matrix and disrupts integrity of vascular basement membrane, thus driving tumor metastatic dissemination. In addition, specific overexpression of ecNrdp1 by Nrdp1-carrying adeno-associated virus or chemical Nrdp1 activator ABPN efficiently mitigates tumor metastasis in mice. Collectively, we explore a new mechanism for VEGF to enhance metastasis and role of Nrdp1 in maintaining the integrity of vascular endothelium, suggesting that ecNrdp1-mediated signaling pathways might become potential target for anti-metastatic therapies.
Collapse
Affiliation(s)
- Qing-Qing Li
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Meng Guo
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China.
| | - Guang-Huan He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Kai-Hua Xi
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Mei-Yi Zhou
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Rong-Yi Shi
- Hainan Academy of Medical Sciences and School of Basic Medicine, Hainan Medical University, Hainan, 570000, China.
- Key Laboratory of Pediatric Hematology and Oncology in National Health Commission, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, SJTU-SM, Shanghai, 200127, China.
| | - Guo-Qiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China.
- Hainan Academy of Medical Sciences and School of Basic Medicine, Hainan Medical University, Hainan, 570000, China.
- Institute of Aging & Tissue Regeneration, State Key Laboratory of Systems Medicine for Cancer, Research Units of Stress and Tumor (2019RU043), Chinese Academy of Medical Sciences, Ren-Ji Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
8
|
Lu Y, Wang H, Chen S, Yang B, Li Y, Li Y. Cystatin SA attenuates gastric cancer cells growth and increases sensitivity to oxaliplatin via PI3K/AKT signaling pathway. J Cancer Res Clin Oncol 2024; 150:244. [PMID: 38717526 PMCID: PMC11078793 DOI: 10.1007/s00432-024-05780-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE Cystatin SA (CST2) belongs to the superfamily of cysteine protease inhibitors. Emerging research indicates that CST2 is often dysregulated across various cancers. Its role and molecular mechanisms in gastric cancer remain underexplored. This study aims to explore the expression and function of CST2 in gastric cancer. METHODS CST2 expression was analyzed and validated through Western blot. CST2 overexpression was induced by lentivirus in GC cells, and the correlation between CST2 expression levels and downstream signaling pathways was assessed. In addition, multiple assays, including cell proliferation, colony formation, wound-healing, and transwell migration/invasion, were considered to ascertain the influence of CST2 overexpression on gastric cancer. The cell cycle and apoptosis were detected by flow cytometry. RESULTS CST2 expression at the protein level was decreased to be reduced in both gastric cancer tissues and cell lines, and CST2 expression attenuate gastric cancer growth, an effect restricted to gastric cancer cells and absent in gastric epithelial GES-1 cells. Furthermore, CST2 was demonstrated to improve chemosensitivity to Oxaliplatin in gastric cancer cells through the PI3K/AKT signaling pathway. CONCLUSION These findings indicate that CST2 is downregulated at the protein level in gastric cancer tissues and cell lines. Additionally, CST2 was found to attenuate the growth of gastric cancer cells and to enhance sensitivity to Oxaliplatin through the PI3K/AKT signaling pathway, specific to gastric cancer cell lines. CST2 may serve as a tumor suppressor gene increasing sensitivity to Oxaliplatin in gastric cancer.
Collapse
Affiliation(s)
- Yida Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Huizhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Sihan Chen
- Taikang Ningbo Hospital, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Bo Yang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Yaxian Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| |
Collapse
|
9
|
Stock C. pH-regulated single cell migration. Pflugers Arch 2024; 476:639-658. [PMID: 38214759 PMCID: PMC11006768 DOI: 10.1007/s00424-024-02907-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024]
Abstract
Over the last two decades, extra- and intracellular pH have emerged as fundamental regulators of cell motility. Fundamental physiological and pathological processes relying on appropriate cell migration, such as embryonic development, wound healing, and a proper immune defense on the one hand, and autoimmune diseases, metastatic cancer, and the progression of certain parasitic diseases on the other, depend on surrounding pH. In addition, migrating single cells create their own localized pH nanodomains at their surface and in the cytosol. By this means, the migrating cells locally modulate their adhesion to, and the re-arrangement and digestion of, the extracellular matrix. At the same time, the cytosolic nanodomains tune cytoskeletal dynamics along the direction of movement resulting in concerted lamellipodia protrusion and rear end retraction. Extracellular pH gradients as found in wounds, inflamed tissues, or the periphery of tumors stimulate directed cell migration, and long-term exposure to acidic conditions can engender a more migratory and invasive phenotype persisting for hours up to several generations of cells after they have left the acidic milieu. In the present review, the different variants of pH-dependent single cell migration are described. The underlying pH-dependent molecular mechanisms such as conformational changes of adhesion molecules, matrix protease activity, actin (de-)polymerization, and signaling events are explained, and molecular pH sensors stimulated by H+ signaling are presented.
Collapse
Affiliation(s)
- Christian Stock
- Department of Gastroenterology, Hepatology, Infectiology & Endocrinology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
10
|
Pathan SU, Kharwar A, Ibrahim MA, Singh SB, Bajaj P. Enzymes as indispensable markers in disease diagnosis. Bioanalysis 2024; 16:485-497. [PMID: 38530222 PMCID: PMC11216522 DOI: 10.4155/bio-2023-0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 03/05/2024] [Indexed: 03/27/2024] Open
Abstract
Enzymes have been used for disease diagnosis for many decades; however, advancements in technology like ELISA and flow cytometry-based detection have significantly increased their use and have increased the sensitivity of detection. Technological advancements in recombinant enzyme production have increased enzymatic stability, and the use of colorimetric-based and florescence-based assays has led to their increased use as biomarkers for disease detection. Enzymes like acid phosphatase, cathepsin, lactate dehydrogenase, thymidine kinase and creatine kinase are indispensable markers for diagnosing cancer, cardiovascular diseases and others. This minireview summarizes various enzymes used in disease diagnosis, their metabolic role, market value and potential as disease markers across various metabolic and other disorders.
Collapse
Affiliation(s)
- Shehabaz Usman Pathan
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| | - Akash Kharwar
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| | - Madaje Amir Ibrahim
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| | - Shashi Bala Singh
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| | - Priyanka Bajaj
- National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, 500037, India
| |
Collapse
|
11
|
Zhang M, Zuo Y, Chen S, Li Y, Xing Y, Yang L, Wang H, Guo R. Antibody-drug conjugates in urothelial carcinoma: scientometric analysis and clinical trials analysis. Front Oncol 2024; 14:1323366. [PMID: 38665947 PMCID: PMC11044263 DOI: 10.3389/fonc.2024.1323366] [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: 10/17/2023] [Accepted: 02/12/2024] [Indexed: 04/28/2024] Open
Abstract
In 2020, bladder cancer, which commonly presents as urothelial carcinoma, became the 10th most common malignancy. For patients with metastatic urothelial carcinoma, the standard first-line treatment remains platinum-based chemotherapy, with immunotherapy serving as an alternative in cases of programmed death ligand 1 expression. However, treatment options become limited upon resistance to platinum and programmed death 1 or programmed death ligand 1 agents. Since the FDA's approval of Enfortumab Vedotin and Sacituzumab Govitecan, the therapeutic landscape has expanded, heralding a shift towards antibody-drug conjugates as potential first-line therapies. Our review employed a robust scientometric approach to assess 475 publications on antibody-drug conjugates in urothelial carcinoma, revealing a surge in related studies since 2018, predominantly led by U.S. institutions. Moreover, 89 clinical trials were examined, with 36 in Phase II and 13 in Phase III, exploring antibody-drug conjugates as both monotherapies and in combination with other agents. Promisingly, novel targets like HER-2 and EpCAM exhibit substantial therapeutic potential. These findings affirm the increasing significance of antibody-drug conjugates in urothelial carcinoma treatment, transitioning them from posterior-line to frontline therapies. Future research is poised to focus on new therapeutic targets, combination therapy optimization, treatment personalization, exploration of double antibody-coupled drugs, and strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Jilin University, Changchun, China
| | - Yuanye Zuo
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Siyi Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yaonan Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Rui Guo
- Department of Clinical Laboratory, First Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Chen YY, Li BP, Wang JF, Wang Y, Luo SS, Lin RJ, Liao XW, Chen JQ. Investigating the prognostic and predictive value of the type II cystatin genes in gastric cancer. BMC Cancer 2023; 23:1122. [PMID: 37978366 PMCID: PMC10657128 DOI: 10.1186/s12885-023-11550-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 10/19/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Accumulating evidence indicates that type II cystatin (CST) genes play a pivotal role in several tumor pathological processes, thereby affecting all stages of tumorigenesis and tumor development. However, the prognostic and predictive value of type II CST genes in GC has not yet been investigated. METHODS The present study evaluated the expression and prognostic value of type II CST genes in GC by using The Cancer Genome Atlas (TCGA) database and the Kaplan-Meier plotter (KM plotter) online database. The type II CST genes related to the prognosis of GC were then screened out. We then validated the expression and prognostic value of these genes by immunohistochemistry. We also used Database for Annotation, Visualization, and Integrated Discovery (DAVID), Gene Multiple Association Network Integration Algorithm (GeneMANIA), Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), nomogram, genome-wide co-expression analysis, and other bioinformatics tools to analyze the value of type II CST genes in GC and the underlying mechanism. RESULTS The data from the TCGA database and the KM plotter online database showed that high expression of CST2 and CST4 was associated with the overall survival (OS) of patients with GC. The immunohistochemical expression analysis showed that patients with high expression of CST4 in GC tissues have a shorter OS than those with low expression of CST4 (HR = 1.85,95%CI: 1.13-3.03, P = 0.015). Multivariate Cox regression analysis confirmed that the high expression level of CST4 was an independent prognostic risk factor for OS. CONCLUSIONS Our findings suggest that CST4 could serve as a tumor marker that affects the prognosis of GC and could be considered as a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Ye-Yang Chen
- Department of General Surgery, The First People's Hospital of Yulin, Yulin, China
| | - Bo-Pei Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun-Fu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ye Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shan-Shan Luo
- Department of Colorectal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Ru-Jing Lin
- Department of General Surgery, The People's Hospital of Binyang, Nanning, China
| | - Xi-Wen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jun-Qiang Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
13
|
Abdelaziz RF, Hussein AM, Kotob MH, Weiss C, Chelminski K, Studenik CR, Aufy M. The Significance of Cathepsin B in Mediating Radiation Resistance in Colon Carcinoma Cell Line (Caco-2). Int J Mol Sci 2023; 24:16146. [PMID: 38003335 PMCID: PMC10671642 DOI: 10.3390/ijms242216146] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Cathepsins (Caths) are lysosomal proteases that participate in various physiological and pathological processes. Accumulating evidence suggests that caths play a multifaceted role in cancer progression and radiotherapy resistance responses. Their proteolytic activity influences the tumor's response to radiation by affecting oxygenation, nutrient availability, and immune cell infiltration within the tumor microenvironment. Cathepsin-mediated DNA repair mechanisms can promote radioresistance in cancer cells, limiting the efficacy of radiotherapy. Additionally, caths have been associated with the activation of prosurvival signaling pathways, such as PI3K/Akt and NF-κB, which can confer resistance to radiation-induced cell death. However, the effectiveness of radiotherapy can be limited by intrinsic or acquired resistance mechanisms in cancer cells. In this study, the regulation and expression of cathepsin B (cath B) in the colon carcinoma cell line (caco-2) before and after exposure to radiation were investigated. Cells were exposed to escalating ionizing radiation doses (2 Gy, 4 Gy, 6 Gy, 8 Gy, and 10 Gy). Analysis of protein expression, in vitro labeling using activity-based probes DCG04, and cath B pull-down revealed a radiation-induced up-regulation of cathepsin B in a dose-independent manner. Proteolytic inhibition of cathepsin B by cathepsin B specific inhibitor CA074 has increased the cytotoxic effect and cell death due to ionizing irradiation treatment in caco-2 cells. Similar results were also obtained after cathepsin B knockout by CRISPR CAS9. Furthermore, upon exposure to radiation treatment, the inhibition of cath B led to a significant upregulation in the expression of the proapoptotic protein BAX, while it induced a significant reduction in the expression of the antiapoptotic protein BCL-2. These results showed that cathepsin B could contribute to ionizing radiation resistance, and the abolishment of cathepsin B, either by inhibition of its proteolytic activity or expression, has increased the caco-2 cells susceptibility to ionizing irradiation.
