1
|
Ye D, Duan X, Guan B, Yuan J, Zhu Y, Shi J, Lu Q, Xu G. Biomarker cystatin B expression correlates with pathogenesis in cervical cancer. J Int Med Res 2024; 52:3000605241233959. [PMID: 38436260 PMCID: PMC10913530 DOI: 10.1177/03000605241233959] [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: 10/30/2023] [Accepted: 01/29/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE Cervical cancer (CC) is one of the most common gynecologic malignancies worldwide. Although rapid improvements have been made regarding its prevention and treatment, little is known about disease pathogenesis and the clinical relevance of reliable biomarkers. The present study evaluated the expression of cystatin B (CSTB) as a potential biomarker of CC. METHODS Tissue microarray analysis and immunohistochemical staining were performed to detect CSTB expression, while CSTB mRNA and protein expression levels of freshly isolated CC tissue were measured by quantitative real-time PCR and western blot, respectively. Bioinformatics were used to analyze the CSTB co-expression network and functional enrichments. RESULTS We observed high CSTB mRNA and protein expression levels in CC tissues, which was confirmed by tissue microarray in a comparison with paired adjacent non-cancerous cervical tissue samples. CSTB gene enrichments and associations with co-expressed genes were also observed. Further analysis showed that elevated CSTB expression was associated with pathological progress in CC. CONCLUSION Our data demonstrate that CSTB has the potential to be used as a tissue biomarker with clinical value in patients with CC, which may aid the development of intervention strategies.
Collapse
Affiliation(s)
- Danjuan Ye
- Department of Obstetrics and Gynaecology, Jinshan Hospital, Fudan University, Shanghai, P. R. China
| | - Xiaoling Duan
- Department of Obstetrics and Gynaecology, Jinshan Hospital, Fudan University, Shanghai, P. R. China
| | - Bin Guan
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Jia Yuan
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yan Zhu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, P. R. China
| | - Jimin Shi
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, P. R. China
| | - Qi Lu
- Department of Obstetrics and Gynaecology, Jinshan Hospital, Fudan University, Shanghai, P. R. China
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, P. R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| |
Collapse
|
2
|
Chiodoni C, Sangaletti S, Lecchi M, Ciniselli CM, Cancila V, Tripodi I, Ratti C, Talarico G, Brich S, De Cecco L, Baili P, Truffi M, Sottotetti F, Piccotti F, Tripodo C, Pruneri G, Triulzi T, Corsi F, Cappelletti V, Di Cosimo S, Verderio P, Colombo MP. A three-gene signature marks the time to locoregional recurrence in luminal-like breast cancer. ESMO Open 2023; 8:101590. [PMID: 37393630 PMCID: PMC10485389 DOI: 10.1016/j.esmoop.2023.101590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/10/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
BACKGROUND Gene expression profiling (GEP)-based prognostic signatures are being rapidly integrated into clinical decision making for systemic management of breast cancer patients. However, GEP remains relatively underdeveloped for locoregional risk assessment. Yet, locoregional recurrence (LRR), especially early after surgery, is associated with poor survival. PATIENTS AND METHODS GEP was carried out on two independent luminal-like breast cancer cohorts of patients developing early (≤5 years after surgery) or late (>5 years) LRR and used, by a training and testing approach, to build a gene signature able to intercept women at risk of developing early LRR. The GEP data of two in silico datasets and of a third independent cohort were used to explore its prognostic value. RESULTS Analysis of the first two cohorts led to the identification of three genes, CSTB, CCDC91 and ITGB1, whose expression, derived by principal component analysis, generated a three-gene signature significantly associated with early LRR in both cohorts (P value <0.001 and 0.005, respectively), overcoming the discriminatory capability of age, hormone receptor status and therapy. Remarkably, the integration of the signature with these clinical variables led to an area under the curve of 0.878 [95% confidence interval (CI) 0.810-0.945]. In in silico datasets we found that the three-gene signature retained its association, showing higher values in the early relapsed patients. Moreover, in the third additional cohort, the signature significantly associated with relapse-free survival (hazard ratio 1.56, 95% CI 1.04-2.35). CONCLUSIONS Our three-gene signature represents a new exploitable tool to aid treatment choice in patients with luminal-like breast cancer at risk of developing early recurrence.
Collapse
Affiliation(s)
- C Chiodoni
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - S Sangaletti
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - M Lecchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Epidemiology and Data Science, Unit of Bioinformatics and Biostatistics, Milan
| | - C M Ciniselli
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Epidemiology and Data Science, Unit of Bioinformatics and Biostatistics, Milan
| | - V Cancila
- University of Palermo School of Medicine, Department of Health Sciences, Tumor Immunology Unit, Palermo
| | - I Tripodi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - C Ratti
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - G Talarico
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan
| | - S Brich
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Pathology, Milan
| | - L De Cecco
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Mechanisms Unit, Milan
| | - P Baili
- Fondazione IRCCS Istituto Nazionale dei Tumori, Analytical Epidemiology and Health Impact Unit, Milan
| | - M Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, Laboratory of Nanomedicine, Pavia
| | - F Sottotetti
- Istituti Clinici Scientifici Maugeri IRCCS, Medical Oncology Unit, Pavia
| | - F Piccotti
- Istituti Clinici Scientifici Maugeri IRCCS, Laboratory of Nanomedicine, Pavia
| | - C Tripodo
- University of Palermo School of Medicine, Department of Health Sciences, Tumor Immunology Unit, Palermo; FIRC Institute of Molecular Oncology (IFOM), Milan
| | - G Pruneri
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Pathology, Milan
| | - T Triulzi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Targeting Unit, Milan
| | - F Corsi
- Istituti Clinici Scientifici Maugeri IRCCS, Surgery Department, Breast Unit, Pavia; Department of Biomedical and Clinical Sciences 'L. Sacco', University of Milan, Milan
| | - V Cappelletti
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Advanced Diagnostics, Biomarkers Unit, Milan, Italy
| | - S Di Cosimo
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Advanced Diagnostics, Biomarkers Unit, Milan, Italy
| | - P Verderio
- Fondazione IRCCS Istituto Nazionale dei Tumori, Department of Epidemiology and Data Science, Unit of Bioinformatics and Biostatistics, Milan
| | - M P Colombo
- Fondazione IRCCS Istituto Nazionale dei Tumori, Experimental Oncology Department, Molecular Immunology Unit, Milan.
