1
|
Zhang J, Qiu Z, Zhang Y, Wang G, Hao H. Intracellular spatiotemporal metabolism in connection to target engagement. Adv Drug Deliv Rev 2023; 200:115024. [PMID: 37516411 DOI: 10.1016/j.addr.2023.115024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/05/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
The metabolism in eukaryotic cells is a highly ordered system involving various cellular compartments, which fluctuates based on physiological rhythms. Organelles, as the smallest independent sub-cell unit, are important contributors to cell metabolism and drug metabolism, collectively designated intracellular metabolism. However, disruption of intracellular spatiotemporal metabolism can lead to disease development and progression, as well as drug treatment interference. In this review, we systematically discuss spatiotemporal metabolism in cells and cell subpopulations. In particular, we focused on metabolism compartmentalization and physiological rhythms, including the variation and regulation of metabolic enzymes, metabolic pathways, and metabolites. Additionally, the intricate relationship among intracellular spatiotemporal metabolism, metabolism-related diseases, and drug therapy/toxicity has been discussed. Finally, approaches and strategies for intracellular spatiotemporal metabolism analysis and potential target identification are introduced, along with examples of potential new drug design based on this.
Collapse
Affiliation(s)
- Jingwei Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Zhixia Qiu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yongjie Zhang
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing, China; Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK-PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing, China.
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
2
|
Senjor E, Kos J, Nanut MP. Cysteine Cathepsins as Therapeutic Targets in Immune Regulation and Immune Disorders. Biomedicines 2023; 11:biomedicines11020476. [PMID: 36831012 PMCID: PMC9953096 DOI: 10.3390/biomedicines11020476] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Cysteine cathepsins, as the most abundant proteases found in the lysosomes, play a vital role in several processes-such as protein degradation, changes in cell signaling, cell morphology, migration and proliferation, and energy metabolism. In addition to their lysosomal function, they are also secreted and may remain functional in the extracellular space. Upregulation of cathepsin expression is associated with several pathological conditions including cancer, neurodegeneration, and immune-system dysregulation. In this review, we present an overview of cysteine-cathepsin involvement and possible targeting options for mitigation of aberrant function in immune disorders such as inflammation, autoimmune diseases, and immune response in cancer.
Collapse
Affiliation(s)
- Emanuela Senjor
- Department of Biotechnology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Milica Perišić Nanut
- Department of Biotechnology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
- Correspondence:
| |
Collapse
|
3
|
Berg AL, Rowson-Hodel A, Wheeler MR, Hu M, Free SR, Carraway KL. Engaging the Lysosome and Lysosome-Dependent Cell Death in Cancer. Breast Cancer 2022. [DOI: 10.36255/exon-publications-breast-cancer-lysosome] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
4
|
Niculescu VF. Cancer genes and cancer stem cells in tumorigenesis: Evolutionary deep homology and controversies. Genes Dis 2022; 9:1234-1247. [PMID: 35873035 PMCID: PMC9293697 DOI: 10.1016/j.gendis.2022.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 12/18/2022] Open
|
5
|
Berg AL, Rowson-Hodel A, Hu M, Keeling M, Wu H, VanderVorst K, Chen JJ, Hatakeyama J, Jilek J, Dreyer CA, Wheeler MR, Yu AM, Li Y, Carraway KL. The Cationic Amphiphilic Drug Hexamethylene Amiloride Eradicates Bulk Breast Cancer Cells and Therapy-Resistant Subpopulations with Similar Efficiencies. Cancers (Basel) 2022; 14:cancers14040949. [PMID: 35205696 PMCID: PMC8869814 DOI: 10.3390/cancers14040949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/07/2022] Open
Abstract
The resistance of cancer cell subpopulations, including cancer stem cell (CSC) populations, to apoptosis-inducing chemotherapeutic agents is a key barrier to improved outcomes for cancer patients. The cationic amphiphilic drug hexamethylene amiloride (HMA) has been previously demonstrated to efficiently kill bulk breast cancer cells independent of tumor subtype or species but acts poorly toward non-transformed cells derived from multiple tissues. Here, we demonstrate that HMA is similarly cytotoxic toward breast CSC-related subpopulations that are resistant to conventional chemotherapeutic agents, but poorly cytotoxic toward normal mammary stem cells. HMA inhibits the sphere-forming capacity of FACS-sorted human and mouse mammary CSC-related cells in vitro, specifically kills tumor but not normal mammary organoids ex vivo, and inhibits metastatic outgrowth in vivo, consistent with CSC suppression. Moreover, HMA inhibits viability and sphere formation by lung, colon, pancreatic, brain, liver, prostate, and bladder tumor cell lines, suggesting that its effects may be applicable to multiple malignancies. Our observations expose a key vulnerability intrinsic to cancer stem cells and point to novel strategies for the exploitation of cationic amphiphilic drugs in cancer treatment.
Collapse
Affiliation(s)
- Anastasia L. Berg
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Ashley Rowson-Hodel
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Michelle Hu
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Michael Keeling
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Hao Wu
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Kacey VanderVorst
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jenny J. Chen
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Jason Hatakeyama
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Joseph Jilek
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Courtney A. Dreyer
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Madelyn R. Wheeler
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Kermit L. Carraway
- Department of Biochemistry and Molecular Medicine, University of California, Sacramento, CA 95817, USA; (A.L.B.); (A.R.-H.); (M.H.); (M.K.); (H.W.); (K.V.); (J.J.C.); (J.H.); (J.J.); (C.A.D.); (M.R.W.); (A.-M.Y.); (Y.L.)
- Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Correspondence:
| |
Collapse
|
6
|
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: 1.0] [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
|
7
|
Gureeva TA, Timoshenko OS, Kugaevskaya EV, Solovyova NI. [Cysteine cathepsins: structure, physiological functions and their role in carcinogenesis]. BIOMEDITSINSKAIA KHIMIIA 2021; 67:453-464. [PMID: 34964439 DOI: 10.18097/pbmc20216706453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Cysteine cathepsins (Cts) also known as thiol proteinases belong to the superfamily of cysteine proteinases (EC 3.4.22). Cts are known as lysosomal proteases responsible for the intracellular proteins degradation. All Cts are synthesized as zymogens, activation of which occurs autocatalytically. Their activity is regulated by endogenous inhibitors. Cts can be secreted into the extracellular environment, which is of particular importance in tumor progression. Extracellular Cts not only hydrolyze extracellular matrix (ECM) proteins, but also contribute to ECM remodeling, processing and/or release of cell adhesion molecules, growth factors, cytokines and chemokines. In cancer, the expression and activity of Cts sharply increase both in cell lysosomes and in the intercellular space, which correlates with neoplastic transformation, invasion, metastasis and leads to further tumor progression. It has been shown that Cts expression depends on the cells type, therefore, their role in the tumor development differs depending on their cellular origin. The mechanism of Cts action in cancer is not limited only by their proteolytic action. The Cts influence on signal transduction pathways associated with cancer development, including the pathway involving growth factors, which is mediated through receptors tyrosine kinases (RTK) and various signaling mitogen-activated protein kinases (MAPK), has been proven. In addition, Cts are able to promote the epithelial-mesenchymal transition (EMT) by activating signal transduction pathways such as Wnt, Notch, and the pathway involving TGF-β. So, Ctc perform specific both destructive and regulatory functions, carrying out proteolysis, both inside and outside the cell.