Collapse
Affiliation(s)
- Ramadan F. Abdelaziz
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (R.F.A.); (A.M.H.); (M.H.K.); (C.W.); (M.A.)
- Division of Human Health, International Atomic Energy Agency, Wagramer Str. 5, 1400 Vienna, Austria;
| | - Ahmed M. Hussein
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (R.F.A.); (A.M.H.); (M.H.K.); (C.W.); (M.A.)
| | - Mohamed H. Kotob
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (R.F.A.); (A.M.H.); (M.H.K.); (C.W.); (M.A.)
| | - Christina Weiss
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (R.F.A.); (A.M.H.); (M.H.K.); (C.W.); (M.A.)
| | - Krzysztof Chelminski
- Division of Human Health, International Atomic Energy Agency, Wagramer Str. 5, 1400 Vienna, Austria;
| | - Christian R. Studenik
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (R.F.A.); (A.M.H.); (M.H.K.); (C.W.); (M.A.)
| | - Mohammed Aufy
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (R.F.A.); (A.M.H.); (M.H.K.); (C.W.); (M.A.)
| |
Collapse
|
14
|
Luo FF, Wang J, Zhang ZF, Lin ST, Huang TJ, Liu BQ, Fan ML, Peng LX, Zheng ST, Yang CF, Huang BJ. SPI1-Mediated Upregulation of the CST1 Gene as an Independent Poor Prognostic Factor Accelerates Metastasis in Esophageal Squamous Cell Carcinoma (ESCC) by Interacting with MMP2. FRONT BIOSCI-LANDMRK 2023; 28:212. [PMID: 37796690 DOI: 10.31083/j.fbl2809212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/09/2023] [Accepted: 04/04/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a highly lethal tumor type, but studies on the ESCC tumor microenvironment are limited. We found that cystatin SN (CST1) plays an important role in the ESCC tumor microenvironment. CST1 has been reported to act as an oncogene in multiple human cancers, but its clinical significance and underlying mechanism in ESCC remain elusive. METHODS We performed ESCC gene expression profiling with data from RNA-sequencing and public databases and found CST1 upregulation in ESCC. Then, we assessed CST1 expression in ESCC by RT‒qPCR and Western blot analysis. In addition, immunohistochemistry (IHC) and enzyme-linked immunosorbent assay (ELISA) were used to estimate the expression of CST1 in ESCC tissue and serum. Moreover, further functional experiments were conducted to verify that the gain and loss of CST1 in ESCC cell lines significantly influenced the proliferation and metastasis of ESCC. Mass spectrometry, coimmunoprecipitation, and gelatin zymography experiments were used to validate the interaction between CST1 and matrix metalloproteinase 2 (MMP2) and the mechanism of CST1 influence on metastasis in ESCC. RESULTS Here, we found that CST1 expression was significantly elevated in ESCC tissues and serum. Moreover, compared with patients with low CST1 expression, patients with high CST1 expression had a worse prognosis. Overall survival (OS) and disease-free survival (DFS) were significantly unfavorable in the high CST1 expression subgroup. Likewise, the CST1 level was significantly increased in ESCC serum compared with healthy control serum, indicating that CST1 may be a potential serum biomarker for diagnosis, with an area under the curve (AUC) = 0.9702 and p < 0.0001 by receiver operating curve (ROC) analysis. Furthermore, upregulated CST1 can promote the motility and metastatic capacity of ESCC in vitro and in vivo by influencing epithelial mesenchymal transition (EMT) and interacting with MMP2 in the tumor microenvironment (TME). CONCLUSIONS Collectively, the results of this study indicated that high CST1 expression mediated by SPI1 in ESCC may serve as a potentially prognostic and diagnostic predictor and as an oncogene to promote motility and metastatic capacity of ESCC by influencing EMT and interacting with MMP2 in the TME.
Collapse
Affiliation(s)
- Fei-Fei Luo
- Department of Experimental Research, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Jing Wang
- Department of Experimental Research, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Zhan-Fei Zhang
- Department of Experimental Research, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong, China
- Department of Cardiothoracic Surgery, Zhongshan People's Hospital, 529403 Zhongshan, Guangdong, China
| | - Si-Ting Lin
- Department of Experimental Research, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong, China
- Department of Radiation Oncology, People's Hospital of Guangxi Zhuang Autonomous Region, 530021 Nanning, Guangxi, China
| | - Tie-Jun Huang
- Department of Nuclear Medicine, The Second People's Hospital of Shenzhen, 518035 Shenzhen, Guangdong, China
| | - Bao-Qi Liu
- Department of Experimental Research, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Mei-Ling Fan
- Department of Experimental Research, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Li-Xia Peng
- Department of Experimental Research, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Shu-Tao Zheng
- Department of Clinical Medical Research Institute, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, China
| | - Chang-Fu Yang
- Department of Oncology, The People's Hospital of Gaozhou, 6664126 Gaozhou, Guangdong, China
| | - Bi-Jun Huang
- Department of Experimental Research, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| |
Collapse
|
15
|
Li S, Pritchard DM, Yu LG. Galectin-3 promotes secretion of proteases that decrease epithelium integrity in human colon cancer cells. Cell Death Dis 2023; 14:268. [PMID: 37055381 PMCID: PMC10102123 DOI: 10.1038/s41419-023-05789-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 04/15/2023]
Abstract
Galectin-3 is a galactoside-binding protein that is commonly overexpressed in many epithelial cancers. It is increasingly recognized as a multi-functional, multi-mode promoter in cancer development, progression, and metastasis. This study reports that galectin-3 secretion by human colon cancer cells induces cancer cell secretion, in an autocrine/paracrine manner, of a number of proteases including cathepsin-B, MMP-1 and MMP-13. The secretion of these proteases causes disruption of epithelial monolayer integrity, increases its permeability and promotes tumour cell invasion. This effect of galectin-3 is shown to be mediated through induction of cellular PYK2-GSK3α/β signalling and can be prevented by the presence of galectin-3 binding inhibitors. This study thus reveals an important mechanism in galectin-3-mediated promotion of cancer progression and metastasis. It provides further evidence to the increased realization of galectin-3 as a potential therapeutic target for the treatment of cancer.
Collapse
Affiliation(s)
- Shun Li
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - David Mark Pritchard
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lu-Gang Yu
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
16
|
Noei-Khesht Masjedi M, Asgari Y, Sadroddiny E. Differential expression analysis in epithelial ovarian cancer using functional genomics and integrated bioinformatics approaches. INFORMATICS IN MEDICINE UNLOCKED 2023. [DOI: 10.1016/j.imu.2023.101172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
17
|
Ćwilichowska N, Świderska KW, Dobrzyń A, Drąg M, Poręba M. Diagnostic and therapeutic potential of protease inhibition. Mol Aspects Med 2022; 88:101144. [PMID: 36174281 DOI: 10.1016/j.mam.2022.101144] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 07/20/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Proteases are enzymes that hydrolyze peptide bonds in proteins and peptides; thus, they control virtually all biological processes. Our understanding of protease function has advanced considerably from nonselective digestive enzymes to highly specialized molecular scissors that orchestrate complex signaling networks through a limited proteolysis. The catalytic activity of proteases is tightly regulated at several levels, ranging from gene expression through trafficking and maturation to posttranslational modifications. However, when this delicate balance is disturbed, many diseases develop, including cancer, inflammatory disorders, diabetes, and neurodegenerative diseases. This new understanding of the role of proteases in pathologic physiology indicates that these enzymes represent excellent molecular targets for the development of therapeutic inhibitors, as well as for the design of chemical probes to visualize their redundant activity. Recently, numerous platform technologies have been developed to identify and optimize protease substrates and inhibitors, which were further used as lead structures for the development of chemical probes and therapeutic drugs. Due to this considerable success, the clinical potential of proteases in therapeutics and diagnostics is rapidly growing and is still not completely explored. Therefore, small molecules that can selectively target aberrant protease activity are emerging in diseases cells. In this review, we describe modern trends in the design of protease drugs as well as small molecule activity-based probes to visualize selected proteases in clinical settings.
Collapse
Affiliation(s)
- Natalia Ćwilichowska
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Karolina W Świderska
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Agnieszka Dobrzyń
- Nencki Institute of Experimental Biology, Ludwika Pasteura 3, 02-093, Warsaw, Poland
| | - Marcin Drąg
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland.
| | - Marcin Poręba
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland.
| |
Collapse
|
18
|
All Roads Lead to Cathepsins: The Role of Cathepsins in Non-Alcoholic Steatohepatitis-Induced Hepatocellular Carcinoma. Biomedicines 2022; 10:biomedicines10102351. [PMID: 36289617 PMCID: PMC9598942 DOI: 10.3390/biomedicines10102351] [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: 08/16/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Cathepsins are lysosomal proteases that are essential to maintain cellular physiological homeostasis and are involved in multiple processes, such as immune and energy regulation. Predominantly, cathepsins reside in the lysosomal compartment; however, they can also be secreted by cells and enter the extracellular space. Extracellular cathepsins have been linked to several pathologies, including non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC). NASH is an increasingly important risk factor for the development of HCC, which is the third leading cause of cancer-related deaths and poses a great medical and economic burden. While information regarding the involvement of cathepsins in NASH-induced HCC (NASH-HCC) is limited, data to support the role of cathepsins in either NASH or HCC is accumulating. Since cathepsins play a role in both NASH and HCC, it is likely that the role of cathepsins is more significant in NASH-HCC compared to HCC derived from other etiologies. In the current review, we provide an overview on the available data regarding cathepsins in NASH and HCC, argue that cathepsins play a key role in the transition from NASH to HCC, and shed light on therapeutic options in this context.