| |
Collapse
|
3
|
Contini C, Serrao S, Manconi B, Olianas A, Iavarone F, Guadalupi G, Messana I, Castagnola M, Masullo C, Bizzarro A, Turck CW, Maccarrone G, Cabras T. Characterization of Cystatin B Interactome in Saliva from Healthy Elderly and Alzheimer’s Disease Patients. Life (Basel) 2023; 13:life13030748. [PMID: 36983903 PMCID: PMC10054399 DOI: 10.3390/life13030748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Cystatin B is a small, multifunctional protein involved in the regulation of inflammation, innate immune response, and neuronal protection and found highly abundant in the brains of patients with Alzheimer’s disease (AD). Recently, our study demonstrated a significant association between the level of salivary cystatin B and AD. Since the protein is able to establish protein-protein interaction (PPI) in different contexts and aggregation-prone proteins and the PPI networks are relevant for AD pathogenesis, and due to the relevance of finding new AD markers in peripheral biofluids, we thought it was interesting to study the possible involvement of cystatin B in PPIs in saliva and to evaluate differences and similarities between AD and age-matched elderly healthy controls (HC). For this purpose, we applied a co-immunoprecipitation procedure and a bottom-up proteomics analysis to purify, identify, and quantify cystatin B interactors. Results demonstrated for the first time the existence of a salivary cystatin B-linked multi-protein complex composed by 82 interactors and largely expressed in the body. Interactors are involved in neutrophil activation, antimicrobial activity, modulation of the cytoskeleton and extra-cellular matrix (ECM), and glucose metabolism. Preliminary quantitative data showed significantly lower levels of triosophosphate isomerase 1 and higher levels of mucin 7, BPI, and matrix Gla protein in AD with respect to HC, suggesting implications associated with AD of altered glucose metabolism, antibacterial activities, and calcification-associated processes. Data are available via ProteomeXchange with identifiers PXD039286 and PXD030679.
Collapse
Affiliation(s)
- Cristina Contini
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Simone Serrao
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Barbara Manconi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
- Correspondence:
| | - Alessandra Olianas
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Federica Iavarone
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Policlinico Universitario “A. Gemelli” Foundation IRCCS, 00168 Rome, Italy
| | - Giulia Guadalupi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Irene Messana
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy
| | - Massimo Castagnola
- Proteomics Laboratory, European Center for Brain Research, (IRCCS) Santa Lucia Foundation, 00168 Rome, Italy
| | - Carlo Masullo
- Department of Neuroscience, Neurology Section, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Christoph W. Turck
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Giuseppina Maccarrone
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Tiziana Cabras
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
4
|
Lu L, Chen Y, Yang Z, Liang S, Zhu S, Liang X. Expression and Regulation of a Novel Decidual Cells-Derived Estrogen Target during Decidualization. Int J Mol Sci 2022; 24:ijms24010302. [PMID: 36613747 PMCID: PMC9820648 DOI: 10.3390/ijms24010302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
During decidualization in rodents, uterine stromal cells undergo extensive reprogramming to differentiate into distinct cell types, forming primary decidual zones (PDZs), secondary decidual zones (SDZs), and layers of undifferentiated stromal cells. The formation of secondary decidual zones is accompanied by extensive angiogenesis. During early pregnancy, besides ovarian estrogen, de novo synthesis of estrogen in the uterus is essential for the progress of decidualization. However, the molecular mechanisms are not fully understood. Studies have shown that Cystatin B (Cstb) is highly expressed in the decidual tissue of the uterus, but the regulation and mechanism of Cstb in the process of decidualization have not been reported. Our results showed that Cstb was highly expressed in mouse decidua and artificially induced deciduoma via in situ hybridization and immunofluorescence. Estrogen stimulates the expression of Cstb through the Estrogen receptor (ER)α. Moreover, in situ synthesis of estrogen in the uterus during decidualization regulates the expression of Cstb. Silencing the expression of Cstb affects the migration ability of stromal cells. Knockdown Cstb by siRNA significantly inhibits the expression of Dtprp, a marker for mouse decidualization. Our study identifies a novel estrogen target, Cstb, during decidualization and reveals that Cstb may play a pivotal role in angiogenesis during mouse decidualization via the Angptl7.