Collapse
Affiliation(s)
- T A Gureeva
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | | |
Collapse
|
8
|
Liu Y, Yao Y, Tao W, Liu F, Yang S, Zhao A, Song D, Li X. Coenzyme Q10 ameliorates BPA-induced apoptosis by regulating autophagy-related lysosomal pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 221:112450. [PMID: 34186417 DOI: 10.1016/j.ecoenv.2021.112450] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 06/13/2023]
Abstract
Bisphenol A (BPA) is a widely distributed environmental endocrine disruptor. The accumulation of BPA has been proved that produce various toxic effects both on human and animals. However, the strategies to reduce the damage of BPA on the body and related mechanisms remain to be studied. Coenzyme Q10 (CoQ10), as a powerful antioxidant, is ubiquitous in many eukaryotic cells, which can improve the integrity of lysosomal membrane, lysosomal degradation function and promote autophagy. Here, we examined the ability of CoQ10 to alleviate oxidative stress and apoptosis in BPA-induced damages in C2C12 cells, and how to alleviate it. Our results showed that BPA treatment significantly reduced cell viability, increased the number of cell apoptosis and ROS production, decreased mitochondrial membrane potential, and inhibited the gene expression of mitochondria biogenesis. Moreover, we demonstrated that exposure to BPA increased expression levels of autophagy protein (LC3-II, p62), inhibited autophagy flux, and disrupted the acidic pH environment of lysosomes. Importantly, CoQ10 supplementation effectively restored these abnormalities caused by BPA. CoQ10 significantly decreased the apoptotic incidence and ROS levels, improved mitochondrial membrane potential. Moreover, CoQ10 improved lysosome function and enhanced autophagy flux. Taken together, our results indicate that CoQ10 supplementation is a feasible and effective way to promote the level of autophagy by improving lysosomal function, thereby reducing the apoptosis caused by BPA accumulation. This study aims to provide evidence for the role of CoQ10 in repairing BPA-induced cell damage in clinical practice.
Collapse
Affiliation(s)
- Yuan Liu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou 311300, China
| | - Yaxin Yao
- Yikon Genomics Company, Ltd., Suzhou 215000, China
| | - Wenjing Tao
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou 311300, China
| | - Feng Liu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou 311300, China
| | - Songbai Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou 311300, China
| | - Ayong Zhao
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou 311300, China
| | - Dan Song
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou 311300, China.
| | - Xiangchen Li
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, Zhejiang A&F University, Hangzhou 311300, China.
| |
Collapse
|
9
|
Protease-triggered bioresponsive drug delivery for the targeted theranostics of malignancy. Acta Pharm Sin B 2021; 11:2220-2242. [PMID: 34522585 PMCID: PMC8424222 DOI: 10.1016/j.apsb.2021.01.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/17/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Proteases have a fundamental role in maintaining physiological homeostasis, but their dysregulation results in severe activity imbalance and pathological conditions, including cancer onset, progression, invasion, and metastasis. This striking importance plus superior biological recognition and catalytic performance of proteases, combining with the excellent physicochemical characteristics of nanomaterials, results in enzyme-activated nano-drug delivery systems (nanoDDS) that perform theranostic functions in highly specific response to the tumor phenotype stimulus. In the tutorial review, the key advances of protease-responsive nanoDDS in the specific diagnosis and targeted treatment for malignancies are emphatically classified according to the effector biomolecule types, on the premise of summarizing the structure and function of each protease. Subsequently, the incomplete matching and recognition between enzyme and substrate, structural design complexity, volume production, and toxicological issues related to the nanocomposites are highlighted to clarify the direction of efforts in nanotheranostics. This will facilitate the promotion of nanotechnology in the management of malignant tumors.
Collapse
|
10
|
miRNA-6715-5p Inhibits Cellular Proliferation and Invasion in Colorectal Cancer by Directly Targeting CST4. JOURNAL OF ONCOLOGY 2021; 2021:7615712. [PMID: 34194498 PMCID: PMC8181091 DOI: 10.1155/2021/7615712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 04/13/2021] [Accepted: 05/22/2021] [Indexed: 12/13/2022]
Abstract
Background Data on the correlation between CST4 and colorectal cancer (CRC) metastasis are scarce. The aim of this study was to analyze CST4 expression and investigate its biological roles and related microRNA- (miRNA-) mediated regulation in CRC. Methods The expression of CST4 was examined in cancer tissues and their corresponding adjacent normal tissues from 40 gastric adenocarcinoma patients. The expression level of CST4 in specimens (cancer and normal tissues) was assessed through immunohistochemistry and/or quantitative polymerase chain reaction. miRNAs targeting CST4 in CRC were predicted by bioinformatics software. CST4 was knocked down in HCT116 cells and candidate miRNAs were transfected into HCT116 cells, and the effects of CST4 knockdown and miRNA transfection on cell proliferation and invasion were examined using CCK8, cell colony formation, and Transwell migration assays. Luciferase double-reporter assays were performed to verify the relationship between miRNA and CST4. Results The expression of CST4 in CRC tissues was significantly higher than that in normal paracancerous tissues, but the results for miRNA-6715-5p were opposite. Regardless of CST4 knockdown or miRNA-6715-5p overexpression, the proliferation and invasion ability of HCT116 cells decreased significantly. Luciferase double-reporter assays showed that the upregulation of miR-6715-5p significantly reduced the luciferase activities of the CST4 3'-UTR plasmid in HCT116 cells. Conclusion CST4 may be involved in CRC proliferation and metastasis. miRNA-6715-5p directly targets CST4 and negatively regulates its expression.
Collapse
|
11
|
Kim HR, Tagirasa R, Yoo E. Covalent Small Molecule Immunomodulators Targeting the Protease Active Site. J Med Chem 2021; 64:5291-5322. [PMID: 33904753 DOI: 10.1021/acs.jmedchem.1c00172] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cells of the immune system utilize multiple proteases to regulate cell functions and orchestrate innate and adaptive immune responses. Dysregulated protease activities are implicated in many immune-related disorders; thus, protease inhibitors have been actively investigated for pharmaceutical development. Although historically considered challenging with concerns about toxicity, compounds that covalently modify the protease active site represent an important class of agents, emerging not only as chemical probes but also as approved drugs. Here, we provide an overview of technologies useful for the study of proteases with the focus on recent advances in chemoproteomic methods and screening platforms. By highlighting covalent inhibitors that have been designed to target immunomodulatory proteases, we identify opportunities for the development of small molecule immunomodulators.
Collapse
Affiliation(s)
- Hong-Rae Kim
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ravichandra Tagirasa
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Euna Yoo
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
12
|
Pišlar A, Tratnjek L, Glavan G, Zidar N, Živin M, Kos J. Neuroinflammation-Induced Upregulation of Glial Cathepsin X Expression and Activity in vivo. Front Mol Neurosci 2020; 13:575453. [PMID: 33328882 PMCID: PMC7714997 DOI: 10.3389/fnmol.2020.575453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/28/2020] [Indexed: 01/12/2023] Open
Abstract
Neuroinflammation is an important factor in the pathogenesis of neurodegenerative diseases. Microglia-derived lysosomal cathepsins have been increasingly recognized as important inflammatory mediators that trigger signaling pathways that aggravate neuroinflammation. In vitro, a contribution to neuroinflammation processes has been shown for cathepsin X: however, the expression patterns and functional role of cathepsin X in neuroinflammatory brain pathology remain elusive. In this study we analyzed the expression, activity, regional distribution and cellular localization of cathepsin X in the rat brain with neuroinflammation-induced neurodegeneration. The unilateral injection of lipopolysaccharide (LPS) induced a strong upregulation of cathepsin X expression and its activity in the ipsilateral striatum. In addition to the striatum, cathepsin X overexpression was detected in other brain areas such as the cerebral cortex, corpus callosum, subventricular zone and external globus pallidus, whereas the upregulation was mainly restricted to activated microglia and reactive astrocytes. Continuous administration of the cathepsin X inhibitor AMS36 indicated protective effects against LPS-induced striatal degeneration, as seen by the attenuated LPS-mediated dilation of the lateral ventricles and partial decreased extent of striatal lesion. Taken together, our results indicate that cathepsin X plays a role as a pathogenic factor in neuroinflammation-induced neurodegeneration and represents a potential therapeutic target for neurodegenerative diseases associated with neuroinflammation.