Collapse
|
19
|
NO news: S-(de)nitrosylation of cathepsins and their relationship with cancer. Anal Biochem 2022; 655:114872. [PMID: 36027970 DOI: 10.1016/j.ab.2022.114872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
Abstract
Tumor formation and progression have been much of a study over the last two centuries. Recent studies have seen different developments for the early diagnosis and treatment of the disease; some of which even promise survival of the patient. Cysteine proteases, mainly cathepsins have been unequivocally identified as putative worthy players of redox imbalance that contribute to the premonition and further progression of cancer by interfering in the normal extracellular and intracellular proteolysis and initiating a proteolytic cascade. The present review article focuses on the study of cancer so far, while establishing facts on how future studies focused on the cellular interrelation between nitric oxide (NO) and cancer, can direct their focus on cathepsins. For a tumor cell to thrive and synergize a cancerous environment, different mutations in the proteolytic and signaling pathways and the proto-oncogenes, oncogenes, and the tumor suppressor genes are made possible through cellular biochemistry and some cancer-stimulating environmental factors. The accumulated findings show that S-nitrosylation of cathepsins under the influence of NO-donors can prevent the invasion of cancer and cause cancer cell death by blocking the activity of cathepsins as well as the major denitrosylase systems using a multi-way approach. Faced with a conundrum of how to fill the gap between the dodging of established cancer hallmarks with cathepsin activity and gaining appropriate research/clinical accreditation using our hypothesis, the scope of this review also explores the interplay and crosstalk between S-nitrosylation and S-(de)nitrosylation of this protease and highlights the utility of charging thioredoxin (Trx) reductase inhibitors, low-molecular-weight dithiols, and Trx mimetics using efficient drug delivery system to prevent the denitrosylation or regaining of cathepsin activity in vivo. In foresight, this raises the prospect that drugs or novel compounds that target cathepsins taking all these factors into consideration could be deployed as alternative or even better treatments for cancer, though further research is needed to ascertain the safety, efficiency and effectiveness of this approach.
Collapse
|
20
|
Strasenburg W, Jóźwicki J, Durślewicz J, Kuffel B, Kulczyk MP, Kowalewski A, Grzanka D, Drewa T, Adamowicz J. Tumor Cell-Induced Platelet Aggregation as an Emerging Therapeutic Target for Cancer Therapy. Front Oncol 2022; 12:909767. [PMID: 35814405 PMCID: PMC9259835 DOI: 10.3389/fonc.2022.909767] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor cells have the ability to induce platelet activation and aggregation. This has been documented to be involved in tumor progression in several types of cancers, such as lung, colon, breast, pancreatic, ovarian, and brain. During the process, platelets protect circulating tumor cells from the deleterious effects of shear forces, shield tumor cells from the immune system, and provide growth factors, facilitating metastatic spread and tumor growth at the original site as well as at the site of metastasis. Herein, we present a wider view on the induction of platelet aggregation by specific factors primarily developed by cancer, including coagulation factors, adhesion receptors, growth factors, cysteine proteases, matrix metalloproteinases, glycoproteins, soluble mediators, and selectins. These factors may be presented on the surface of tumor cells as well as in their microenvironment, and some may trigger more than just one simple receptor-ligand mechanism. For a better understanding, we briefly discuss the physiological role of the factors in the platelet activation process, and subsequently, we provide scientific evidence and discuss their potential role in the progression of specific cancers. Targeting tumor cell-induced platelet aggregation (TCIPA) by antiplatelet drugs may open ways to develop new treatment modalities. On the one hand, it may affect patients' prognosis by enhancing known therapies in advanced-stage tumors. On the other hand, the use of drugs that are mostly easily accessible and widely used in general practice may be an opportunity to propose an unparalleled antitumor prophylaxis. In this review, we present the recent discoveries of mechanisms by which cancer cells activate platelets, and discuss new platelet-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Wiktoria Strasenburg
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Jakub Jóźwicki
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Justyna Durślewicz
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Błażej Kuffel
- Department of General and Oncological Urology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Martyna Parol Kulczyk
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Adam Kowalewski
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Tomasz Drewa
- Department of General and Oncological Urology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Jan Adamowicz
- Department of General and Oncological Urology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
21
|
Al‐Qahtani SM, Gadalla SE, Guo M, Ericsson C, Hägerstrand D, Nistér M. The association between Annexin A2 and epithelial cell adhesion molecule in breast cancer cells. Cancer Rep (Hoboken) 2022; 5:e1498. [PMID: 34240826 PMCID: PMC9124509 DOI: 10.1002/cnr2.1498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/05/2021] [Accepted: 06/14/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The epithelial cell adhesion molecule (EpCAM) is a type I transmembrane and glycosylated protein, which is overexpressed in many neoplasms. However, EpCAM has no known ligand partners and the mechanisms by which it functions are not fully understood. AIM This study was performed to discover novel partners of EpCAM, which may provide a better understanding of its functions. METHODS The membrane fraction of the ERα+ noninvasive breast cancer cell line ZR-75-1 and MCF-7 was extracted and followed by co-immunoprecipitation of EpCAM using C-10, a mouse monoclonal antibody raised against amino acids 24-93 of the EpCAM molecule. As a negative control, MDA-MB-231 and Hs578T were used since they express a negligible amount of EpCAM and are known as EpCAM-/low ERα-/low invasive and tumorigenic breast cancer cell lines. RESULTS Annexin A2 (ANXA2) was found to be selectively and differentially co-immunoprecipitated with EpCAM in the ERα+ breast cancer cells MCF-7 and ZR-75-1. ANXA2 is a multifunctional protein and known to act as a co-receptor for tissue plasminogen activator (tPA) on the surface of endothelial and cancer cells, thereby affecting fibrinolytic activity and neoangiogenesis as well as invasive and metastatic properties. In this study, the association between EpCAM and ANXA2 was found to affect the activity of tPA. CONCLUSION This study concludes that ANXA2 co-localizes with EpCAM at the plasma membrane, and the co-localization may have functional implications. Data suggest that EpCAM supports ANXA2 to function as a co-receptor for the tPA, and that EpCAM has a regulatory function on the expression and subcellular localization of ANXA2.
Collapse
Affiliation(s)
- Saad Misfer Al‐Qahtani
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
- Department of Pathology, College of Medicine and Najran University HospitalNajran UniversityNajranSaudi Arabia
| | | | - Min Guo
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
| | | | | | - Monica Nistér
- Department of Oncology‐PathologyKarolinska InstitutetStockholmSweden
| |
Collapse
|
22
|
Rodriguez-Rios M, Megia-Fernandez A, Norman DJ, Bradley M. Peptide probes for proteases - innovations and applications for monitoring proteolytic activity. Chem Soc Rev 2022; 51:2081-2120. [PMID: 35188510 DOI: 10.1039/d1cs00798j] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Proteases are excellent biomarkers for a variety of diseases, offer multiple opportunities for diagnostic applications and are valuable targets for therapy. From a chemistry-based perspective this review discusses and critiques the most recent advances in the field of substrate-based probes for the detection and analysis of proteolytic activity both in vitro and in vivo.
Collapse
Affiliation(s)
- Maria Rodriguez-Rios
- EaStCHEM School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ Edinburgh, UK.
| | - Alicia Megia-Fernandez
- EaStCHEM School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ Edinburgh, UK.
| | - Daniel J Norman
- Technical University of Munich, Trogerstrasse, 30, 81675, Munich, Germany
| | - Mark Bradley
- EaStCHEM School of Chemistry, University of Edinburgh, David Brewster Road, EH9 3FJ Edinburgh, UK.
| |
Collapse
|
23
|
Sheyi R, de la Torre BG, Albericio F. Linkers: An Assurance for Controlled Delivery of Antibody-Drug Conjugate. Pharmaceutics 2022; 14:pharmaceutics14020396. [PMID: 35214128 PMCID: PMC8874516 DOI: 10.3390/pharmaceutics14020396] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 12/15/2022] Open
Abstract
As one of the major therapeutic options for cancer treatment, chemotherapy has limited selectivity against cancer cells. Consequently, this therapeutic strategy offers a small therapeutic window with potentially high toxicity and thus limited efficacy of doses that can be tolerated by patients. Antibody-drug conjugates (ADCs) are an emerging class of anti-cancer therapeutic drugs that can deliver highly cytotoxic molecules directly to cancer cells. To date, twelve ADCs have received market approval, with several others in clinical stages. ADCs have become a powerful class of therapeutic agents in oncology and hematology. ADCs consist of recombinant monoclonal antibodies that are covalently bound to cytotoxic chemicals via synthetic linkers. The linker has a key role in ADC outcomes because its characteristics substantially impact the therapeutic index efficacy and pharmacokinetics of these drugs. Stable linkers and ADCs can maintain antibody concentration in blood circulation, and they do not release the cytotoxic drug before it reaches its target, thus resulting in minimum off-target effects. The linkers used in ADC development can be classified as cleavable and non-cleavable. The former, in turn, can be grouped into three types: hydrazone, disulfide, or peptide linkers. In this review, we highlight the various linkers used in ADC development and their design strategy, release mechanisms, and future perspectives.
Collapse
Affiliation(s)
- Rotimi Sheyi
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Beatriz G. de la Torre
- Kwazulu-Natal Research Innovation and Sequencing Platform (KRISP), College of Health Sciences, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: (B.G.d.l.T.); (F.A.); Tel.: +27-614-047-528 (B.G.d.l.T.); +27-6140-09144 (F.A.)
| | - Fernando Albericio
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban 4001, South Africa;
- Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
- Networking Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Department of Organic Chemistry, University of Barcelona, 08028 Barcelona, Spain
- Correspondence: (B.G.d.l.T.); (F.A.); Tel.: +27-614-047-528 (B.G.d.l.T.); +27-6140-09144 (F.A.)
| |
Collapse
|
24
|
Sharma C, Kang SC. Molecular dynamic simulation (MDS) and in vitro cathepsin-B inhibitory activity of decrusin angelate, ibuprofen, and thymol. Nat Prod Res 2022; 36:1020-1025. [PMID: 33148043 DOI: 10.1080/14786419.2020.1843030] [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: 10/23/2022]
Abstract
Attenuation of cathepsin B (CATB) proteolytic activity and/or inhibition serves as a potential therapeutic target in cancer metastasis. Herein, we determined the specificity of FDA approved potential anti-cancer natural flavonoid decursinol angelate (DA), thymol (TH) and a propionic acid derivative ibuprofen (IB), for the inactivation of CATB. We used enzymatic assay, computational and in vitro methods for the identification of the best candidate. Out of these we found DA can inhibit CATB with lowest IC50 measured after one hour of incubation using Z-Phe-Arg-4MβNA (BANA) as a substrate. Docking analysis suggested favorable interaction of DA with the catalytic site residues (GLN23, CYS26, HIS110, HIS111) of CATB (PDB Id: 1HUC) were responsible for the inhibition of its proteolytic activity. Additionally, in vitro quantification with human colorectal carcinoma (HCT 116) revealed, DA rapidly inactivates CATB as compared with commercial synthetic inhibitor CA074 with no cellular toxicity towards normal colon cells (CCD 841).