Collapse
|
5
|
Jiang Y, Han L, Xue M, Wang T, Zhu Y, Xiong C, Shi M, Li H, Hai W, Huo Y, Shen B, Jiang L, Chen H. Cystatin B increases autophagic flux by sustaining proteolytic activity of cathepsin B and fuels glycolysis in pancreatic cancer: CSTB orchestrates autophagy and glycolysis in PDAC. Clin Transl Med 2022; 12:e1126. [PMID: 36495123 PMCID: PMC9736795 DOI: 10.1002/ctm2.1126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Both autophagy and glycolysis are essential for pancreatic ductal adenocarcinoma (PDAC) survival due to desmoplasia. We investigated whether targeting a hub gene which participates in both processes could be an efficient strategy for PDAC treatment. METHODS The expression pattern of glycolysis signatures (GS) and autophagy signatures (AS) and their correlation with cystatin B (CSTB) in PDAC were analysed. It was discovered how CSTB affected the growth, glycolysis, and autophagy of PDAC cells. We assessed competitive binding to cathepsin B (CTSB) between CSTB and cystatin C (CSTC) via immunoprecipitation (IP) and immunofluorescence (IF). Chromatin immunoprecipitation quantitative polymerase chain reaction (ChIP-qPCR) and luciferase reporter gene assays were used to unveil the mechanism underlying CSTB upregulation. The expression pattern of CSTB was examined in clinical samples and KrasG12D/+, Trp53R172H/+, Pdx1-Cre (KPC) mice. RESULTS GS and AS were enriched and closely associated in PDAC tissues. CSTB increased autophagic flux and provided substrates for glycolysis. CSTB knockdown attenuated the proliferation of PDAC cells and patient-derived xenografts. The liquid chromatography-tandem mass spectrometry assay indicated CSTB interacted with CTSB and contributed to the proteolytic activity of CTSB in lysosomes. IF and IP assays demonstrated that CSTB competed with CSTC to bind to CTSB. Mutation of the key sites of CSTB abolished the interaction between CSTB and CTSB. CSTB was highly expressed in PDAC due to H3K27acetylation and SP1 expression. High expression of CSTB in PDAC was observed in tissue microarray and patients' serum samples. CONCLUSIONS Our work demonstrated the tumorigenic roles of autophagy and glycolysis in PDAC. CSTB is a key role in orchestrating these processes to ensure energy supply of PDAC cells.
Collapse
Affiliation(s)
- Yongsheng Jiang
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lijie Han
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Meilin Xue
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ting Wang
- Department of PathologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Youwei Zhu
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Cheng Xiong
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Minmin Shi
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hongzhe Li
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wangxi Hai
- Department of Nuclear MedicineRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yanmiao Huo
- Department of Biliary‐Pancreatic SurgeryRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP. R. China
| | - Baiyong Shen
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina,Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Lingxi Jiang
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hao Chen
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina,Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina,State Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghaiChina,Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
6
|
Bousquet PA, Manna D, Sandvik JA, Arntzen MØ, Moreno E, Sandvig K, Krengel U. SILAC-based quantitative proteomics and microscopy analysis of cancer cells treated with the N-glycolyl GM3-specific anti-tumor antibody 14F7. Front Immunol 2022; 13:994790. [PMID: 36439103 PMCID: PMC9682173 DOI: 10.3389/fimmu.2022.994790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/20/2022] [Indexed: 08/11/2024] Open
Abstract
Cancer immunotherapy represents a promising approach to specifically target and treat cancer. The most common mechanisms by which monoclonal antibodies kill cells include antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity and apoptosis, but also other mechanisms have been described. 14F7 is an antibody raised against the tumor-associated antigen NeuGc GM3, which was previously reported to kill cancer cells without inducing apoptotic pathways. The antibody was reported to induce giant membrane lesions in tumor cells, with apparent changes in the cytoskeleton. Here, we investigated the effect of humanized 14F7 on HeLa cells using stable isotope labeling with amino acids in cell culture (SILAC) in combination with LC-MS and live cell imaging. 14F7 did not kill the HeLa cells, however, it caused altered protein expression (MS data are available via ProteomeXchange with identifier PXD024320). Several cytoskeletal and nucleic-acid binding proteins were found to be strongly down-regulated in response to antibody treatment, suggesting how 14F7 may induce membrane lesions in cells that contain higher amounts of NeuGc GM3. The altered expression profile identified in this study thus contributes to an improved understanding of the unusual killing mechanism of 14F7.
Collapse
Affiliation(s)
| | - Dipankar Manna
- Department of Chemistry, University of Oslo, Oslo, Norway
| | | | | | - Ernesto Moreno
- Facultad de Ciencias Básicas, Universidad de Medellín, Medellín, Colombia
| | - Kirsten Sandvig
- Department of Biosciences, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ute Krengel
- Department of Chemistry, University of Oslo, Oslo, Norway
| |
Collapse
|
7
|
Contini C, Serrao S, Manconi B, Olianas A, Iavarone F, Bizzarro A, Masullo C, Castagnola M, Messana I, Diaz G, Cabras T. Salivary Proteomics Reveals Significant Changes in Relation to Alzheimer's Disease and Aging. J Alzheimers Dis 2022; 89:605-622. [PMID: 35912740 DOI: 10.3233/jad-220246] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Aging is a risk factor for several pathologies as Alzheimer's disease (AD). Great interest exists, therefore, in discovering diagnostic biomarkers and indicators discriminating biological aging and health status. To this aim, omic investigations of biological matrices, as saliva, whose sampling is easy and non-invasive, offer great potential. OBJECTIVE Investigate the salivary proteome through a statistical comparison of the proteomic data by several approaches to highlight quali-/quantitative variations associated specifically either to aging or to AD occurrence, and, thus, able to classify the subjects. METHODS Salivary proteomic data of healthy controls under-70 (adults) and over-70 (elderly) years old, and over-70 AD patients, obtained by liquid chromatography/mass spectrometry, were analyzed by multiple Mann-Whitney test, Kendall correlation, and Random-Forest (RF) analysis. RESULTS Almost all the investigated proteins/peptides significantly decreased in relation to aging in elderly subjects, with or without AD, in comparison with adults. AD subjects exhibited the highest levels of α-defensins, thymosin β4, cystatin B, S100A8 and A9. Correlation tests also highlighted age/disease associated differences. RF analysis individuated quali-/quantitative variations in 20 components, as oxidized S100A8 and S100A9, α-defensin 3, P-B peptide, able to classify with great accuracy the subjects into the three groups. CONCLUSION The findings demonstrated a strong change of the salivary protein profile in relation to the aging. Potential biomarkers candidates of AD were individuated in peptides/proteins involved in antimicrobial defense, innate immune system, inflammation, and in oxidative stress. RF analysis revealed the feasibility of the salivary proteome to discriminate groups of subjects based on age and health status.