Collapse
Affiliation(s)
- Anja Pišlar
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Larisa Tratnjek
- Institute of Pathophysiology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia.,Institute of Cell Biology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Gordana Glavan
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Nace Zidar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Marko Živin
- Institute of Pathophysiology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Janko Kos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.,Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
13
|
Pišlar A, Mitrović A, Sabotič J, Pečar Fonović U, Perišić Nanut M, Jakoš T, Senjor E, Kos J. The role of cysteine peptidases in coronavirus cell entry and replication: The therapeutic potential of cathepsin inhibitors. PLoS Pathog 2020; 16:e1009013. [PMID: 33137165 PMCID: PMC7605623 DOI: 10.1371/journal.ppat.1009013] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Over the last 2 decades, several coronaviruses (CoVs) have crossed the species barrier into humans, causing highly prevalent and severe respiratory diseases, often with fatal outcomes. CoVs are a large group of enveloped, single-stranded, positive-sense RNA viruses, which encode large replicase polyproteins that are processed by viral peptidases to generate the nonstructural proteins (Nsps) that mediate viral RNA synthesis. Papain-like peptidases (PLPs) and chymotrypsin-like cysteine 3C-like peptidase are essential for coronaviral replication and represent attractive antiviral drug targets. Furthermore, CoVs utilize the activation of their envelope spike glycoproteins by host cell peptidases to gain entry into cells. CoVs have evolved multiple strategies for spike protein activation, including the utilization of lysosomal cysteine cathepsins. In this review, viral and host peptidases involved in CoV cell entry and replication are discussed in depth, with an emphasis on papain-like cysteine cathepsins. Furthermore, important findings on cysteine peptidase inhibitors with regard to virus attenuation are highlighted as well as the potential of such inhibitors for future treatment strategies for CoV-related diseases.
Collapse
Affiliation(s)
- Anja Pišlar
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Ana Mitrović
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Jerica Sabotič
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Urša Pečar Fonović
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | - Tanja Jakoš
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Emanuela Senjor
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Janko Kos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| |
Collapse
|
14
|
Leto G, Sepporta MV. The potential of cystatin C as a predictive biomarker in breast cancer. Expert Rev Anticancer Ther 2020; 20:1049-1056. [PMID: 32990495 DOI: 10.1080/14737140.2020.1829481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Breast cancer (BCa) is the leading cause of cancer-related deaths among women. Numerous efforts are being directed toward identifying novel tissue and/or circulating molecular markers that may help clinicians in detecting early-stage BCa patients and in providing an accurate estimation of the prognosis and prediction of response to clinical treatments. In this setting, emerging evidence has indicated Cystatin C (Cyst C), as the most potent endogenous inhibitor of cysteine cathepsins, as a possible useful marker in the clinical management of BCa patients. AREAS COVERED This review analyzes the results of emerging studies underpinning a potential clinical role of Cyst C, as additional marker in BCa. EXPERT OPINION Cyst C expression levels have been reported to be altered in tumor tissues and/or in biological fluids of BCa patients. Furthermore, clinical evidence has highlighted a significant correlation between altered Cyst C levels in tumor tissues and/or biological fluids and some clinco-biological parameters of BCa progression. These findings provide evidence for a potential clinical use of Cyst C as a novel marker to improve the clinical and therapeutic management of BCa patients and as a gauge for better clarifying the role of cysteine proteinases in the various steps of BCa progression.
Collapse
Affiliation(s)
- Gaetano Leto
- Laboratory of Experimental Pharmacology, Department of Health Promotion Sciences, School of Medicine, University of Palermo , Palermo, Italy
| | - Maria Vittoria Sepporta
- Pediatric Unit, Department Women-Mother-Children, Pediatric Hematology-Oncology Research Laboratory, Lausanne University Hospital , Lausanne, Switzerland
| |
Collapse
|
15
|
He X, Lin Z, Ning J, Li N, Cui X, Zhao B, Hong F, Miao J. Promoting TTC4 and HSP70 interaction and translocation of annexin A7 to lysosome inhibits apoptosis in vascular endothelial cells. FASEB J 2020; 34:12932-12945. [PMID: 33000523 DOI: 10.1096/fj.202000067r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 07/12/2020] [Accepted: 07/21/2020] [Indexed: 01/16/2023]
Abstract
We previously demonstrated that Tetraticopeptide 4 (TTC4) inhibited apoptosis in vascular endothelial cells (VEC) deprived of serum and fibroblast growth factor 2 (FGF-2). In this study, we aimed to resolve the mechanism of TTC4 inhibiting VEC apoptosis. TTC4, predicted as a HSP70 co-chaperone protein, may regulate the fate of cells by affecting the activity of HSP70, however, there is no experimental evidence showing the interaction of TTC4 and HSP70. Using Co-immunoprecipitation (Co-IP), we demonstrated that TTC4 interacted with HSP70. If HSP70 was knockdown, TTC4 no longer suppressed apoptosis. Furthermore, we found ABO, an inhibitor of annexin A7 (ANXA7) GTPase, could promote the interaction of TTC4 and HSP70 and the translocation of ANXA7 to lysosome. At the same time, ABO inhibited the interaction of HSP70 and ANXA7. Moreover, Akt, as a downstream effector of HSP70 was upregulated, and ANXA7 translocating to lysosome protected the stability of lysosomal membrane. Here, we discovered a special mechanism by which TTC4 inhibited apoptosis via HSP70 in VECs. On the one hand, increasing TTC4 and HSP70 interaction upregulated Akt that inhibited apoptosis. On the other hand, decreasing HSP70 and ANXA7 interaction promoted the translocation of ANXA7 to lysosome, which inhibited apoptosis through protecting the lysosomal membrane stability.
Collapse
Affiliation(s)
- Xiaoying He
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, P.R. China
| | - Zhaomin Lin
- Institute of Medical Science, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Junya Ning
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, P.R. China
| | - Na Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, P.R. China
| | - Xiaoling Cui
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, P.R. China
| | - Baoxiang Zhao
- Institute of Organic Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Jinan, P.R. China
| | - Fanzhen Hong
- Department of Obstetrics, The Second Hospital of Shandong University, Jinan, P.R. China
| | - Junying Miao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, P.R. China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, P.R. China
| |
Collapse
|
16
|
Al-Hashimi A, Venugopalan V, Sereesongsaeng N, Tedelind S, Pinzaru AM, Hein Z, Springer S, Weber E, Führer D, Scott CJ, Burden RE, Brix K. Significance of nuclear cathepsin V in normal thyroid epithelial and carcinoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118846. [PMID: 32910988 DOI: 10.1016/j.bbamcr.2020.118846] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/29/2020] [Indexed: 12/11/2022]
Abstract
Altered expression and/or localization of cysteine cathepsins is believed to involve in thyroid diseases including cancer. Here, we examined the localization of cathepsins B and V in human thyroid tissue sections of different pathological conditions by immunolabeling and morphometry. Cathepsin B was mostly found within endo-lysosomes as expected. In contrast, cathepsin V was detected within nuclei, predominantly in cells of cold nodules, follicular and papillary thyroid carcinoma tissue, while it was less often detected in this unusual localization in hot nodules and goiter tissue. To understand the significance of nuclear cathepsin V in thyroid cells, this study aimed to establish a cellular model of stable nuclear cathepsin V expression. As representative of a specific form lacking the signal peptide and part of the propeptide, N-terminally truncated cathepsin V fused to eGFP recapitulated the nuclear localization of endogenous cathepsin V throughout the cell cycle in Nthy-ori 3-1 cells. Interestingly, the N-terminally truncated cathepsin V-eGFP was more abundant in the nuclei during S phase. These findings suggested a possible contribution of nuclear cathepsin V forms to cell cycle progression. Indeed, we found that N-terminally truncated cathepsin V-eGFP expressing cells were more proliferative than those expressing full-length cathepsin V-eGFP or wild type controls. We conclude that a specific molecular form of cathepsin V localizes to the nucleus of thyroid epithelial and carcinoma cells, where it might involve in deregulated pathways leading to hyperproliferation. These findings highlight the necessity to better understand cathepsin trafficking in health and disease. In particular, cell type specificity of mislocalization of cysteine cathepsins, which otherwise act in a functionally redundant manner, seems to be important to understand their non-canonical roles in cell cycle progression.