Collapse
Affiliation(s)
- Chanchal Sharma
- Department of Biotechnology, College of Engineering, Daegu University, Gyeongsan, Republic of Korea
| | - Sun Chul Kang
- Department of Biotechnology, College of Engineering, Daegu University, Gyeongsan, Republic of Korea
| |
Collapse
|
25
|
Kos J, Mitrović A, Perišić Nanut M, Pišlar A. Lysosomal peptidases – Intriguing roles in cancer progression and neurodegeneration. FEBS Open Bio 2022; 12:708-738. [PMID: 35067006 PMCID: PMC8972049 DOI: 10.1002/2211-5463.13372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/04/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022] Open
Abstract
Lysosomal peptidases are hydrolytic enzymes capable of digesting waste proteins that are targeted to lysosomes via endocytosis and autophagy. Besides intracellular protein catabolism, they play more specific roles in several other cellular processes and pathologies, either within lysosomes, upon secretion into the cell cytoplasm or extracellular space, or bound to the plasma membrane. In cancer, lysosomal peptidases are generally associated with disease progression, as they participate in crucial processes leading to changes in cell morphology, signaling, migration, and invasion, and finally metastasis. However, they can also enhance the mechanisms resulting in cancer regression, such as apoptosis of tumor cells or antitumor immune responses. Lysosomal peptidases have also been identified as hallmarks of aging and neurodegeneration, playing roles in oxidative stress, mitochondrial dysfunction, abnormal intercellular communication, dysregulated trafficking, and the deposition of protein aggregates in neuronal cells. Furthermore, deficiencies in lysosomal peptidases may result in other pathological states, such as lysosomal storage disease. The aim of this review was to highlight the role of lysosomal peptidases in particular pathological processes of cancer and neurodegeneration and to address the potential of lysosomal peptidases in diagnosing and treating patients.
Collapse
Affiliation(s)
- Janko Kos
- University of Ljubljana Faculty of Pharmacy Aškerčeva 7 1000 Ljubljana Slovenia
- Jožef Stefan Institute Department of Biotechnology Jamova 39 1000 Ljubljana Slovenia
| | - Ana Mitrović
- Jožef Stefan Institute Department of Biotechnology Jamova 39 1000 Ljubljana Slovenia
| | - Milica Perišić Nanut
- Jožef Stefan Institute Department of Biotechnology Jamova 39 1000 Ljubljana Slovenia
| | - Anja Pišlar
- University of Ljubljana Faculty of Pharmacy Aškerčeva 7 1000 Ljubljana Slovenia
| |
Collapse
|
26
|
Kumar AA, Buckley BJ, Ranson M. The Urokinase Plasminogen Activation System in Pancreatic Cancer: Prospective Diagnostic and Therapeutic Targets. Biomolecules 2022; 12:152. [PMID: 35204653 PMCID: PMC8961517 DOI: 10.3390/biom12020152] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy that features high recurrence rates and the poorest prognosis of all solid cancers. The urokinase plasminogen activation system (uPAS) is strongly implicated in the pathophysiology and clinical outcomes of patients with pancreatic ductal adenocarcinoma (PDAC), which accounts for more than 90% of all pancreatic cancers. Overexpression of the urokinase-type plasminogen activator (uPA) or its cell surface receptor uPAR is a key step in the acquisition of a metastatic phenotype via multiple mechanisms, including the increased activation of cell surface localised plasminogen which generates the serine protease plasmin. This triggers multiple downstream processes that promote tumour cell migration and invasion. Increasing clinical evidence shows that the overexpression of uPA, uPAR, or of both is strongly associated with worse clinicopathological features and poor prognosis in PDAC patients. This review provides an overview of the current understanding of the uPAS in the pathogenesis and progression of pancreatic cancer, with a focus on PDAC, and summarises the substantial body of evidence that supports the role of uPAS components, including plasminogen receptors, in this disease. The review further outlines the clinical utility of uPAS components as prospective diagnostic and prognostic biomarkers for PDAC, as well as a rationale for the development of novel uPAS-targeted therapeutics.
Collapse
Affiliation(s)
- Ashna A. Kumar
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Benjamin J. Buckley
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; (A.A.K.); (B.J.B.)
- School of Chemistry and Molecular Biosciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
27
|
Van de Walle T, Cools L, Mangelinckx S, D'hooghe M. Recent contributions of quinolines to antimalarial and anticancer drug discovery research. Eur J Med Chem 2021; 226:113865. [PMID: 34655985 DOI: 10.1016/j.ejmech.2021.113865] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 09/01/2021] [Accepted: 09/20/2021] [Indexed: 12/28/2022]
Abstract
Quinoline, a privileged scaffold in medicinal chemistry, has always been associated with a multitude of biological activities. Especially in antimalarial and anticancer research, quinoline played (and still plays) a central role, giving rise to the development of an array of quinoline-containing pharmaceuticals in these therapeutic areas. However, both diseases still affect millions of people every year, pointing to the necessity of new therapies. Quinolines have a long-standing history as antimalarial agents, but established quinoline-containing antimalarial drugs are now facing widespread resistance of the Plasmodium parasite. Nevertheless, as evidenced by a massive number of recent literature contributions, they are still of great value for future developments in this field. On the other hand, the number of currently approved anticancer drugs containing a quinoline scaffold are limited, but a strong increase and interest in quinoline compounds as potential anticancer agents can be seen in the last few years. In this review, a literature overview of recent contributions made by quinoline-containing compounds as potent antimalarial or anticancer agents is provided, covering publications between 2018 and 2020.
Collapse
Affiliation(s)
- Tim Van de Walle
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000, Ghent, Belgium
| | - Lore Cools
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000, Ghent, Belgium
| | - Sven Mangelinckx
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000, Ghent, Belgium
| | - Matthias D'hooghe
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000, Ghent, Belgium.
| |
Collapse
|
28
|
He P, Sheng J, Qi J, Bai X, Li J, Wang F, Yuan Y, Zheng X. STAT3-induced NCK1 elevation promotes migration of triple-negative breast cancer cells via regulating ERK1/2 signaling. Mol Biol Rep 2021; 49:267-278. [PMID: 34846647 DOI: 10.1007/s11033-021-06868-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Noncatalytic region of tyrosine kinase 1 (NCK1) plays a key role in extracellular matrix degradation, which is required for the metastasis of triple-negative breast cancer (TNBC). However, the role NCK1 plays in the metastatic progression of TNBC is unknown. METHODS AND RESULTS Based on online databases, NCK1 was found to be highly expressed in TNBC as compared to normal breast-like subjects, which was also confirmed using TNBC cells and a tissue microarray. NCK1 expression gradually decreased with increased tumor stage. High NCK1 expression displayed a poor prognosis in lymph node-positive metastatic TNBC patients, but not in lymph node-negative patients. Using transwell assays and immunoblotting, we confirmed that NCK1 overexpression promoted, while NCK1 downregulation inhibited migration capabilities, as well as the expression of matrix metalloproteinases (MMP2/9), uridylyl phosphate adenosine, and plasminogen activator inhibitor-1 in TNBC cells. Mechanistically, NCK1 upregulation mediated the activation of MMP2/9 through ERK1/2 activity. Signal transducer and activator of transcription 3 (STAT3) was positively correlated with NCK1. STAT3 could directly bind to the promoter region of NCK1 to promote its expression and was accompanied by the elevation of MMP2/9 and ERK1/2 signaling, which were partially abolished by the knockdown of NCK1 in TNBC cells. CONCLUSIONS NCK1 may serve as a diagnostic and prognostic marker of metastatic TNBC. STAT3 upregulation promoted the expression of NCK1, which subsequently induced the migration and activity of MMPs in a ERK1/2 signaling-dependent manner in TNBC cells. NCK1 is a promising target for improving TNBC migration.
Collapse
Affiliation(s)
- Peina He
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Jianyun Sheng
- Department of Gynecotokology, Pingdingshan First People's Hospital, Pingdingshan, 410402, China
| | - Jinxu Qi
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Xianguang Bai
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Jiaxin Li
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Fubao Wang
- Department of Gynecotokology, Pingdingshan First People's Hospital, Pingdingshan, 410402, China
| | - Yamin Yuan
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Xinhua Zheng
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China.
| |
Collapse
|
29
|
Song Y, Wright JG, Anderson MJ, Rajendran S, Ren Z, Hua DH, Koehne JE, Meyyappan M, Li J. Quantitative Detection of Cathepsin B Activity in Neutral pH Buffers Using Gold Microelectrode Arrays: Toward Direct Multiplex Analyses of Extracellular Proteases in Human Serum. ACS Sens 2021; 6:3621-3631. [PMID: 34546741 DOI: 10.1021/acssensors.1c01175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Proteases are critical signaling molecules and prognostic biomarkers for many diseases including cancer. There is a strong demand for multiplex bioanalytical techniques that can rapidly detect the activity of extracellular proteases with high sensitivity and specificity. This study demonstrates an activity-based electrochemical biosensor of a 3 × 3 gold microelectrode array for the detection of cathepsin B activity in human serum diluted in a neutral buffer. Proteolysis of ferrocene-labeled peptide substrates functionalized on 200 × 200 μm microelectrodes is measured simultaneously over the nine channels by AC voltammetry. The protease activity is represented by the inverse of the exponential decay time constant (1/τ), which equals to (kcat/KM)[CB] based on the Michaelis-Menten model. An enhanced activity of the recombinant human cathepsin B (rhCB) is observed in a low-ionic-strength phosphate buffer at pH = 7.4, giving a very low limit of detection of 8.49 × 10-4 s-1 for activity and 57.1 pM for the active rhCB concentration that is comparable to affinity-based enzyme-linked immunosorbent assay (ELISA). The cathepsin B presented in the human serum sample is validated by ELISA, which mainly detects the inactive proenzyme, while the electrochemical biosensor specifically measures the active cathepsin B and shows significantly higher decay rates when rhCB and human serum are activated. Analyses of the kinetic electrochemical measurements with spiked active cathepsin B in human serum provide further assessment of the protease activity in the complex sample. This study lays the foundation to develop the gold microelectrode array into a multiplex biosensor for rapid detection of the activity of extracellular proteases toward cancer diagnosis and treatment assessment.