Collapse
Affiliation(s)
- Cristina Contini
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Simone Serrao
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Barbara Manconi
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Alessandra Olianas
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Federica Iavarone
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Catholic University of the Sacred Heart, Rome, Italy.,Policlinico Universitario "A. Gemelli" Foundation -IRCCS, Rome, Italy
| | | | - Carlo Masullo
- Department of Neuroscience, Section Neurology, Catholic University of the Sacred Heart, Rome, Italy
| | - Massimo Castagnola
- Proteomics laboratory, European Centre for Research on the Brain, "Santa Lucia" Foundation -IRCCS, Rome, Italy
| | - Irene Messana
- Institute of Chemical Sciences and Technologies "Giulio Natta", National Research Council, Rome, Italy
| | - Giacomo Diaz
- Department of Biomedical Sciences University of Cagliari Cagliari, Italy
| | - Tiziana Cabras
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
8
|
Rudzinska-Radecka M, Frolova AS, Balakireva AV, Gorokhovets NV, Pokrovsky VS, Sokolova DV, Korolev DO, Potoldykova NV, Vinarov AZ, Parodi A, Zamyatnin AA. In Silico, In Vitro, and Clinical Investigations of Cathepsin B and Stefin A mRNA Expression and a Correlation Analysis in Kidney Cancer. Cells 2022; 11:1455. [PMID: 35563761 PMCID: PMC9101197 DOI: 10.3390/cells11091455] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 02/06/2023] Open
Abstract
The cysteine protease Cathepsin B (CtsB) plays a critical role in multiple signaling pathways, intracellular protein degradation, and processing. Endogenous inhibitors regulate its enzymatic activity, including stefins and other cystatins. Recent data proved that CtsB is implicated in tumor extracellular matrix remodeling, cell invasion, and metastasis: a misbalance between cathepsins and their natural inhibitors is often considered a sign of disease progression. In the present study, we investigated CtsB and stefin A (StfA) expression in renal cell carcinoma (RCC). mRNA analysis unveiled a significant CTSB and STFA increase in RCC tissues compared to adjacent non-cancerogenic tissues and a higher CtsB expression in malignant tumors than in benign renal neoplasms. Further analysis highlighted a positive correlation between CtsB and StfA expression as a function of patient sex, age, tumor size, grade, lymph node invasion, metastasis occurrence, and survival. Alternative overexpression and silencing of CtsB and StfA confirmed the correlation expression between these proteins in human RCC-derived cells through protein analysis and fluorescent microscopy. Finally, the ectopic expression of CtsB and StfA increased RCC cell proliferation. Our data strongly indicated that CtsB and StfA expression play an important role in RCC development by mutually stimulating their expression in RCC progression.
Collapse
Affiliation(s)
- Magdalena Rudzinska-Radecka
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
| | - Anastasia S. Frolova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
| | - Anastasia V. Balakireva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Neonila V. Gorokhovets
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
| | - Vadim S. Pokrovsky
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
- Laboratory of Combined Treatment, N.N. Blokhin Cancer Research Center, 115478 Moscow, Russia
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Darina V. Sokolova
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
- Laboratory of Combined Treatment, N.N. Blokhin Cancer Research Center, 115478 Moscow, Russia
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Dmitry O. Korolev
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.V.P.); (A.Z.V.)
| | - Natalia V. Potoldykova
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.V.P.); (A.Z.V.)
| | - Andrey Z. Vinarov
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia; (D.O.K.); (N.V.P.); (A.Z.V.)
| | - Alessandro Parodi
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
| | - Andrey A. Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (M.R.-R.); (A.S.F.); (A.V.B.); (N.V.G.); (A.P.)
- Department of Biotechnology, Sirius University of Science and Technology, 1 Olympic Ave., 354340 Sochi, Russia; (V.S.P.); (D.V.S.)
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- Department of Immunology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
9
|
Reinheckel T, Tholen M. Low level lysosomal membrane permeabilization for limited release and sub-lethal functions of cathepsin proteases in the cytosol and nucleus. FEBS Open Bio 2022; 12:694-707. [PMID: 35203107 PMCID: PMC8972055 DOI: 10.1002/2211-5463.13385] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/21/2022] [Accepted: 02/15/2022] [Indexed: 11/12/2022] Open
Abstract
For a long time, lysosomes were purely seen as organelles in charge of garbage disposal within the cell. They destroy any cargo delivered into their lumen with a plethora of highly potent hydrolytic enzymes, including various proteases. In case of damage to their limiting membranes, the lysosomes release their soluble content with detrimental outcomes for the cell. In recent years however, this view of the lysosome changed towards acknowledging it as a platform for integration of manifold intra- and extracellular signals. Even impaired lysosomal membrane integrity is no longer considered to be a one-way street to cell death. Increasing evidence suggests that lysosomal enzymes, mainly cathepsin proteases, can be released in a spatially and temporarily restricted manner that is compatible with cellular survival. This way, cathepsins can act in the cytosol and the nucleus, where they affect important cellular processes such as cell division. Here, we review this evidence and discuss the routes and molecular mechanisms by which the cathepsins may reach their unusual destination.