Collapse
Affiliation(s)
- Alaa Al-Hashimi
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Vaishnavi Venugopalan
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | | | - Sofia Tedelind
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Alexandra M Pinzaru
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Zeynep Hein
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Sebastian Springer
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Ekkehard Weber
- Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Hollystrasse 1, D-06114 Halle-Saale, Germany
| | - Dagmar Führer
- Universität Duisburg-Essen, Universitätsklinikum Essen (AöR), Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Hufeland Strasse 55, D-45177 Essen, Germany
| | - Christopher J Scott
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Roberta E Burden
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany.
| |
Collapse
|
17
|
Rudzińska M, Parodi A, Maslova VD, Efremov YM, Gorokhovets NV, Makarov VA, Popkov VA, Golovin AV, Zernii EY, Zamyatnin AA. Cysteine Cathepsins Inhibition Affects Their Expression and Human Renal Cancer Cell Phenotype. Cancers (Basel) 2020; 12:cancers12051310. [PMID: 32455715 PMCID: PMC7281206 DOI: 10.3390/cancers12051310] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 12/30/2022] Open
Abstract
Renal cancer would greatly benefit from new therapeutic strategies since, in advanced stages, it is refractory to classical chemotherapeutic approaches. In this context, lysosomal protease cysteine cathepsins may represent new pharmacological targets. In renal cancer, they are characterized by a higher expression, and they were shown to play a role in its aggressiveness and spreading. Traditional studies in the field were focused on understanding the therapeutic potentialities of cysteine cathepsin inhibition, while the direct impact of such therapeutics on the expression of these enzymes was often overlooked. In this work, we engineered two fluoromethyl ketone-based peptides with inhibitory activity against cathepsins to evaluate their potential anticancer activity and impact on the lysosomal compartment in human renal cancer. Molecular modeling and biochemical assays confirmed the inhibitory properties of the peptides against cysteine cathepsin B and L. Different cell biology experiments demonstrated that the peptides could affect renal cancer cell migration and organization in colonies and spheroids, while increasing their adhesion to biological substrates. Finally, these peptide inhibitors modulated the expression of LAMP1, enhanced the expression of E-cadherin, and altered cathepsin expression. In conclusion, the inhibition of cysteine cathepsins by the peptides was beneficial in terms of cancer aggressiveness; however, they could affect the overall expression of these proteases.
Collapse
Affiliation(s)
- Magdalena Rudzińska
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.); (A.P.); (N.V.G.); (V.A.M.); (A.V.G.); (E.Y.Z.)
| | - Alessandro Parodi
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.); (A.P.); (N.V.G.); (V.A.M.); (A.V.G.); (E.Y.Z.)
| | - Valentina D. Maslova
- Faculty of Bioengineering and Bioinformatics, Moscow State University, 119992 Moscow, Russia;
| | - Yuri M. Efremov
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia;
| | - Neonila V. Gorokhovets
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.); (A.P.); (N.V.G.); (V.A.M.); (A.V.G.); (E.Y.Z.)
| | - Vladimir A. Makarov
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.); (A.P.); (N.V.G.); (V.A.M.); (A.V.G.); (E.Y.Z.)
| | - Vasily A. Popkov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
| | - Andrey V. Golovin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.); (A.P.); (N.V.G.); (V.A.M.); (A.V.G.); (E.Y.Z.)
- Faculty of Bioengineering and Bioinformatics, Moscow State University, 119992 Moscow, Russia;
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Evgeni Y. Zernii
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.); (A.P.); (N.V.G.); (V.A.M.); (A.V.G.); (E.Y.Z.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.); (A.P.); (N.V.G.); (V.A.M.); (A.V.G.); (E.Y.Z.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia;
- Correspondence: ; Tel.: +74-95-622-9843
| |
Collapse
|
18
|
Kim GD. Sirt1-Mediated Anti-Aging Effects of Houttuynia cordata Extract in a High Glucose-Induced Endothelial Cell-Aging Model. Prev Nutr Food Sci 2020; 25:108-112. [PMID: 32292763 PMCID: PMC7143013 DOI: 10.3746/pnf.2020.25.1.108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/12/2020] [Indexed: 12/28/2022] Open
Abstract
Houttuynia cordata (HC) is a herb widely used in traditional Asian medicine as an ingredient in complex prescriptions. HC is known for its anti-leukemic, anti-oxidative, and anti-inflammatory properties. However, its anti-vascular endothelial aging efficacy and the underlying mechanisms are not fully understood. In this study, we investigated the anti-aging effects of HC in a high glucose (HG)-induced endothelial cell (EC)-aging model. Treatment with HC (40 μg/mL) increased migration of ECs, and increased phosphorylation of extracellular signal-regulated kinases and p38 in a dose-dependent manner. Following HG treatment (30 mM), HC significantly decreased the number of senescence-associated β-galactosidase positive cells, which are the biomarkers for aging, in a dose-dependent manner. Based on levels of phosphorylation, HC (40 μg/mL) was shown to increase expression of sirtuin1 (Sirt1) and endothelial nitric oxide synthase (eNOS) by 74.4% and 328.2%, respectively. Furthermore, treatment of HG-induced senescent ECs with HC (40 μg/mL) significantly increased nitric oxide production (P<0.05). These results demonstrate that HC both increases EC migration and regulates the Sirt1/eNOS pathway, suggesting HC has potential for protecting ECs against HG-induced aging.
Collapse
Affiliation(s)
- Gi Dae Kim
- Department of Food and Nutrition, Kyungnam University, Gyeongnam 51767, Korea
| |
Collapse
|
19
|
Mitschke J, Burk UC, Reinheckel T. The role of proteases in epithelial-to-mesenchymal cell transitions in cancer. Cancer Metastasis Rev 2020; 38:431-444. [PMID: 31482486 DOI: 10.1007/s10555-019-09808-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Changing the characteristics of cells from epithelial states to mesenchymal properties is a key process involved in developmental and physiological processes as well as in many diseases with cancer as the most prominent example. Nowadays, a great deal of work and literature concerns the understanding of the process of epithelial-to-mesenchymal transition (EMT) in terms of its molecular regulation and its implications for cancer. Similar statements can certainly be made regarding the investigation of the more than 500 proteases typically encoded by a mammalian genome. Specifically, the impact of proteases on tumor biology has been a long-standing topic of interest. However, although EMT actively regulates expression of many proteases and proteolytic enzymes are clearly involved in survival, division, differentiation, and movements of cells, information on the diverse roles of proteases in EMT has been rarely compiled. Here we aim to conceptually connect the scientific areas of "EMT" and "protease" research by describing how several important classes of proteolytic enzymes are regulated by EMT and how they are involved in initiation and execution of the EMT program. To do so, we briefly introduce the evolving key features of EMT and its regulation followed by discussion of protease involvement in this process.
Collapse
Affiliation(s)
- Julia Mitschke
- Institute of Molecular Medicine and Cell Research, University of Freiburg, 79104, Freiburg, Germany
| | - Ulrike C Burk
- Institute of Molecular Medicine and Cell Research, University of Freiburg, 79104, Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, University of Freiburg, 79104, Freiburg, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) Heidelberg, partner site Freiburg, 79106, Freiburg, Germany. .,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
20
|
Lysosomal peptidases in innate immune cells: implications for cancer immunity. Cancer Immunol Immunother 2019; 69:275-283. [PMID: 31813053 DOI: 10.1007/s00262-019-02447-0] [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: 07/29/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
Abstract
Cathepsins are lysosomal peptidases involved in intracellular protein catabolism as well as in various other physiological and pathological processes. Several members of the family, most notably cathepsins B, S, K and L, are frequently overexpressed in cancer and have been associated with remodeling of the proteins of the extracellular matrix, a process leading to tumor cell migration, invasion and metastasis. In addition, lysosomal cathepsins play a role in innate and adaptive immunity, regulation of antigen presentation, Toll-like receptor signaling, cytokine secretion, apoptosis, autophagy, differentiation, migration and cytotoxicity. In cancer, the cells of innate immunity, such as myeloid cells, are often subverted to the regulatory immunosuppressive phenotype. Most studies indicate that lysosomal cathepsins reinforce the pro-tumoral activity of myeloid-derived suppressor cells and tumor-associated macrophages as well as of neutrophils. On the other hand, in cytotoxic natural killer cells, tumor cells suppress lysosomal peptidases in their activation of perforin and granzymes, thus diminishing their killing ability. With multifaceted actions, lysosomal peptidases constitute an important regulatory mechanism for fine-tuning the anti-tumor immune response.