Collapse
Affiliation(s)
- Yang Song
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Jestin Gage Wright
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Morgan J. Anderson
- NASA Ames Research Center, Moffett Field, California 94035, United States
| | - Sabari Rajendran
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Zhaoyang Ren
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Duy H. Hua
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| | - Jessica E. Koehne
- NASA Ames Research Center, Moffett Field, California 94035, United States
| | - M. Meyyappan
- NASA Ames Research Center, Moffett Field, California 94035, United States
| | - Jun Li
- Department of Chemistry, Kansas State University, Manhattan, Kansas 66506, United States
| |
Collapse
|
30
|
Huang L, Bai F, Zhang Y, Zhang S, Jin T, Wei X, Zhou X, Lin M, Xie Y, He C, Lin Q, Xie T, Ding Y. Preliminary study of genome-wide association identified novel susceptibility genes for thyroid-related hormones in Chinese population. Genes Genomics 2021; 44:1031-1038. [PMID: 34533693 DOI: 10.1007/s13258-021-01165-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/11/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Thyroid hormones are critical regulators of metabolism, development and growth in mammals. However, the genetic association of thyroid-related hormones in the Chinese Han population is not fully understood. OBJECTIVE We aimed to identify the genetic loci associated with circulating thyroid-related hormones concentrations in the healthy Chinese Han population. METHODS Genotyping was performed in 124 individuals using Applied Biosystems™ Axiom™ PMDA, and 796,288 single nucleotide polymorphisms (SNPs) were available for the GWAS analysis. For replication, eleven SNPs were selected as candidate loci for genotyping by Agena MassARRAY platform in additional samples (313 subjects). The values of p < 5 × 10- 6 suggest a suggestively significant genome-wide association with circulating thyroid-related hormones concentrations. RESULTS We identified that rs11178277 (PTPRB, p = 4.88 × 10- 07) and rs7320337 (LMO7DN-KCTD12, p = 1.22 × 10- 06) were associated with serum FT3 level. Three SNPs (rs4850041 in LOC105373394-LINC01249: p = 3.55 × 10- 06, rs6867291 in LINC02208: p = 2.40 × 10- 06 and rs79508321 in WWOX: p = 3.35 × 10- 06) were related to circulating T3 level. Rs12474167 (LOC105373394-LINC01249, p = 1.65 × 10- 06) and rs1864553 (IWS1, p = 2.00 × 10- 06) were associated with circulating T4 concentration. The association with TGA concentration was for rs17163542 in DISP1 (p = 3.46 × 10- 06) and rs12601151 in NOG-C17orf67 (p = 2.72 × 10- 07). Two genome-level significant SNPs (rs2114707 in LINC01314, p = 1.69 × 10- 06 and rs12601151, p = 1.41 × 10- 07) associated with serum TMA concentration were identified. Moreover, rs6083269 (CST1-CST2, p = 3.36 × 10- 06) was a significant locus for circulating TSH level. In replication, rs12601151 in NOG-C17orf67 was still associated with serum TGA level (p = 0.012). CONCLUSIONS The GWAS reported 11 new suggestively significant loci associated with circulating thyroid-related hormones levels among the Chinese Han population. These findings represented suggestively biological candidates for circulating thyroid-related hormones levels and provided new insights into the mechanisms of regulating serum TGA concentration.
Collapse
Affiliation(s)
- Liang Huang
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China
- Xincun Central Health Center, Lingshui Li Autonomous County, Lingshui, 572426, Hainan, People's Republic of China
| | - Fenghua Bai
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China
- Science and Education Office, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China
| | - Yutian Zhang
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China
- Department of General Practice, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China
| | - Shanshan Zhang
- Xi'an 21st Century Biological Science and Technology Co., Ltd, Xi'an, 712000, Shaanxi, People's Republic of China
| | - Tianbo Jin
- Xi'an 21st Century Biological Science and Technology Co., Ltd, Xi'an, 712000, Shaanxi, People's Republic of China
| | - Xingwei Wei
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China
- Department of General Practice, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China
| | - Xiaoli Zhou
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China
- Department of General Practice, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China
| | - Mei Lin
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China
- Department of General Practice, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China
| | - Yufei Xie
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China
- Department of General Practice, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China
| | - Chanyi He
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China
- Department of General Practice, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China
| | - Qi Lin
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China
- Department of General Practice, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China
| | - Tian Xie
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China.
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China.
| | - Yipeng Ding
- Hainan Affiliated Hospital of Hainan Medical University, #19, Xiuhua Road, Xiuying District, Haikou, 570311, Hainan, People's Republic of China.
- Department of General Practice, Hainan General Hospital, Haikou, 570311, Hainan, People's Republic of China.
| |
Collapse
|
31
|
Scott J, Deng Q, Vendrell M. Near-Infrared Fluorescent Probes for the Detection of Cancer-Associated Proteases. ACS Chem Biol 2021; 16:1304-1317. [PMID: 34315210 PMCID: PMC8383269 DOI: 10.1021/acschembio.1c00223] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022]
Abstract
Proteases are enzymes capable of catalyzing protein breakdown, which is critical across many biological processes. There are several families of proteases, each of which perform key functions through the degradation of specific proteins. As our understanding of cancer improves, it has been demonstrated that several proteases can be overactivated during the progression of cancer and contribute to malignancy. Optical imaging systems that employ near-infrared (NIR) fluorescent probes to detect protease activity offer clinical promise, both for early detection of cancer as well as for the assessment of personalized therapy. In this Review, we review the design of NIR probes and their successful application for the detection of different cancer-associated proteases.
Collapse
Affiliation(s)
- Jamie
I. Scott
- Centre
for Inflammation Research, The University
of Edinburgh, EH16 4TJ Edinburgh, United Kingdom
| | - Qinyi Deng
- Centre
for Inflammation Research, The University
of Edinburgh, EH16 4TJ Edinburgh, United Kingdom
| | - Marc Vendrell
- Centre
for Inflammation Research, The University
of Edinburgh, EH16 4TJ Edinburgh, United Kingdom
| |
Collapse
|
32
|
Boosting the oxidase-like activity of platinum nanozyme in MBTH-TOOS chromogenic system for detection of trypsin and its inhibitor. Talanta 2021; 234:122647. [PMID: 34364456 DOI: 10.1016/j.talanta.2021.122647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 11/23/2022]
Abstract
Nanozymes, as a new type of artificial enzyme, have recently become a research hotspot in the field of catalysis and biomedicine. However, the application of nanozyme is limited by catalytic activity changes of different substrates and low specificity. This work shows that citrate-capped platinum nanoparticles (Cit-PtNPs) exhibit stronger oxidase-like activity than other platinum nanozymes at different pH when 3-methyl-2-benzothiazolinonehydrazone hydrochloride (MBTH) and n-ethyl-n- (2-hydroxy-3-sulfopropyl)-m-toluidine sodium salt (TOOS) were used as chromogenic substrates. This phenomenon has important reference value for different nanozymes to choose chromogenic substrates in catalysis. In MBTH-TOOS chromogenic system, MBTH (-NH) radical is first produced during the reaction through catalytic oxidation of Cit-PtNPs, which reacts with TOOS to produce a colorless compound. The blue-purple quinoid dye was produced through the dismutation of the colorless compound. The catalytic mechanism of the oxidase-like activity of Cit-PtNPs is that two-electron reduction process and four-electron reduction process are simultaneously carried out in the catalytic process. Furthermore, to solve the problem of low specificity of metal nanozymes, protamine is designed as aggregation promoter of Cit-PtNPs and the specifichydrolysis substrate of trypsin. In this work, it can achieve one-step detection of trypsin by the boosting oxidase activity of Cit-PtNPs at pH8. The catalytic activity of Cit-PtNPs is proportional to the concentration of trypsin. The linear range for trypsin is 1.0-70.0 ngmL-1 and the limit of detection is measured to be 0.6 ngmL-1. This novel method has also been successfully applied to the detection of inhibitors and trypsin in urine samples.
Collapse
|
33
|
Hershberger KK, Gauger AJ, Bronstein LM. Utilizing Stimuli Responsive Linkages to Engineer and Enhance Polymer Nanoparticle-Based Drug Delivery Platforms. ACS APPLIED BIO MATERIALS 2021; 4:4720-4736. [PMID: 35007022 DOI: 10.1021/acsabm.1c00351] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The devastating nature of cancer continues to be one of the leading causes of death in the world. Chemotherapy is among the most common forms of cancer treatment but comes with a host of adverse effects caused by the therapeutic agents damaging healthy tissue and organs. To limit these side effects, scientists have been designing stimuli responsive drug delivery vessels for targeted release. This Review focuses on the incorporation of stimuli responsive linkages in targeted drug delivery systems to enhance therapeutic efficiency. These platforms are primarily employed to control the distribution of anticancer agents in the body to reduce the adverse side effects caused by their toxicities. We will outline how drug delivery vessels are constructed so that exposure to select environmental and external stimuli releases the enclosed drug only at the target site. Stimuli responsive components are integrated within drug delivery vessels in the form of cross-linkers, polymers, and surface modifications. The changes, these moieties undergo upon stimuli exposure, cascade into larger scale alterations to the platforms, resulting in complete disassembly, reversible morphological variations, and enhanced cellular uptake. The ability for these modes of delivery to be initiated exclusively under stimuli exposure allows for release of toxic therapeutic agents to be confined only to the affected area.