Collapse
Affiliation(s)
- Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Biological Signaling Studies BIOSS, Albert Ludwigs University, Freiburg, Germany
| | - Martina Tholen
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany
| |
Collapse
|
10
|
Xu TT, Zeng XW, Wang XH, Yang LX, Luo G, Yu T. Cystatin-B Negatively Regulates the Malignant Characteristics of Oral Squamous Cell Carcinoma Possibly Via the Epithelium Proliferation/Differentiation Program. Front Oncol 2021; 11:707066. [PMID: 34504787 PMCID: PMC8421684 DOI: 10.3389/fonc.2021.707066] [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: 05/08/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
Disturbance in the proteolytic process is one of the malignant signs of tumors. Proteolysis is highly orchestrated by cysteine cathepsin and its inhibitors. Cystatin-B (CSTB) is a general cysteine cathepsin inhibitor that prevents cysteine cathepsin from leaking from lysosomes and causing inappropriate proteolysis. Our study found that CSTB was downregulated in both oral squamous cell carcinoma (OSCC) tissues and cells compared with normal controls. Immunohistochemical analysis showed that CSTB was mainly distributed in the epithelial structure of OSCC tissues, and its expression intensity was related to the grade classification. A correlation analysis between CSTB and clinical prognosis was performed using gene expression data and clinical information acquired from The Cancer Genome Atlas (TCGA) database. Patients with lower expression levels of CSTB had shorter disease-free survival times and poorer clinicopathological features (e.g., lymph node metastases, perineural invasion, low degree of differentiation, and advanced tumor stage). OSCC cell models overexpressing CSTB were constructed to assess the effects of CSTB on malignant biological behaviors and upregulation of CSTB inhibited cell proliferation, migration, and invasion in vitro. Weighted gene correlation network analysis (WGCNA) and gene set enrichment analysis (GSEA) were performed based on the TCGA data to explore potential mechanisms, and CSTB appeared to correlate with squamous epithelial proliferation-differentiation processes, such as epidermal cell differentiation and keratinization. Moreover, in WGCNA, the gene module most associated with CSTB expression (i.e., the brown module) was also the one most associated with grade classification. Upregulation of CSTB promoted the expression levels of markers (LOR, IVL, KRT5/14, and KRT1/10), reflecting a tendency for differentiation and keratinization in vitro. Gene expression profile data of the overexpressed CSTB cell line were obtained by RNA sequencing (RNA-seq) technology. By comparing the GSEA enrichment results of RNA-seq data (from the OSCC models overexpressing CSTB) and existing public database data, three gene sets (i.e., apical junction, G2/M checkpoint, etc.) and six pathways (e.g., NOTCH signaling pathway, glycosaminoglycan degradation, mismatch repair, etc.) were enriched in the data from both sources. Overall, our study shows that CSTB is downregulated in OSCC and might regulate the malignant characteristics of OSCC via the epithelial proliferation/differentiation program.
Collapse
Affiliation(s)
- Tian-Tian Xu
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Xiao-Wen Zeng
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Xin-Hong Wang
- Department of Oral Pathology and Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Lu-Xi Yang
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Gang Luo
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Ting Yu
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| |
Collapse
|
11
|
Contini C, Olianas A, Serrao S, Deriu C, Iavarone F, Boroumand M, Bizzarro A, Lauria A, Faa G, Castagnola M, Messana I, Manconi B, Masullo C, Cabras T. Top-Down Proteomics of Human Saliva Highlights Anti-inflammatory, Antioxidant, and Antimicrobial Defense Responses in Alzheimer Disease. Front Neurosci 2021; 15:668852. [PMID: 34121996 PMCID: PMC8189262 DOI: 10.3389/fnins.2021.668852] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Alzheimer disease (AD) is the most prevalent neurodegenerative disease in the elderly, characterized by accumulation in the brain of misfolded proteins, inflammation, and oxidative damage leading to neuronal cell death. By considering the viewpoint that AD onset and worsening may be influenced by environmental factors causing infection, oxidative stress, and inflammatory reaction, we investigated the changes of the salivary proteome in a population of patients with respect to that in healthy controls (HCs). Indeed, the possible use of saliva as a diagnostic tool has been explored in several oral and systemic diseases. Moreover, the oral cavity continuously established adaptative and protective processes toward exogenous stimuli. In the present study, qualitative/quantitative variations of 56 salivary proteoforms, including post-translationally modified derivatives, have been analyzed by RP-HPLC-ESI-IT-MS and MS/MS analyses, and immunological methods were applied to validate MS results. The salivary protein profile of AD patients was characterized by significantly higher levels of some multifaceted proteins and peptides that were either specific to the oral cavity or also expressed in other body districts: (i) peptides involved in the homeostasis of the oral cavity; (ii) proteins acting as ROS/RNS scavengers and with a neuroprotective role, such as S100A8, S100A9, and their glutathionylated and nitrosylated proteoforms; cystatin B and glutathionylated and dimeric derivatives; (iii) proteins with antimicrobial activity, such as α-defensins, cystatins A and B, histatin 1, statherin, and thymosin β4, this last with a neuroprotective role at the level of microglia. These results suggested that, in response to injured conditions, Alzheimer patients established defensive mechanisms detectable at the oral level. Data are available via ProteomeXchange with identifier PXD021538.
Collapse
Affiliation(s)
- Cristina Contini
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Alessandra Olianas
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Simone Serrao
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Carla Deriu
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Federica Iavarone
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Mozhgan Boroumand
- Laboratorio di Proteomica, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Alessandra Bizzarro
- UOC Continuità Assistenziale, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Alessandra Lauria
- UOC Continuità Assistenziale, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Gavino Faa
- Dipartimento di Scienze Mediche e Sanità Pubblica, University of Cagliari, Cagliari, Italy
| | - Massimo Castagnola
- Laboratorio di Proteomica, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Irene Messana
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Barbara Manconi
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| | - Carlo Masullo
- Dipartimento di Neuroscienze, Sez. Neurologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Tiziana Cabras
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, Cagliari, Italy
| |
Collapse
|
12
|
Di Matteo F, Pipicelli F, Kyrousi C, Tovecci I, Penna E, Crispino M, Chambery A, Russo R, Ayo-Martin AC, Giordano M, Hoffmann A, Ciusani E, Canafoglia L, Götz M, Di Giaimo R, Cappello S. Cystatin B is essential for proliferation and interneuron migration in individuals with EPM1 epilepsy. EMBO Mol Med 2020; 12:e11419. [PMID: 32378798 PMCID: PMC7278547 DOI: 10.15252/emmm.201911419] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/18/2022] Open
Abstract
Progressive myoclonus epilepsy (PME) of Unverricht–Lundborg type (EPM1) is an autosomal recessive neurodegenerative disorder with the highest incidence of PME worldwide. Mutations in the gene encoding cystatin B (CSTB) are the primary genetic cause of EPM1. Here, we investigate the role of CSTB during neurogenesis in vivo in the developing mouse brain and in vitro in human cerebral organoids (hCOs) derived from EPM1 patients. We find that CSTB (but not one of its pathological variants) is secreted into the mouse cerebral spinal fluid and the conditioned media from hCOs. In embryonic mouse brain, we find that functional CSTB influences progenitors’ proliferation and modulates neuronal distribution by attracting interneurons to the site of secretion via cell‐non‐autonomous mechanisms. Similarly, in patient‐derived hCOs, low levels of functional CSTB result in an alteration of progenitor's proliferation, premature differentiation, and changes in interneurons migration. Secretion and extracellular matrix organization are the biological processes particularly affected as suggested by a proteomic analysis in patients’ hCOs. Overall, our study sheds new light on the cellular mechanisms underlying the development of EPM1.