Collapse
|
21
|
Yuan L, Zou C, Ge W, Liu Y, Hu B, Wang J, Lin B, Li Y, Ma E. A novel cathepsin L inhibitor prevents the progression of idiopathic pulmonary fibrosis. Bioorg Chem 2019; 94:103417. [PMID: 31744600 DOI: 10.1016/j.bioorg.2019.103417] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 01/13/2023]
Abstract
In previous work, the target of asperphenamate as a natural product was successfully determined as cathepsin by the natural product consensus pharmacophore strategy. In order to develop accurate SAR (structure-activity relationship) of asperphenamate-type cathepsin inhibitor, we chose several novel analogs with heterocyclic moiety to perform further study. The molecular simulation showed that 4-pyridyl derivative 3 with the greatest cathepsin inhibitory activity presented new interaction modes with protein utilizing its B-ring moiety. And then molecular dynamics (MD) simulation further revealed that 3 and cathepsin kept stable interaction in the binding site, which validated the molecular docking results. In view that cathepsins play an important role in fibrosis and some cathepsin inhibitors display the therapeutic ability for fibrosis, we investigated the anti-fibrotic effect of 3in vitro and in vivo. The results indicated that 3 displayed the strongest inhibitory effect on the formation of α-SMA and collagen I as the protein markers of fibrosis among all tested derivatives. Further in vivo assay confirmed that 3 indeed showed significant inhibitory ability against pulmonary fibrosis by the method of H&E and Masson staining as well as immunohistochemical staining for characteristic α-SMA proteins.
Collapse
Affiliation(s)
- Lei Yuan
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China; Institute of Drug Research in Medicine Capital of China, Benxi 117000, PR China
| | - Chunyang Zou
- Department of Pharmacy, Liaoning Vocational College of Medicine, Shenyang 110101, PR China
| | - Wentao Ge
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China; Institute of Drug Research in Medicine Capital of China, Benxi 117000, PR China
| | - Yutong Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Baichun Hu
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China
| | - Bin Lin
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, Shenyang 110016, PR China; Institute of Drug Research in Medicine Capital of China, Benxi 117000, PR China
| | - Yanchun Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Enlong Ma
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
22
|
Cui Y, Sun D, Song R, Zhang S, Liu X, Wang Y, Meng F, Lan Y, Han J, Pan S, Liang S, Zhang B, Guo H, Liu Y, Lu Z, Liu L. Upregulation of cystatin SN promotes hepatocellular carcinoma progression and predicts a poor prognosis. J Cell Physiol 2019; 234:22623-22634. [PMID: 31106426 PMCID: PMC6767558 DOI: 10.1002/jcp.28828] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022]
Abstract
Cystatin SN, a specific cysteine protease inhibitor, is thought to be involved in various malignant tumors. Therefore, we evaluated the role of cystatin SN in hepatocellular carcinoma (HCC). Notably, cystatin SN was elevated in tumorous samples and cells. Moreover, overexpression of cystatin SN was correlated with tumor diameter and TNM stage. Cox multivariate analysis displayed that cystatin SN was an independent prognosis indicator and that high cystatin SN level was associated with a dismal prognosis. Moreover, cystatin SN enhancement facilitated the proliferation, migratory, and invasive potential of Huh7 and HCCLM3 cells, whereas cystatin SN knockdown caused the opposite effect. Cystatin SN also modulated the epithelial‐mesenchymal transition progression through the PI3K/AKT pathway. In vivo cystatin SN promoted HCCLM3 cell growth and metastasis in xenograft mice model. Thus, cystatin SN was involved in HCC progression and could be a latent target for HCC treatment.
Collapse
Affiliation(s)
- Yifeng Cui
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Dan Sun
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ruipeng Song
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Shugeng Zhang
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Xirui Liu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Yan Wang
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Fanzheng Meng
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Yaliang Lan
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Jihua Han
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Shangha Pan
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Shuhang Liang
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Bo Zhang
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Hongrui Guo
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Yufeng Liu
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Zhaoyang Lu
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Lianxin Liu
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| |
Collapse
|
23
|
Cystatins in cancer progression: More than just cathepsin inhibitors. Biochimie 2019; 166:233-250. [PMID: 31071357 DOI: 10.1016/j.biochi.2019.05.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/03/2019] [Indexed: 12/20/2022]
Abstract
Cystatins are endogenous and reversible inhibitors of cysteine peptidases that are important players in cancer progression. Besides their primary role as regulators of cysteine peptidase activity, cystatins are involved in cancer development and progression through proteolysis-independent mechanisms. Mechanistic studies of cystatin function revealed that they affect all stages of cancer progression including tumor growth, apoptosis, invasion, metastasis and angiogenesis. Recently, the involvement of cystatins in the antitumor immune responses was reported. In this review, we discuss molecular mechanisms and clinical aspects of cystatins in cancer. Altered expression of cystatins in cancer resulting in harmful excessive cysteine peptidase activity has been a subject of several studies in order to find correlations with clinical outcome and therapy response. However, involvement in anti-tumor immune response and signaling cascades leading to cancer progression designates cystatins as possible targets for development of new anti-tumor drugs.
Collapse
|
24
|
Lo CW, Kryvalap Y, Sheu TJ, Chang CH, Czyzyk J. Cellular proliferation in mouse and human pancreatic islets is regulated by serpin B13 inhibition and downstream targeting of E-cadherin by cathepsin L. Diabetologia 2019; 62:822-834. [PMID: 30824970 DOI: 10.1007/s00125-019-4834-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS We previously reported that exposure to antibodies neutralising serpin B13, a protease inhibitor expressed in exocrine pancreatic ducts, promotes beta cell proliferation, underscoring the importance of a functional relationship between exocrine and endocrine pancreas. The aim of the present study was to identify the molecular events that link inhibition of serpin B13 to islet cell proliferation. METHODS We used an in vitro culture system consisting of isolated pancreatic islets, an extract of pancreatic ductal epithelium and a monoclonal antibody (mAb) to serpin B13 or IgG isotype control. In vivo studies involved treatment of mice with these mAbs. RESULTS The catalytic activity of cathepsin L (CatL), a cysteine protease target of serpin B13, was augmented in the pancreas of mice injected with serpin B13 mAb. Furthermore, the addition of serpin B13 mAb to the islets, together with the pancreatic ductal epithelium lysate, caused CatL-dependent cleavage of E-cadherin and concomitant upregulation of REG genes, ultimately leading to beta cell proliferation. Direct blockade of E-cadherin with mAb also markedly enhanced REG gene induction, while chemical inhibition of β-catenin, a binding target of E-cadherin, prevented the serpin B13 mAb-induced upregulation of REG genes. CONCLUSIONS/INTERPRETATION Our work implicates the CatL-E-cadherin-REG pathway in the regulation of islet cell proliferation in response to signals generated in exocrine pancreatic tissue and demonstrates that protease activity may promote adaptive changes in the islets. DATA AVAILABILITY Microarray data that support the findings of this study have been deposited in Gene Expression Omnibus (GEO) with the accession no. GSE125151.