Collapse
Affiliation(s)
- Kian K Hershberger
- Indiana University, Department of Chemistry, Bloomington, 800 East Kirkwood Avenue, Indiana 47405, United States
| | - Andrew J Gauger
- Indiana University, Department of Chemistry, Bloomington, 800 East Kirkwood Avenue, Indiana 47405, United States
| | - Lyudmila M Bronstein
- Indiana University, Department of Chemistry, Bloomington, 800 East Kirkwood Avenue, Indiana 47405, United States.,A.N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, 28 Vavilov Street, Moscow, 119991 Russia.,King Abdulaziz University, Faculty of Science, Department of Physics, P.O. Box 80303, Jeddah 21589, Saudi Arabia
| |
Collapse
|
34
|
Protease Substrate-Independent Universal Assay for Monitoring Digestion of Native Unmodified Proteins. Int J Mol Sci 2021; 22:ijms22126362. [PMID: 34198602 PMCID: PMC8231992 DOI: 10.3390/ijms22126362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022] Open
Abstract
Proteases are a group of enzymes with a catalytic function to hydrolyze peptide bonds of proteins. Proteases regulate the activity, signaling mechanism, fate, and localization of many proteins, and their dysregulation is associated with various pathological conditions. Proteases have been identified as biomarkers and potential therapeutic targets for multiple diseases, such as acquired immunodeficiency syndrome, cardiovascular diseases, osteoporosis, type 2 diabetes, and cancer, where they are essential to disease progression. Thus, protease inhibitors and inhibitor-like molecules are interesting drug candidates. To study proteases and their substrates and inhibitors, simple, rapid, and sensitive protease activity assays are needed. Existing fluorescence-based assays enable protease monitoring in a high-throughput compatible microtiter plate format, but the methods often rely on either molecular labeling or synthetic protease targets that only mimic the hydrolysis site of the true target proteins. Here, we present a homogenous, label-free, and time-resolved luminescence utilizing the protein-probe method to assay proteases with native and denatured substrates at nanomolar sensitivity. The developed protein-probe method is not restricted to any single protein or protein target class, enabling digestion and substrate fragmentation studies with the natural unmodified substrate proteins. The versatility of the assay for studying protease targets was shown by monitoring the digestion of a substrate panel with different proteases. These results indicate that the protein-probe method not only monitors the protease activity and inhibition, but also studies the substrate specificity of individual proteases.
Collapse
|
35
|
Sá JDO, Trino LD, Oliveira AK, Lopes AFB, Granato DC, Normando AGC, Santos ES, Neves LX, Carnielli CM, Paes Leme AF. Proteomic approaches to assist in diagnosis and prognosis of oral cancer. Expert Rev Proteomics 2021; 18:261-284. [PMID: 33945368 DOI: 10.1080/14789450.2021.1924685] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Oral squamous cell carcinoma (OSCC) ranks among the top 10 leading causes of cancer worldwide, with 5-year survival rate of about 50%, high lymph node metastasis, and relapse rates. The OSCC diagnosis, prognosis, and treatment are mostly based on the clinical TNM classification. There is an urgent need for the discovery of biomarkers and therapeutic targets to assist in the clinical decision-making process.Areas covered: We summarize proteomic studies of the OSCC tumor, immune microenvironment, potential liquid biopsy sites, and post-translational modifications trying to retrieve information in the discovery and verification or (pre)validation phases. The search strategy was based on the combination of MeSH terms and expert refinement.Expert opinion: Untargeted combined with targeted proteomics are strategies that provide reliable and reproducible quantitation of proteins and are the methods of choice of many groups worldwide. Undoubtedly, proteomics has been contributing to the understanding of OSCC progression and uncovers potential candidates as biomarker or therapeutic targets. Nevertheless, none of these targets are available in the clinical practice yet. The scientific community needs to overcome the limitations by investing in robust experimental designs to strengthen the value of the findings, leveraging the translation of knowledge, and further supporting clinical decisions.
Collapse
Affiliation(s)
- Jamile De Oliveira Sá
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil.,Departamento De Diagnóstico Oral, Faculdade De Odontologia De Piracicaba, Universidade Estadual De Campinas (UNICAMP), Piracicaba, Brazil
| | - Luciana Daniele Trino
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Ana Karina Oliveira
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Ariane Fidelis Busso Lopes
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Daniela Campos Granato
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Ana Gabriela Costa Normando
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil.,Departamento De Diagnóstico Oral, Faculdade De Odontologia De Piracicaba, Universidade Estadual De Campinas (UNICAMP), Piracicaba, Brazil
| | - Erison Santana Santos
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil.,Departamento De Diagnóstico Oral, Faculdade De Odontologia De Piracicaba, Universidade Estadual De Campinas (UNICAMP), Piracicaba, Brazil
| | - Leandro Xavier Neves
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Carolina Moretto Carnielli
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| | - Adriana Franco Paes Leme
- Laboratório Nacional De Biociências (Lnbio), Centro Nacional De Pesquisa Em Energia E Materiais (CNPEM), Campinas, Brazil
| |
Collapse
|
36
|
Zhang Z, Yue P, Lu T, Wang Y, Wei Y, Wei X. Role of lysosomes in physiological activities, diseases, and therapy. J Hematol Oncol 2021; 14:79. [PMID: 33990205 PMCID: PMC8120021 DOI: 10.1186/s13045-021-01087-1] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
Long known as digestive organelles, lysosomes have now emerged as multifaceted centers responsible for degradation, nutrient sensing, and immunity. Growing evidence also implicates role of lysosome-related mechanisms in pathologic process. In this review, we discuss physiological function of lysosomes and, more importantly, how the homeostasis of lysosomes is disrupted in several diseases, including atherosclerosis, neurodegenerative diseases, autoimmune disorders, pancreatitis, lysosomal storage disorders, and malignant tumors. In atherosclerosis and Gaucher disease, dysfunction of lysosomes changes cytokine secretion from macrophages, partially through inflammasome activation. In neurodegenerative diseases, defect autophagy facilitates accumulation of toxic protein and dysfunctional organelles leading to neuron death. Lysosomal dysfunction has been demonstrated in pathology of pancreatitis. Abnormal autophagy activation or inhibition has been revealed in autoimmune disorders. In tumor microenvironment, malignant phenotypes, including tumorigenesis, growth regulation, invasion, drug resistance, and radiotherapy resistance, of tumor cells and behaviors of tumor-associated macrophages, fibroblasts, dendritic cells, and T cells are also mediated by lysosomes. Based on these findings, a series of therapeutic methods targeting lysosomal proteins and processes have been developed from bench to bedside. In a word, present researches corroborate lysosomes to be pivotal organelles for understanding pathology of atherosclerosis, neurodegenerative diseases, autoimmune disorders, pancreatitis, and lysosomal storage disorders, and malignant tumors and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Ziqi Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Pengfei Yue
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Tianqi Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| |
Collapse
|
37
|
Giribaldi J, Smith JJ, Schroeder CI. Recent developments in animal venom peptide nanotherapeutics with improved selectivity for cancer cells. Biotechnol Adv 2021; 50:107769. [PMID: 33989705 DOI: 10.1016/j.biotechadv.2021.107769] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/06/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023]
Abstract
Animal venoms are a rich source of bioactive peptides that efficiently modulate key receptors and ion channels involved in cellular excitability to rapidly neutralize their prey or predators. As such, they have been a wellspring of highly useful pharmacological tools for decades. Besides targeting ion channels, some venom peptides exhibit strong cytotoxic activity and preferentially affect cancer over healthy cells. This is unlikely to be driven by an evolutionary impetus, and differences in tumor cells and the tumor microenvironment are probably behind the serendipitous selectivity shown by some venom peptides. However, strategies such as bioconjugation and nanotechnologies are showing potential to improve their selectivity and potency, thereby paving the way to efficiently harness new anticancer mechanisms offered by venom peptides. This review aims to highlight advances in nano- and chemotherapeutic tools and prospective anti-cancer drug leads derived from animal venom peptides.
Collapse
Affiliation(s)
- Julien Giribaldi
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Jennifer J Smith
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Christina I Schroeder
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| |
Collapse
|
38
|
Wallin H, Hunaiti S, Abrahamson M. Externally added cystatin C reduces growth of A375 melanoma cells by increasing cell cycle time. FEBS Open Bio 2021; 11:1645-1658. [PMID: 33837649 PMCID: PMC8167853 DOI: 10.1002/2211-5463.13162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/31/2021] [Accepted: 04/08/2021] [Indexed: 12/21/2022] Open
Abstract
Some secreted cysteine protease inhibitors of the cystatin family appear to affect intracellular proteolysis and growth of human cells, as a result of internalization. Here, we studied the effects of external addition of the most abundant human cystatin, cystatin C, on viability and proliferation of cancer cells in culture. A dose‐dependent decrease in viable cells was seen for A375 melanoma, MCF‐7 breast cancer, and PC‐3 prostate cancer cells cultured in 1–5 µm cystatin C after 24 h. Real‐time assessment of growth rates in A375 cell cultures for 48 h by digital holographic microscopy showed an increased doubling time for cells cultured in the presence of 5 µm cystatin C (20.1 h) compared with control cells (14.7 h). A prolonged doubling time was already observed during the first 12 h, indicating a rapid general decrease in cell proliferation at the population level. Tracking of individual cells in phase holographic images showed that dividing cells incubated with 5 µm cystatin C underwent fewer mitoses during 48 h than control cells. In addition, the time between cell divisions was longer, especially for the first cell cycle. Incubation with the variant W106F‐cystatin C (with high cellular uptake rate) resulted in a lower number of viable cells and a prolonged doubling time than when cells were incubated with wild‐type cystatin C, but no effect was observed for (R24A,R25A)‐cystatin C (low cellular uptake). Thus, cystatin C causes prolonged cell division leading to decreased proliferation of melanoma cells, and internalization seems to be a prerequisite for this effect.
Collapse
Affiliation(s)
- Hanna Wallin
- Division of Clinical Chemistry & Pharmacology, Department of Laboratory Medicine, Lund University, Sweden
| | - Samar Hunaiti
- Division of Clinical Chemistry & Pharmacology, Department of Laboratory Medicine, Lund University, Sweden
| | - Magnus Abrahamson
- Division of Clinical Chemistry & Pharmacology, Department of Laboratory Medicine, Lund University, Sweden
| |
Collapse
|
39
|
Rana A, Bhatnagar S. Advancements in folate receptor targeting for anti-cancer therapy: A small molecule-drug conjugate approach. Bioorg Chem 2021; 112:104946. [PMID: 33989916 DOI: 10.1016/j.bioorg.2021.104946] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/17/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Targeted delivery combined with controlled release of drugs has a crucial role in future of personalized medicine. The majority of cancer drugs are intended to interfere with one or more cellular events. Anticancer agents can also be toxic to healthy cells, as healthy cells may also need to proliferate and avoid apoptosis. The focus of this review covers the principles, advantages, drawbacks and summarize criteria that must be met for design of small molecule-drug conjugates (SMDCs) to achieve the desired therapeutic potency with minimal toxicity. SMDCs are composed of a targeting ligand, a releasable bridge, a spacer, and a therapeutic payload. We summarize the criteria for the effective design that influences the selection of tumor specific receptor and optimum elements in the design of SMDCs. We also discuss the criteria for selecting the optimal therapeutic drug payload, spacer and linker. The linker chemistries and cleavage strategies are also discussed. Finally, we review the folate receptor targeting SMDCs that are in preclinical development and in clinical trials.