Collapse
Affiliation(s)
- Francesco Di Matteo
- Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Fabrizia Pipicelli
- Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | | | - Isabella Tovecci
- Max Planck Institute of Psychiatry, Munich, Germany.,Department of Biology, University Federico II, Naples, Italy
| | - Eduardo Penna
- Department of Biology, University Federico II, Naples, Italy
| | | | - Angela Chambery
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Rosita Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Ane Cristina Ayo-Martin
- Max Planck Institute of Psychiatry, Munich, Germany.,International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | | | | | - Emilio Ciusani
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Magdalena Götz
- Physiological Genomics, Biomedical Center (BMC), Ludwig-Maximilians-Universitaet (LMU), Planegg/Martinsried, Germany.,Helmholtz Center Munich, Biomedical Center (BMC), Institute of Stem Cell Research, Planegg/Martinsried, Germany.,SyNergy Excellence Cluster, Munich, Germany
| | - Rossella Di Giaimo
- Max Planck Institute of Psychiatry, Munich, Germany.,Department of Biology, University Federico II, Naples, Italy
| | | |
Collapse
|
13
|
Kavčič N, Butinar M, Sobotič B, Hafner Česen M, Petelin A, Bojić L, Zavašnik Bergant T, Bratovš A, Reinheckel T, Turk B. Intracellular cathepsin C levels determine sensitivity of cells to leucyl-leucine methyl ester-triggered apoptosis. FEBS J 2020; 287:5148-5166. [PMID: 32319717 DOI: 10.1111/febs.15326] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 02/06/2020] [Accepted: 03/05/2020] [Indexed: 12/20/2022]
Abstract
L-leucyl-leucine methyl ester (LLOMe) is a lysosomotropic detergent, which was evaluated in clinical trials in graft-vs-host disease because it very efficiently killed monocytic cell lines. It was also shown to efficiently trigger apoptosis in cancer cells, suggesting that the drug might have potential in anticancer therapy. Using U-937 and THP-1 promonocytes as models for monocytic cells, U-87-MG and HeLa cells as models for cancer cells, and noncancerous HEK293 cells, we show that the drug triggers rapid cathepsin C-dependent lysosomal membrane permeabilization, followed by the release of other cysteine cathepsins into the cytosol and subsequent apoptosis. However, monocytes were found to be far more sensitive to the drug than the cancer and noncancer cells, which is most likely a consequence of the much higher intracellular levels of cathepsin C-the most upstream molecule in the pathway-in monocytic cell lines as compared to cancer cells. Overexpression of cathepsin C in HEK293 cells substantially enhances their sensitivity to the drug, consistent with the crucial role of cathepsin C. Major involvement of cysteine cathepsins B, S, and L in the downstream signaling pathway to mitochondrial cell death was confirmed in two gene ablation models, including the ablation of the major cytosolic inhibitor of cysteine cathepsins, stefin B, in primary mouse cancer cells, and simultaneous ablation of two major cathepsins, B and L, in mouse embryonic fibroblasts (MEFs). Deletion of stefin B resulted in sensitizing primary murine breast cancer cells to cell death without affecting the release of cathepsins, whereas simultaneous ablation of cathepsins B and L largely protected MEFs against cell death. However, due to the extreme sensitivity of monocytes to LLOMe, it appears that the drug may not be suitable for anticancer therapy due to risk of systemic toxicity.
Collapse
Affiliation(s)
- Nežka Kavčič
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Miha Butinar
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Barbara Sobotič
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Maruša Hafner Česen
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Ana Petelin
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Health Sciences, University of Primorska, Izola, Slovenia
| | - Lea Bojić
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Tina Zavašnik Bergant
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Andreja Bratovš
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia.,Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Thomas Reinheckel
- Medical Faculty, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs University, Freiburg, Germany.,German Cancer Consortium (DKTK) partner site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Boris Turk
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, Ljubljana, Slovenia
| |
Collapse
|
14
|
Hölzen L, Parigiani MA, Reinheckel T. Tumor cell- and microenvironment-specific roles of cysteine cathepsins in mouse models of human cancers. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140423. [PMID: 32247787 DOI: 10.1016/j.bbapap.2020.140423] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/20/2020] [Accepted: 03/29/2020] [Indexed: 12/22/2022]
Abstract
The human genome encodes for 11 papain-like endolysosomal cysteine peptidases, collectively known as the cysteine cathepsins. Based on their biochemical properties and with the help of experiments in cell culture, the cysteine cathepsins have acquired a reputation as promotors of progression and metastasis of various cancer entities. However, tumors are known to be complex tissues in which non-cancerous cells are also critical for tumorigenesis. Here we discuss the results of the intense investigation of cathepsins in mouse models of human cancers. We focus on models in immunocompetent mice, because only such models allow for analysis of cathepsins in a fully functional tumor microenvironment. An important outcome of those studies was the identification of cancer-promoting cathepsins in tumor-associated macrophages. Another interesting outcome of these animal studies was the identification of a homeostatic tumor-suppressive role for cathepsin L in skin and intestinal cancers. Taken together, these in vivo findings provide a basis for the use of cysteine cathepsins as therapeutic targets, prodrug activators, or as proteases for imaging tumors.