Collapse
Affiliation(s)
- Chi-Wen Lo
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY, USA
| | - Yury Kryvalap
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Washington Ave SE, Minneapolis, MN, 55455, USA
| | - Tzong-Jen Sheu
- The Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| | - Ching-Ho Chang
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Jan Czyzyk
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Washington Ave SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
25
|
Cogo F, Williams R, Burden RE, Scott CJ. Application of nanotechnology to target and exploit tumour associated proteases. Biochimie 2019; 166:112-131. [PMID: 31029743 DOI: 10.1016/j.biochi.2019.04.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023]
Abstract
Proteases are hydrolytic enzymes fundamental for a variety of physiological processes, but the loss of their regulation leads to aberrant functions that promote onset and progression of many diseases including cancer. Proteases have been implicated in almost every hallmark of cancer and whilst widely investigated for tumour therapy, clinical adoption of protease inhibitors as drugs remains a challenge due to issues such as off-target toxicity and inability to achieve therapeutic doses at the disease site. Now, nanotechnology-based solutions and strategies are emerging to circumvent these issues. In this review, preclinical advances in approaches to enhance the delivery of protease drugs and the exploitation of tumour-derived protease activities to promote targeting of nanomedicine formulations is examined. Whilst this field is still in its infancy, innovations to date suggest that nanomedicine approaches to protease targeting or inhibition may hold much therapeutic and diagnostic potential.
Collapse
Affiliation(s)
- Francesco Cogo
- Centre for Cancer Research and Cell Biology, 97 Lisburn Road, BT9 7AE, UK
| | - Rich Williams
- Centre for Cancer Research and Cell Biology, 97 Lisburn Road, BT9 7AE, UK
| | - Roberta E Burden
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | | |
Collapse
|
26
|
Pišlar A, Jewett A, Kos J. Cysteine cathepsins: Their biological and molecular significance in cancer stem cells. Semin Cancer Biol 2018; 53:168-177. [DOI: 10.1016/j.semcancer.2018.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022]
|
27
|
Wyganowska-Świątkowska M, Tarnowski M, Murtagh D, Skrzypczak-Jankun E, Jankun J. Proteolysis is the most fundamental property of malignancy and its inhibition may be used therapeutically (Review). Int J Mol Med 2018; 43:15-25. [PMID: 30431071 PMCID: PMC6257838 DOI: 10.3892/ijmm.2018.3983] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 09/06/2018] [Indexed: 12/22/2022] Open
Abstract
The mortality rates of cancer patients decreased by ~1.5% per year between 2001 and 2015, although the decrease depends on patient sex, ethnic group and type of malignancy. Cancer remains a significant global health problem, requiring a search for novel treatments. The most common property of malignant tumors is their capacity to invade adjacent tissue and to metastasize, and this cancer aggressiveness is contingent on overexpression of proteolytic enzymes. The components of the plasminogen activation system (PAS) and the metal-loproteinase family [mainly matrix metalloproteinases (MMPs)] are overexpressed in malignant tumors, driving the local invasion, metastasis and angiogenesis. This is the case for numerous types of cancer, such as breast, colon, prostate and oral carcinoma, among others. Present chemotherapeutics agents typically attack all dividing cells; however, for future therapeutic agents to be clinically successful, they need to be highly selective for a specific protein(s) and act on the cancerous tissues without adverse systemic effects. Inhibition of proteolysis in cancerous tissue has the ability to attenuate tumor invasion, angiogenesis and migration. For that purpose, inhibiting both PAS and MMPs may be another approach, since the two groups of enzymes are overexpressed in cancer. In the present review, the roles and new findings on PAS and MMP families in cancer formation, growth and possible treatments are discussed.
Collapse
Affiliation(s)
| | | | - Daniel Murtagh
- Urology Research Center, Department of Urology, Health Science Campus, The University of Toledo, Toledo, OH 43614‑2598, USA
| | - Ewa Skrzypczak-Jankun
- Urology Research Center, Department of Urology, Health Science Campus, The University of Toledo, Toledo, OH 43614‑2598, USA
| | - Jerzy Jankun
- Urology Research Center, Department of Urology, Health Science Campus, The University of Toledo, Toledo, OH 43614‑2598, USA
| |
Collapse
|
28
|
The transcription factor C/EBP α controls the role of cystatin F during the differentiation of monocytes to macrophages. Eur J Cell Biol 2018; 97:463-473. [PMID: 30033148 DOI: 10.1016/j.ejcb.2018.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/13/2018] [Accepted: 07/14/2018] [Indexed: 12/30/2022] Open
Abstract
Cystatin F is an inhibitor of cysteine peptidases expressed solely in immune cells. It is the only type II cystatin able to enter endosomal/lysosomal vesicles and to regulate directly the activity of intracellular cysteine cathepsins. Its expression in promonocytic U937 and promyeloblastic HL-60 cells is highly upregulated but, after differentiation with phorbol 12-myristate 13-acetate - PMA, its levels drop significantly. In contrast, the activities of intracellular cysteine cathepsins C, L and S are higher in differentiated cells than in non-differentiated ones due, presumably, to the lower inhibitory capacity of cystatin F. Using immunofluorescence confocal microscopy, proximity ligation assay and co-immunoprecipitation, cathepsins C, L and S were confirmed to be the main interacting partners of cystatin F in U937 and HL-60 cells. The promoter region of the cystatin F gene, CST7, contains a unique binding site for transcription factor C/EBP α, one of the main myeloid differentiation instructors. Using the chromatin immunoprecipitation assay, C/EBP α was shown to bind to CST7 gene in U937 cells. Following cell differentiation with PMA, the binding of C/EBP α was decreased significantly. The protein level of C/EBP α was also significantly lower in differentiated than in non-differentiated cells. It was shown that, during monocyte to macrophage differentiation, the endosomal/lysosomal proteolytic activity can be regulated by cystatin F whose expression is under the control of transcriptional factor C/EBP α.
Collapse
|
29
|
Leto G, Crescimanno M, Flandina C. On the role of cystatin C in cancer progression. Life Sci 2018; 202:152-160. [DOI: 10.1016/j.lfs.2018.04.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/17/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023]
|
30
|
Breuer C, Lemke C, Schmitz J, Bartz U, Gütschow M. Synthesis and kinetic evaluation of ethyl acrylate and vinyl sulfone derived inhibitors for human cysteine cathepsins. Bioorg Med Chem Lett 2018; 28:2008-2012. [DOI: 10.1016/j.bmcl.2018.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/30/2018] [Accepted: 05/01/2018] [Indexed: 11/17/2022]
|
31
|
Estévez-Sarmiento F, Hernández E, Brouard I, León F, García C, Quintana J, Estévez F. 3'-Hydroxy-3,4'-dimethoxyflavone-induced cell death in human leukaemia cells is dependent on caspases and reactive oxygen species and attenuated by the inhibition of JNK/SAPK. Chem Biol Interact 2018; 288:1-11. [PMID: 29630880 DOI: 10.1016/j.cbi.2018.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/07/2018] [Accepted: 04/05/2018] [Indexed: 01/15/2023]
Abstract
Flavonoids are phenolic substances that appear to exert beneficial effects in several chronic diseases, including cancer. Structure-activity relationships of the cytotoxic activity of a series of flavonols and their 3-methyl ether derivatives established that 3'-hydroxy-3,4'-dimethoxyflavone (flavonoid 11) displayed strong cytotoxicity against human leukaemia cell lines (HL-60, U-937 and MOLT-3), and cells that over-express the anti-apoptotic proteins, Bcl-2 and Bcl-xL, and against P-glycoprotein-overexpressing K-562/ADR cells. This compound induced G2-M cell cycle arrest and it was a potent apoptotic inducer on HL-60, MOLT-3, U-937 and U-937/Bcl-2 cell lines. Cell death was (i) mediated by caspase activation, since it was prevented by the non-specific caspase inhibitor z-VAD-fmk and reduced by a selective caspase-9 inhibitor, (ii) associated with cytochrome c release, the dissipation of the inner mitochondrial membrane potential (ΔΨm) and the activation of the mitogen-activated protein kinase pathway and (iii) partially blocked by the inhibition of c-jun NH2 terminal kinases/stress activated protein kinases (JNK/SAPK) signalling and by the free-radical scavenger N-acetyl-l-cysteine.