Collapse
Affiliation(s)
- Abhilash Rana
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| | - Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Sector125, Noida, Uttar Pradesh, India.
| |
Collapse
|
40
|
Plasmin and Plasminogen System in the Tumor Microenvironment: Implications for Cancer Diagnosis, Prognosis, and Therapy. Cancers (Basel) 2021; 13:cancers13081838. [PMID: 33921488 PMCID: PMC8070608 DOI: 10.3390/cancers13081838] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In this review, we present a detailed discussion of how the plasminogen-activation system is utilized by tumor cells in their unrelenting attack on the tissues surrounding them. Plasmin is an enzyme which is responsible for digesting several proteins that hold the tissues surrounding solid tumors together. In this process tumor cells utilize the activity of plasmin to digest tissue barriers in order to leave the tumour site and spread to other parts of the body. We specifically focus on the role of plasminogen receptor—p11 which is an important regulatory protein that facilitates the conversion of plasminogen to plasmin and by this means promotes the attack by the tumour cells on their surrounding tissues. Abstract The tumor microenvironment (TME) is now being widely accepted as the key contributor to a range of processes involved in cancer progression from tumor growth to metastasis and chemoresistance. The extracellular matrix (ECM) and the proteases that mediate the remodeling of the ECM form an integral part of the TME. Plasmin is a broad-spectrum, highly potent, serine protease whose activation from its precursor plasminogen is tightly regulated by the activators (uPA, uPAR, and tPA), the inhibitors (PAI-1, PAI-2), and plasminogen receptors. Collectively, this system is called the plasminogen activation system. The expression of the components of the plasminogen activation system by malignant cells and the surrounding stromal cells modulates the TME resulting in sustained cancer progression signals. In this review, we provide a detailed discussion of the roles of plasminogen activation system in tumor growth, invasion, metastasis, and chemoresistance with specific emphasis on their role in the TME. We particularly review the recent highlights of the plasminogen receptor S100A10 (p11), which is a pivotal component of the plasminogen activation system.
Collapse
|
41
|
Morais C, Rajandram R, Blakeney JS, Iyer A, Suen JY, Johnson DW, Gobe GC, Fairlie DP, Vesey DA. Expression of protease activated receptor-2 is reduced in renal cell carcinoma biopsies and cell lines. PLoS One 2021; 16:e0248983. [PMID: 33765016 PMCID: PMC7993771 DOI: 10.1371/journal.pone.0248983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/09/2021] [Indexed: 01/09/2023] Open
Abstract
Expression of the protease sensing receptor, protease activated receptor-2 (PAR2), is elevated in a variety of cancers and has been promoted as a potential therapeutic target. With the development of potent antagonists for this receptor, we hypothesised that they could be used to treat renal cell carcinoma (RCC). The expression of PAR2 was, therefore, examined in human RCC tissues and selected RCC cell lines. Histologically confirmed cases of RCC, together with paired non-involved kidney tissue, were used to produce a tissue microarray (TMA) and to extract total tissue RNA. Immunohistochemistry and qPCR were then used to assess PAR2 expression. In culture, RCC cell lines versus primary human kidney tubular epithelial cells (HTEC) were used to assess PAR2 expression by qPCR, immunocytochemistry and an intracellular calcium mobilization assay. The TMA revealed an 85% decrease in PAR2 expression in tumour tissue compared with normal kidney tissue. Likewise, qPCR showed a striking reduction in PAR2 mRNA in RCC compared with normal kidney. All RCC cell lines showed lower levels of PAR2 expression than HTEC. In conclusion, we found that PAR2 was reduced in RCC compared with normal kidney and is unlikely to be a target of interest in the treatment of this type of cancer.
Collapse
Affiliation(s)
- Christudas Morais
- Centre for Kidney Disease Research, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, Australia
| | - Retnagowri Rajandram
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Persekutuan, Malaysia
| | - Jade S. Blakeney
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Abishek Iyer
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jacky Y. Suen
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David W. Johnson
- Centre for Kidney Disease Research, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Glenda C. Gobe
- Centre for Kidney Disease Research, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - David P. Fairlie
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David A. Vesey
- Centre for Kidney Disease Research, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
- * E-mail:
| |
Collapse
|
42
|
van Leeuwen T, Araman C, Pieper Pournara L, Kampstra ASB, Bakkum T, Marqvorsen MHS, Nascimento CR, Groenewold GJM, van der Wulp W, Camps MGM, Janssen GMC, van Veelen PA, van Westen GJP, Janssen APA, Florea BI, Overkleeft HS, Ossendorp FA, Toes REM, van Kasteren SI. Bioorthogonal protein labelling enables the study of antigen processing of citrullinated and carbamylated auto-antigens. RSC Chem Biol 2021; 2:855-862. [PMID: 34212151 PMCID: PMC8190914 DOI: 10.1039/d1cb00009h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 11/21/2022] Open
Abstract
Proteolysis is fundamental to many biological processes. In the immune system, it underpins the activation of the adaptive immune response: degradation of antigenic material into short peptides and presentation thereof on major histocompatibility complexes, leads to activation of T-cells. This initiates the adaptive immune response against many pathogens. Studying proteolysis is difficult, as the oft-used polypeptide reporters are susceptible to proteolytic sequestration themselves. Here we present a new approach that allows the imaging of antigen proteolysis throughout the processing pathway in an unbiased manner. By incorporating bioorthogonal functionalities into the protein in place of methionines, antigens can be followed during degradation, whilst leaving reactive sidechains open to templated and non-templated post-translational modifications, such as citrullination and carbamylation. Using this approach, we followed and imaged the post-uptake fate of the commonly used antigen ovalbumin, as well as the post-translationally citrullinated and/or carbamylated auto-antigen vinculin in rheumatoid arthritis, revealing differences in antigen processing and presentation.
Collapse
Affiliation(s)
- Tyrza van Leeuwen
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Can Araman
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Linda Pieper Pournara
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Arieke S B Kampstra
- Department of Rheumatology, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - Thomas Bakkum
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Mikkel H S Marqvorsen
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Clarissa R Nascimento
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - G J Mirjam Groenewold
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Willemijn van der Wulp
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Marcel G M Camps
- Department of Immunology, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - George M C Janssen
- Center for Proteomics and Metabolomics, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - Gerard J P van Westen
- Computational Drug Discovery, Drug Discovery and Safety, LACDR, Leiden University Leiden The Netherlands
| | - Antonius P A Janssen
- Department of Molecular Physiology, Leiden Institute of Chemistry and the Oncode Institute, Leiden University Leiden The Netherlands
| | - Bogdan I Florea
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Herman S Overkleeft
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| | - Ferry A Ossendorp
- Department of Immunology, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - René E M Toes
- Department of Rheumatology, Leiden University Medical Center P.O. Box 9600 2300 RC Leiden The Netherlands
| | - Sander I van Kasteren
- Division of Bio-organic Synthesis, Leiden Institute of Chemistry and the Institute of Chemical Immunology, Leiden University Leiden The Netherlands
| |
Collapse
|
43
|
Varshosaz J, Fard MM, Mirian M, Hassanzadeh F. Targeted Nanoparticles for Co-delivery of 5-FU and Nitroxoline, a Cathepsin B Inhibitor, in HepG2 Cells of Hepatocellular Carcinoma. Anticancer Agents Med Chem 2021; 20:346-358. [PMID: 31566137 DOI: 10.2174/1871520619666190930124746] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/27/2019] [Accepted: 07/08/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND The first choice of treatment in Hepatocellular Carcinoma (HCC) is 5-fluorouracil (5-FU). Nitroxoline (NIT), a potent inhibitor of Cathepsin B, impairs tumor progression by decreased extracellular matrix degradation. The objective of the current project was designed to target nanoparticles for co-delivery of 5-FU and NIT in order to enhance the 5-FU cytotoxic effects and reduce the metastatic properties of HepG2 cells. METHODS 5-FU and NIT were loaded in chitosan-chondroitin nanoparticles. To target the CD44 receptors of HepG2 cells, Hyaluronic Acid (HA) was conjugated to the chondroitin by adipic acid dihydrazide and the conjugation was confirmed by FTIR and 1HNMR. After physicochemical characterization and optimization of the processing variables, MTT assay was done on HepG2 and NIH3T3 cell lines to determine the cytotoxic properties of HA targeted nanoparticles. Migration of the cells was studied to compare the co-delivery of the drugs with each drug alone. RESULTS The optimized nanoparticles showed the particle size of 244.7±16.3nm, PDI of 0.30±0.03, drug entrapment efficiency of 46.3±5.0% for 5-FU and 75.1±0.9% for NIT. The drug release efficiency up to 8 hours was about 37.6±0.9% for 5-FU and 62.9±0.7% for NIT. The co-delivery of 5-FU and NIT in targeted nanoparticles showed significantly more cytotoxicity than the mixture of the two free drugs, non-targeted nanoparticles or each drug alone and reduced the IC50 value of 5-FU from 3.31±0.65μg/ml to 0.17±0.03μg/ml and the migration of HepG2 cells was also reduced to five-fold. CONCLUSION Co-delivery of 5-FU and NIT by HA targeted chitosan-chondroitin nanoparticles may be promising in HCC.
Collapse
Affiliation(s)
- Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Monireh M Fard
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Mirian
- Department of Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farshid Hassanzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
44
|
Small Molecule-Based Prodrug Targeting Prostate Specific Membrane Antigen for the Treatment of Prostate Cancer. Cancers (Basel) 2021; 13:cancers13030417. [PMID: 33499427 PMCID: PMC7865627 DOI: 10.3390/cancers13030417] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 11/16/2022] Open
Abstract
Metastatic castration-resistant prostate cancer poses a serious clinical problem with poor outcomes and remains a deadly disease. New targeted treatment options are urgently needed. PSMA is highly expressed in prostate cancer and has been an attractive biomarker for the treatment of prostate cancer. In this study, we explored the feasibility of targeted delivery of an antimitotic drug, monomethyl auristatin E (MMAE), to tumor tissue using a small-molecule based PSMA lig-and. With the aid of Cy5.5, we found that a cleavable linker is vital for the antitumor activity of the ligand-drug conjugate and have developed a new PSMA-targeting prodrug, PSMA-1-VcMMAE. In in vitro studies, PSMA-1-VcMMAE was 48-fold more potent in killing PSMA-positive PC3pip cells than killing PSMA-negative PC3flu cells. In in vivo studies, PSMA-1-VcMMAE significantly inhibited tumor growth leading to prolonged animal survival in different animal models, including metastatic prostate cancer models. Compared to anti-PSMA antibody-MMAE conjugate (PSMA-ADC) and MMAE, PSMA-1-VcMMAE had over a 10-fold improved maximum tolerated dose, resulting in improved therapeutic index. The small molecule-drug conjugates reported here can be easily synthesized and are more cost efficient than anti-body-drug conjugates. The therapeutic profile of the PSMA-1-VcMMAE encourages further clin-ical development for the treatment of advanced prostate cancer.