Collapse
Affiliation(s)
- Lena Hölzen
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Maria Alejandra Parigiani
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
15
|
Tian C, Öhlund D, Rickelt S, Lidström T, Huang Y, Hao L, Zhao RT, Franklin O, Bhatia SN, Tuveson DA, Hynes RO. Cancer Cell-Derived Matrisome Proteins Promote Metastasis in Pancreatic Ductal Adenocarcinoma. Cancer Res 2020; 80:1461-1474. [PMID: 32029550 PMCID: PMC7127978 DOI: 10.1158/0008-5472.can-19-2578] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/20/2019] [Accepted: 01/30/2020] [Indexed: 12/23/2022]
Abstract
The prognosis for pancreatic ductal adenocarcinoma (PDAC) remains poor despite decades of effort. The abundant extracellular matrix (ECM) in PDAC comprises a major fraction of the tumor mass and plays various roles in promoting resistance to therapies. However, nonselective depletion of ECM has led to poor patient outcomes. Consistent with that observation, we previously showed that individual matrisome proteins derived from stromal cells correlate with either long or short patient survival. In marked contrast, those derived from cancer cells correlate strongly with poor survival. Here, we studied three cancer cell-derived matrisome proteins that are significantly overrepresented during PDAC progression, AGRN (agrin), SERPINB5 (serine protease inhibitor B5), and CSTB (cystatin B). Using both overexpression and knockdown experiments, we demonstrate that all three are promoters of PDAC metastasis. Furthermore, these proteins operate at different metastatic steps. AGRN promoted epithelial-to-mesenchymal transition in primary tumors, whereas SERPINB5 and CSTB enhanced late steps in the metastatic cascade by elevating invadopodia formation and in vivo extravasation. All three genes were associated with a poor prognosis in human patients and high levels of SERPINB5, secreted by cancer cells and deposited in the ECM, correlated with poor patient prognosis. This study provides strong evidence that cancer cell-derived matrisome proteins can be causal in promoting tumorigenesis and metastasis and lead to poor patient survival. Therefore, compared with the bulk matrix, mostly made by stromal cells, precise interventions targeting cancer cell-derived matrisome proteins, such as AGRN, SERPINB5, and CSTB, may represent preferred potential therapeutic targets. SIGNIFICANCE: This study provides insights into the biological roles of cancer cell-derived matrisome proteins in PDAC and supports the notion that these proteins are protumorigenic and better therapeutic targets.
Collapse
Affiliation(s)
- Chenxi Tian
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Daniel Öhlund
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Steffen Rickelt
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Tommy Lidström
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Ying Huang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Liangliang Hao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Renee T Zhao
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Oskar Franklin
- Department of Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | | | - Richard O Hynes
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| |
Collapse
|
16
|
Guan W, Wang X, Lin Q, Zhang J, Ren W, Xu G. Transforming growth factor‑β/miR‑143‑3p/cystatin B axis is a therapeutic target in human ovarian cancer. Int J Oncol 2019; 55:267-276. [PMID: 31180557 DOI: 10.3892/ijo.2019.4815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/24/2019] [Indexed: 12/24/2022] Open
Abstract
We previously reported that cystatin B (CSTB) is a progression marker of human ovarian cancer (OC); however, the regulatory mechanism of CSTB and its function in OC remain unclear. The present study aimed to explore the mechanism underlying transforming growth factor-β (TGF‑β) 1‑mediated CSTB regulation, and to examine the function of CSTB on OC cell proliferation and apoptosis. Using the online program, miRWalk, a microRNA (miR)‑143‑3p was detected, which contains a homologous sequence of the potential binding site to the 3'‑untranslated region (3'‑UTR) of CSTB. A dual‑luciferase reporter assay confirmed the interaction between miR‑143‑3p and CSTB 3'‑UTR. Treating OC cells with miR‑143‑3p mimics or inhibitors resulted in a decrease or an increase of CSTB expression at mRNA and protein levels, respectively. Additionally, CSTB was significantly overexpressed, whereas miR‑143‑3p was downregulated in human OC tissues compared with normal ovarian tissues. A negative correlation between miR‑143‑3p and CSTB mRNA expression was observed in ovarian malignant tumors. The levels of primary and mature miR‑143‑3p expression were upregulated in OC cells after TGF‑β1 treatment; the action of TGF‑β1 was abolished in the presence of an inhibitor of TGF‑β type I receptor. These results indicated an axis between TGF‑β, miR‑143‑3p and CSTB in OC cells. Furthermore, high levels of CSTB expression were associated with the poor overall survival of patients with OC. Knockdown of CSTB resulted in a decrease in OC cell proliferation and arrested cells in G2/M phase. In addition, suppression of CSTB induced cell apoptosis. In conclusion, CSTB was overexpressed and miR‑143‑3p was downregulated in ovarian malignant tumors. Mature miR‑143‑3p directly bound CSTB 3'‑UTR, leading to a decrease in CSTB expression in OC cells, which was regulated by TGF‑β1. Our findings suggest the potential therapeutic application of targeting the TGF‑β/miR‑143‑3p/CSTB axis for treating patients with OC.