Collapse
Affiliation(s)
- Francisco Estévez-Sarmiento
- Departamento de Bioquímica y Biología Molecular, Unidad Asociada al Consejo Superior de Investigaciones Científicas (CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de las Palmas de Gran Canaria, Spain
| | - Elisa Hernández
- Departamento de Bioquímica y Biología Molecular, Unidad Asociada al Consejo Superior de Investigaciones Científicas (CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de las Palmas de Gran Canaria, Spain
| | - Ignacio Brouard
- Instituto de Productos Naturales y Agrobiología, CSIC, La Laguna, Tenerife, Spain
| | - Francisco León
- Instituto de Productos Naturales y Agrobiología, CSIC, La Laguna, Tenerife, Spain
| | - Celina García
- Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Departamento de Química Orgánica, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - José Quintana
- Departamento de Bioquímica y Biología Molecular, Unidad Asociada al Consejo Superior de Investigaciones Científicas (CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de las Palmas de Gran Canaria, Spain
| | - Francisco Estévez
- Departamento de Bioquímica y Biología Molecular, Unidad Asociada al Consejo Superior de Investigaciones Científicas (CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias, Universidad de las Palmas de Gran Canaria, Spain.
| |
Collapse
|
32
|
Lu HJ, Yan J, Jin PY, Zheng GH, Qin SM, Wu DM, Lu J, Zheng YL. MicroRNA-152 inhibits tumor cell growth while inducing apoptosis via the transcriptional repression of cathepsin L in gastrointestinal stromal tumor. Cancer Biomark 2018; 21:711-722. [PMID: 29278883 DOI: 10.3233/cbm-170809] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE MicroRNAs are widely thought to play a regulatory role in gene expression. Although the more unique microRNA expression profiles have been reported in several tumors, there remains a scarcity of knowledge in relation to microRNA expression profiles in GISTs. During this study, through the alteration in the expression of microRNA-152 (miR-152) in gastrointestinal stromal tumor (GIST) cells, we subsequently evaluated its ability to influence the processes associated with cancer, including proliferation, migration, invasion, and apoptosis, as well as the associated mechanisms. METHODS The expression of miR-152 and cathepsin L (CTSL) in GIST cell lines (GIST882, GIST430, GIST48 and GIST-T1) and normal gastric mucosal cell line RGM-1 were determined. A series of miR-152 mimics, miR-152 inhibitors, and siRNA against CTSL were introduced to treat GIST-T1 cells with the lowest miR-152 and the highest CTSL were assessed. Cell viability, cell cycle entry, apoptosis, and cell migration/invasion were all evaluated by means of CCK-8 assay, flow cytometry analyses of Annexin V-FITC/PI staining, and transwell assays. RESULTS The target prediction program and luciferase reporter gene assay verified CTSL is the target of miR-152. Regarding the biological significance of miR-152, siRNA knockdown and ectopic expression studies revealed that miR-152 mimic or siRNA against CTSL exposure reduced cell viability and migration/invasion, which resulted in more cells arrested at the S stage, and induced apoptosis. MiR-152 inhibitor exposure was observed to have induced effects on CTSL cells as opposed to those induced by that of the miR-152 mimics. In contrast, miR-152 downregulation abrogated the effects induced by siRNA against CTSL treatment. CONCLUSION The key findings of this study provided evidence suggesting that miR-152 functions by means of binding to CTSL to induce GIST cell apoptosis and inhibit proliferation, migration, and invasion. The anti-tumor role of miR-152 makes it an attractive therapeutic target for GIST.
Collapse
Affiliation(s)
- Hong-Jie Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.,Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Jing Yan
- Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221009, Jiangsu, China.,Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Pei-Ying Jin
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China.,Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Gui-Hong Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Su-Ming Qin
- Department of Oncology, Linyi People's Hospital, Linyi 276003, Shandong, China
| | - Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou 221116, Jiangsu, China
| |
Collapse
|
33
|
Fan J, Wang S, Zhang X, Chen W, Li Y, Yang P, Cao Z, Wang Y, Lu W, Ju D. Quantum Dots Elicit Hepatotoxicity through Lysosome-Dependent Autophagy Activation and Reactive Oxygen Species Production. ACS Biomater Sci Eng 2018; 4:1418-1427. [PMID: 33418671 DOI: 10.1021/acsbiomaterials.7b00824] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Jiajun Fan
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Shaofei Wang
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Xuyao Zhang
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Wei Chen
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Yubin Li
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Ping Yang
- Instrumental Analysis Center, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Zhonglian Cao
- Instrumental Analysis Center, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yichen Wang
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Weiyue Lu
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| | - Dianwen Ju
- Minhang Branch, Zhongshan Hospital, Fudan University/Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai 201199, P. R. China
- Department of Microbiological and Biochemical Pharmacy and Key Lab of Smart Drug Delivery MOE, School of Pharmacy, Fudan University, Shanghai 201203, P. R. China
| |
Collapse
|
34
|
Hesketh GG, Wartosch L, Davis LJ, Bright NA, Luzio JP. The Lysosome and Intracellular Signalling. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2018; 57:151-180. [PMID: 30097775 DOI: 10.1007/978-3-319-96704-2_6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In addition to being the terminal degradative compartment of the cell's endocytic and autophagic pathways, the lysosome is a multifunctional signalling hub integrating the cell's response to nutrient status and growth factor/hormone signalling. The cytosolic surface of the limiting membrane of the lysosome is the site of activation of the multiprotein complex mammalian target of rapamycin complex 1 (mTORC1), which phosphorylates numerous cell growth-related substrates, including transcription factor EB (TFEB). Under conditions in which mTORC1 is inhibited including starvation, TFEB becomes dephosphorylated and translocates to the nucleus where it functions as a master regulator of lysosome biogenesis. The signalling role of lysosomes is not limited to this pathway. They act as an intracellular Ca2+ store, which can release Ca2+ into the cytosol for both local effects on membrane fusion and pleiotropic effects within the cell. The relationship and crosstalk between the lysosomal and endoplasmic reticulum (ER) Ca2+ stores play a role in shaping intracellular Ca2+ signalling. Lysosomes also perform other signalling functions, which are discussed. Current views of the lysosomal compartment recognize its dynamic nature. It includes endolysosomes, autolysosome and storage lysosomes that are constantly engaged in fusion/fission events and lysosome regeneration. How signalling is affected by individual lysosomal organelles being at different stages of these processes and/or at different sites within the cell is poorly understood, but is discussed.
Collapse
Affiliation(s)
- Geoffrey G Hesketh
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Lena Wartosch
- Department of Clinical Biochemistry and Cambridge Institute for Medical Research, School of Clinical Medicine, Wellcome Trust/MRC Building, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
| | - Luther J Davis
- Department of Clinical Biochemistry and Cambridge Institute for Medical Research, School of Clinical Medicine, Wellcome Trust/MRC Building, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
| | - Nicholas A Bright
- Department of Clinical Biochemistry and Cambridge Institute for Medical Research, School of Clinical Medicine, Wellcome Trust/MRC Building, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK
| | - J Paul Luzio
- Department of Clinical Biochemistry and Cambridge Institute for Medical Research, School of Clinical Medicine, Wellcome Trust/MRC Building, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
35
|
Cysteine cathepsins B and X promote epithelial-mesenchymal transition of tumor cells. Eur J Cell Biol 2017; 96:622-631. [DOI: 10.1016/j.ejcb.2017.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 01/28/2023] Open
|
36
|
Mitrović A, Sosič I, Kos Š, Tratar UL, Breznik B, Kranjc S, Mirković B, Gobec S, Lah T, Serša G, Kos J. Addition of 2-(ethylamino)acetonitrile group to nitroxoline results in significantly improved anti-tumor activity in vitro and in vivo. Oncotarget 2017; 8:59136-59147. [PMID: 28938624 PMCID: PMC5601720 DOI: 10.18632/oncotarget.19296] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/10/2017] [Indexed: 12/26/2022] Open
Abstract
Lysosomal cysteine peptidase cathepsin B, involved in multiple processes associated with tumor progression, is validated as a target for anti-cancer therapy. Nitroxoline, a known antimicrobial agent, is a potent and selective inhibitor of cathepsin B, hence reducing tumor progression in vitro and in vivo. In order to further improve its anti-cancer properties we developed a number of derivatives using structure-based chemical synthesis. Of these, the 7-aminomethylated derivative (compound 17) exhibited significantly improved kinetic properties over nitroxoline, inhibiting cathepsin B endopeptidase activity selectively. In the present study, we have evaluated its anti-cancer properties. It was more effective than nitroxoline in reducing tumor cell invasion and migration, as determined in vitro on two-dimensional cell models and tumor spheroids, under either endpoint or real time conditions. Moreover, it exhibited improved action over nitroxoline in impairing tumor growth in vivo in LPB mouse fibrosarcoma tumors in C57Bl/6 mice. Taken together, the addition of a 2-(ethylamino)acetonitrile group to nitroxoline at position 7 significantly improves its pharmacological characteristics and its potential for use as an anti-cancer drug.