Collapse
|
45
|
Ishida K, Nakamura Y, Ohta T, Oe Y. A Molecular Probe with Both Chromogenic and Fluorescent Units for Detecting Serine Proteases. Molecules 2021; 26:molecules26020482. [PMID: 33477543 PMCID: PMC7831087 DOI: 10.3390/molecules26020482] [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: 12/07/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 11/16/2022] Open
Abstract
A molecular probe with l-phenylalanine p-nitroanilide and l-lysin 4-methylcoumaryl-7-amide, in which these amino acid derivatives are connected through a succinic-acid spacer, was prepared. Trypsin and papain were detected by blue-fluorescence emission of generated 7-amino-4-methylcoumarin (AMC). α-Chymotrypsin and nattokinase were detected from both the blue-fluorescence emission of AMC and the UV absorbance of p-nitroaniline. In addition, different time courses of p-nitroaniline and AMC were observed between the reaction of P1 with α-chymotrypsin and that with nattokinase. In the case of nattokinase, both the fluorescence emission and UV absorbance slowly increased. In contrast, the increasing UV absorbance was saturated at the early stage of the reaction of the present probe with chymotrypsin, whereas the fluorescence emission continuously increased in the following stages.
Collapse
Affiliation(s)
| | | | | | - Yohei Oe
- Correspondence: ; Tel./Fax: +81-774-65-6505
| |
Collapse
|
46
|
Jmel MA, Aounallah H, Bensaoud C, Mekki I, Chmelař J, Faria F, M’ghirbi Y, Kotsyfakis M. Insights into the Role of Tick Salivary Protease Inhibitors during Ectoparasite-Host Crosstalk. Int J Mol Sci 2021; 22:E892. [PMID: 33477394 PMCID: PMC7831016 DOI: 10.3390/ijms22020892] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 02/07/2023] Open
Abstract
Protease inhibitors (PIs) are ubiquitous regulatory proteins present in all kingdoms. They play crucial tasks in controlling biological processes directed by proteases which, if not tightly regulated, can damage the host organism. PIs can be classified according to their targeted proteases or their mechanism of action. The functions of many PIs have now been characterized and are showing clinical relevance for the treatment of human diseases such as arthritis, hepatitis, cancer, AIDS, and cardiovascular diseases, amongst others. Other PIs have potential use in agriculture as insecticides, anti-fungal, and antibacterial agents. PIs from tick salivary glands are special due to their pharmacological properties and their high specificity, selectivity, and affinity to their target proteases at the tick-host interface. In this review, we discuss the structure and function of PIs in general and those PI superfamilies abundant in tick salivary glands to illustrate their possible practical applications. In doing so, we describe tick salivary PIs that are showing promise as drug candidates, highlighting the most promising ones tested in vivo and which are now progressing to preclinical and clinical trials.
Collapse
Affiliation(s)
- Mohamed Amine Jmel
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005 České Budějovice, Czech Republic; (M.A.J.); (C.B.); (I.M.)
| | - Hajer Aounallah
- Institut Pasteur de Tunis, Université de Tunis El Manar, LR19IPTX, Service d’Entomologie Médicale, Tunis 1002, Tunisia; (H.A.); (Y.M.)
- Innovation and Development Laboratory, Innovation and Development Center, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Chaima Bensaoud
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005 České Budějovice, Czech Republic; (M.A.J.); (C.B.); (I.M.)
| | - Imen Mekki
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005 České Budějovice, Czech Republic; (M.A.J.); (C.B.); (I.M.)
- Faculty of Science, University of South Bohemia in České Budějovice, 37005 České Budějovice, Czech Republic;
| | - Jindřich Chmelař
- Faculty of Science, University of South Bohemia in České Budějovice, 37005 České Budějovice, Czech Republic;
| | - Fernanda Faria
- Innovation and Development Laboratory, Innovation and Development Center, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Youmna M’ghirbi
- Institut Pasteur de Tunis, Université de Tunis El Manar, LR19IPTX, Service d’Entomologie Médicale, Tunis 1002, Tunisia; (H.A.); (Y.M.)
| | - Michalis Kotsyfakis
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005 České Budějovice, Czech Republic; (M.A.J.); (C.B.); (I.M.)
| |
Collapse
|
47
|
Xiao D, Zhao L, Xie F, Fan S, Liu L, Li W, Cao R, Li S, Zhong W, Zhou X. A bifunctional molecule-based strategy for the development of theranostic antibody-drug conjugate. Am J Cancer Res 2021; 11:2550-2563. [PMID: 33456559 PMCID: PMC7806464 DOI: 10.7150/thno.51232] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are being developed worldwide with the potential to revolutionize current cancer treatment strategies. Developing novel theranostic ADCs with therapeutic utility and imaging capability is an attractive and challenging subject that promises advances in the field of personalized medicine. In this work, we propose a bifunctional molecule-based strategy for the development of theranostic ADCs. Methods: We developed a theranostic ADC consisting of the anti-Her2 antibody Mil40, monomethyl auristatin E (MMAE) as the active payload, and a 7-amino-3-hydroxyethyl-coumarin (7-AHC)-based dipeptide linker, which functions as a novel bifunctional fluorescence probe that allows self-elimination cleavage in the presence of cathepsin B for payload release and fluorophore activation. The on-off fluorescence properties and the antitumor effect in vitro and in vivo were investigated. Results: A 48-fold fluorescence enhancement was observed within 1 h when the 7-AHC-based linker was exposed to cathepsin B. Cleavage upon exposure to cathepsin B allows MMAE and fluorophore intracellular release and the monitoring of MMAE distribution using confocal microscopy. Additionally, the newly developed ADC retains the advantages of traditional p-aminobenzyloxycarbonyl-containing ADCs, such as good stability (t1/2 > 7 days) and high activity in vitro (IC50 = 0.09-3.74 nM). Importantly, the theranostic ADC exhibited the equivalent antitumor efficacy to the marketed ADC T-DM1 in the classic breast cancer model. Conclusion: We suggest that the present strategy can be universally applied in all p-aminobenzyloxycarbonyl-containing ADCs. Overall, theranostic ADCs may play a role in developing new theranostic systems and promoting personalized medicine research.
Collapse
|
48
|
Potential urinary monitoring of the enhanced permeability and retention effect using MMP-2-responsive poly(ethylene glycol) derivatives. J Control Release 2021; 329:513-523. [DOI: 10.1016/j.jconrel.2020.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/21/2020] [Accepted: 09/02/2020] [Indexed: 01/04/2023]
|
49
|
Van de Walle T, Briand M, Mitrović A, Sosič I, Gobec S, Kos J, Persoons L, Daelemans D, De Jonghe S, Ubiparip Z, Desmet T, Van Hecke K, Mangelinckx S, D'hooghe M. Synthesis of Novel Nitroxoline Analogs with Potent Cathepsin B Exopeptidase Inhibitory Activity. ChemMedChem 2020; 15:2477-2490. [PMID: 32744405 DOI: 10.1002/cmdc.202000402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Indexed: 01/02/2023]
Abstract
Nitroxoline, a well-known antimicrobial agent, has been identified in several independent studies, and on different molecular targets, as a promising candidate to be repurposed for cancer treatment. One specific target of interest concerns cathepsin B, a lysosomal peptidase involved in the degradation of the extracellular matrix (ECM), leading to tumor invasion, metastasis and angiogenesis. However, dedicated optimization of the nitroxoline core is needed to actually deliver a nitroxoline-based antitumor drug candidate. Within that context, 34 novel nitroxoline analogs were synthesized and evaluated for their relative cathepsin B inhibitory activity, their antiproliferative properties and their antimicrobial activity. More than twenty analogs were shown to exert a similar or even slightly higher cathepsin B inhibitory activity compared to nitroxoline. The implemented modifications of the nitroxoline scaffold and the resulting SAR information can form an eligible basis for further optimization toward more potent cathepsin B inhibitors in the quest for a clinical nitroxoline-based antitumor agent.
Collapse
Affiliation(s)
- Tim Van de Walle
- SynbioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Marina Briand
- SynbioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Ana Mitrović
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000, Ljubljana, Slovenia
| | - Izidor Sosič
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Stanislav Gobec
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000, Ljubljana, Slovenia.,Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Leentje Persoons
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Dirk Daelemans
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Steven De Jonghe
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Zorica Ubiparip
- Centre for Synthetic Biology (CSB), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Tom Desmet
- Centre for Synthetic Biology (CSB), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Kristof Van Hecke
- Xstruct, Department of Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S3, 9000, Ghent, Belgium
| | - Sven Mangelinckx
- SynbioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Matthias D'hooghe
- SynbioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| |
Collapse
|
50
|
Wang CH, Wang LK, Wu CC, Chen ML, Kuo CY, Shyu RY, Tsai FM. Cathepsin V Mediates the Tazarotene-induced Gene 1-induced Reduction in Invasion in Colorectal Cancer Cells. Cell Biochem Biophys 2020; 78:483-494. [PMID: 32918681 DOI: 10.1007/s12013-020-00940-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/28/2020] [Indexed: 01/06/2023]
Abstract
Tazarotene-induced gene 1 (TIG1) is a retinoid acid receptor-responsive gene involved in cell differentiation and tumorigenesis. Aberrant methylation of CpG islands in the TIG1 promoter is found in multiple cancers. Currently, the exact mechanism underlying the anticancer effect of TIG1 is unknown. Here, we show that TIG1 interacts with cathepsin V (CTSV), which reduces CTSV stability and subsequently affects the production of activated urokinase-type plasminogen activator (uPA), an epithelial-mesenchymal transition-associated protein. Ectopic expression of CTSV increased the expression of activated uPA and the number of migrated and invaded cells, whereas ectopic TIG1 expression reversed the effects of CTSV on the uPA signaling pathway. Similar patterns in the production of activated uPA and number of migrated and invaded cells were also observed in TIG1-expressing and CTSV-knockdown cells. The results suggest that CTSV may participate in TIG1-regulated uPA activity and the associated downstream signaling pathway.
Collapse
Affiliation(s)
- Chun-Hua Wang
- Department of Dermatology, Taipei Tzuchi Hospital, Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan
- School of Medicine, Tzu Chi University, Hualien, 970, Taiwan
| | - Lu-Kai Wang
- Radiation Biology Core Laboratory, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Linkou, Taoyuan, 333, Taiwan
| | - Chang-Chieh Wu
- Department of Surgery, Tri-Service General Hospital Keelung Branch, National Defense Medical Center, Keelung, 202, Taiwan
| | - Mao-Liang Chen
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan
| | - Rong-Yaun Shyu
- Department of Internal Medicine, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan.
| | - Fu-Ming Tsai
- Department of Research, Taipei Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation, New Taipei City, 231, Taiwan.
| |
Collapse
|