Collapse
Affiliation(s)
- Ana Mitrović
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Izidor Sosič
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Špela Kos
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia
| | - Urša Lampreht Tratar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia.,International Postgraduate School Jožef Stefan, 1000 Ljubljana, Slovenia
| | - Simona Kranjc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia
| | - Bojana Mirković
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Tamara Lah
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| | - Gregor Serša
- Department of Experimental Oncology, Institute of Oncology Ljubljana, 1000 Ljubljana, Slovenia
| | - Janko Kos
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia.,Department of Biotechnology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
| |
Collapse
|
37
|
Kramer L, Turk D, Turk B. The Future of Cysteine Cathepsins in Disease Management. Trends Pharmacol Sci 2017; 38:873-898. [PMID: 28668224 DOI: 10.1016/j.tips.2017.06.003] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/23/2017] [Accepted: 06/05/2017] [Indexed: 02/06/2023]
Abstract
Since the discovery of the key role of cathepsin K in bone resorption, cysteine cathepsins have been investigated by pharmaceutical companies as drug targets. The first clinical results from targeting cathepsins by activity-based probes and substrates are paving the way for the next generation of molecular diagnostic imaging, whereas the majority of antibody-drug conjugates currently in clinical trials depend on activation by cathepsins. Finally, cathepsins have emerged as suitable vehicles for targeted drug delivery. It is therefore timely to review the future of cathepsins in drug discovery. We focus here on inflammation-associated diseases because dysregulation of the immune system accompanied by elevated cathepsin activity is a common feature of these conditions.
Collapse
Affiliation(s)
- Lovro Kramer
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, 1000 Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, 1000 Ljubljana, Slovenia
| | - Dušan Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, 1000 Ljubljana, Slovenia; Center of Excellence CIPKEBIP, Jamova 39, 1000 Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, 1000 Ljubljana, Slovenia; Center of Excellence CIPKEBIP, Jamova 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia.
| |
Collapse
|
38
|
Rhana P, Trivelato RR, Beirão PSL, Cruz JS, Rodrigues ALP. Is there a role for voltage-gated Na+ channels in the aggressiveness of breast cancer? ACTA ACUST UNITED AC 2017; 50:e6011. [PMID: 28591378 PMCID: PMC5463531 DOI: 10.1590/1414-431x20176011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/11/2017] [Indexed: 12/19/2022]
Abstract
Breast cancer is the most common cancer among women and its metastatic potential is responsible for numerous deaths. Thus, the need to find new targets for improving treatment, and even finding the cure, becomes increasingly greater. Ion channels are known to participate in several physiological functions, such as muscle contraction, cell volume regulation, immune response and cell proliferation. In breast cancer, different types of ion channels have been associated with tumorigenesis. Recently, voltage-gated Na+ channels (VGSC) have been implicated in the processes that lead to increased tumor aggressiveness. To explain this relationship, different theories, associated with pH changes, gene expression and intracellular Ca2+, have been proposed in an attempt to better understand the role of these ion channels in breast cancer. However, these theories are having difficulty being accepted because most of the findings are contrary to the present scientific knowledge. Several studies have shown that VGSC are related to different types of cancer, making them a promising pharmacological target against this debilitating disease. Molecular biology and cell electrophysiology have been used to look for new forms of treatment aiming to reduce aggressiveness and the disease progress.
Collapse
Affiliation(s)
- P Rhana
- Laboratório de Câncer de Mama, Canais Iônicos e AMP Cíclico, Faculdade de Ciências Humanas, Sociais e da Saúde, Universidade FUMEC, Belo Horizonte, MG, Brasil.,Laboratório de Membranas Excitáveis e de Biologia Cardiovascular, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - R R Trivelato
- Laboratório de Câncer de Mama, Canais Iônicos e AMP Cíclico, Faculdade de Ciências Humanas, Sociais e da Saúde, Universidade FUMEC, Belo Horizonte, MG, Brasil
| | - P S L Beirão
- Laboratório de Membranas Excitáveis e de Biologia Cardiovascular, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - J S Cruz
- Laboratório de Membranas Excitáveis e de Biologia Cardiovascular, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - A L P Rodrigues
- Laboratório de Câncer de Mama, Canais Iônicos e AMP Cíclico, Faculdade de Ciências Humanas, Sociais e da Saúde, Universidade FUMEC, Belo Horizonte, MG, Brasil
| |
Collapse
|
39
|
Dai DN, Li Y, Chen B, Du Y, Li SB, Lu SX, Zhao ZP, Zhou AJ, Xue N, Xia TL, Zeng MS, Zhong Q, Wei WD. Elevated expression of CST1 promotes breast cancer progression and predicts a poor prognosis. J Mol Med (Berl) 2017; 95:873-886. [PMID: 28523467 PMCID: PMC5515997 DOI: 10.1007/s00109-017-1537-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/09/2017] [Accepted: 04/24/2017] [Indexed: 12/14/2022]
Abstract
Cystatin SN (CST1) belongs to the type 2 cystatin (CST) superfamily, which restricts the proteolytic activities of cysteine proteases. CST1 has been recently considered to be involved in the development of several human cancers. However, the prognostic significance and function of CST1 in breast cancer remains unknown. In the current study, we found that CST1 was generally upregulated in breast cancer at both mRNA and protein level. Furthermore, overall survival (OS) and disease-free survival (DFS) in the low CST1 expression subgroup were significantly superior to the high CST1 expression subgroup (OS, p < 0.001; DFS, p < 0.001), which indicated that CST1 expression level was closely correlated to the survival risk of these patients. Univariate and multivariate analyses demonstrated that CST1 expression was an independent prognostic factor, the same as ER status and nodal status. Next, CST1 overexpression promoted breast cancer cell proliferation, clonogenicity, migration, and invasion abilities. By contrast, knockdown of CST1 attenuated these malignant characteristics in breast cancer cells. Collectively, our study indicates that CST1 cannot only serve as a significant prognostic indicator but also as a potential therapeutic target for breast cancer. KEY MESSAGES High CST1 expression is negatively correlated with survival of breast cancer patients. CST1 promotes cell proliferation, clone formation, and metastasis in breast cancer cells. CST1 is a novel potential prognostic biomarker and therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Da-Nian Dai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, 651 East Dongfeng Road, Guangzhou, 510060, China
| | - Yan Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Bo Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, 651 East Dongfeng Road, Guangzhou, 510060, China
| | - Yong Du
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shi-Bing Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shi-Xun Lu
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhi-Ping Zhao
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ai-Jun Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ning Xue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Tian-Liang Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, 651 East Dongfeng Road, Guangzhou, 510060, China.
| | - Wei-Dong Wei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
- Department of Breast Oncology, Sun Yat-Sen University Cancer Center, 651 East Dongfeng Road, Guangzhou, 510060, China.
| |
Collapse
|
40
|
Affiliation(s)
- Ira Skvortsova
- Laboratory for Experimental and Translational Research on Radiation Oncology (EXTRO-Lab), Department of Therapeutic Radiology and Oncology, Innsbruck Medical University, Innsbruck, Austria.
| |
Collapse
|