Collapse
Affiliation(s)
- Wencai Guan
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Xingxing Wang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Qunbo Lin
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Jinguo Zhang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Weimin Ren
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
17
|
Liu GM, Zeng HD, Zhang CY, Xu JW. Identification of a six-gene signature predicting overall survival for hepatocellular carcinoma. Cancer Cell Int 2019; 19:138. [PMID: 31139015 PMCID: PMC6528264 DOI: 10.1186/s12935-019-0858-2] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 05/13/2019] [Indexed: 02/08/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) remains a major challenge for public health worldwide. Considering the great heterogeneity of HCC, more accurate prognostic models are urgently needed. To identify a robust prognostic gene signature, we conduct this study. Materials and methods Level 3 mRNA expression profiles and clinicopathological data were obtained in The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC). GSE14520 dataset from the gene expression omnibus (GEO) database was downloaded to further validate the results in TCGA. Differentially expressed mRNAs between HCC and normal tissue were investigated. Univariate Cox regression analysis and lasso Cox regression model were performed to identify and construct the prognostic gene signature. Time-dependent receiver operating characteristic (ROC), Kaplan–Meier curve, multivariate Cox regression analysis, nomogram, and decision curve analysis (DCA) were used to assess the prognostic capacity of the six-gene signature. The prognostic value of the gene signature was further validated in independent GSE14520 cohort. Gene Set Enrichment Analyses (GSEA) was performed to further understand the underlying molecular mechanisms. The performance of the prognostic signature in differentiating between normal liver tissues and HCC were also investigated. Results A novel six-gene signature (including CSE1L, CSTB, MTHFR, DAGLA, MMP10, and GYS2) was established for HCC prognosis prediction. The ROC curve showed good performance in survival prediction in both the TCGA HCC cohort and the GSE14520 validation cohort. The six-gene signature could stratify patients into a high- and low-risk group which had significantly different survival. Cox regression analysis showed that the six-gene signature could independently predict OS. Nomogram including the six-gene signature was established and shown some clinical net benefit. Furthermore, GSEA revealed several significantly enriched oncological signatures and various metabolic process, which might help explain the underlying molecular mechanisms. Besides, the prognostic signature showed a strong ability for differentiating HCC from normal tissues. Conclusions Our study established a novel six-gene signature and nomogram to predict overall survival of HCC, which may help in clinical decision making for individual treatment. Electronic supplementary material The online version of this article (10.1186/s12935-019-0858-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gao-Min Liu
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, No. 38 Huangtang Road, Meizhou, 514000 China
| | - Hua-Dong Zeng
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, No. 38 Huangtang Road, Meizhou, 514000 China
| | - Cai-Yun Zhang
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, No. 38 Huangtang Road, Meizhou, 514000 China
| | - Ji-Wei Xu
- Department of Hepatobiliary Surgery, Meizhou People's Hospital, No. 38 Huangtang Road, Meizhou, 514000 China
| |
Collapse
|
18
|
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: 62] [Impact Index Per Article: 10.3] [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
|
19
|
Vidak E, Javoršek U, Vizovišek M, Turk B. Cysteine Cathepsins and their Extracellular Roles: Shaping the Microenvironment. Cells 2019; 8:cells8030264. [PMID: 30897858 PMCID: PMC6468544 DOI: 10.3390/cells8030264] [Citation(s) in RCA: 267] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 12/17/2022] Open
Abstract
For a long time, cysteine cathepsins were considered primarily as proteases crucial for nonspecific bulk proteolysis in the endolysosomal system. However, this view has dramatically changed, and cathepsins are now considered key players in many important physiological processes, including in diseases like cancer, rheumatoid arthritis, and various inflammatory diseases. Cathepsins are emerging as important players in the extracellular space, and the paradigm is shifting from the degrading enzymes to the enzymes that can also specifically modify extracellular proteins. In pathological conditions, the activity of cathepsins is often dysregulated, resulting in their overexpression and secretion into the extracellular space. This is typically observed in cancer and inflammation, and cathepsins are therefore considered valuable diagnostic and therapeutic targets. In particular, the investigation of limited proteolysis by cathepsins in the extracellular space is opening numerous possibilities for future break-through discoveries. In this review, we highlight the most important findings that establish cysteine cathepsins as important players in the extracellular space and discuss their roles that reach beyond processing and degradation of extracellular matrix (ECM) components. In addition, we discuss the recent developments in cathepsin research and the new possibilities that are opening in translational medicine.
Collapse
Affiliation(s)
- Eva Vidak
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Urban Javoršek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | - Matej Vizovišek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich Otto-Stern-Weg 3, 8093 Zürich, Switzerland.
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia.
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
20
|
Dynamic matrisome: ECM remodeling factors licensing cancer progression and metastasis. Biochim Biophys Acta Rev Cancer 2018; 1870:207-228. [DOI: 10.1016/j.bbcan.2018.09.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/07/2018] [Accepted: 09/30/2018] [Indexed: 01/04/2023]
|
21
|
Librizzi M, Caradonna F, Cruciata I, Dębski J, Sansook S, Dadlez M, Spencer J, Luparello C. Molecular Signatures Associated with Treatment of Triple-Negative MDA-MB231 Breast Cancer Cells with Histone Deacetylase Inhibitors JAHA and SAHA. Chem Res Toxicol 2017; 30:2187-2196. [PMID: 29129070 DOI: 10.1021/acs.chemrestox.7b00269] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Jay Amin hydroxamic acid (JAHA; N8-ferrocenylN1-hydroxy-octanediamide) is a ferrocene-containing analogue of the histone deacetylase inhibitor (HDACi) suberoylanilide hydroxamic acid (SAHA). JAHA's cytotoxic activity on MDA-MB231 triple negative breast cancer (TNBC) cells at 72 h has been previously demonstrated with an IC50 of 8.45 μM. JAHA's lethal effect was found linked to perturbations of cell cycle, mitochondrial activity, signal transduction, and autophagy mechanisms. To glean novel insights on how MDA-MB231 breast cancer cells respond to the cytotoxic effect induced by JAHA, and to compare the biological effect with the related compound SAHA, we have employed a combination of differential display-PCR, proteome analysis, and COMET assay techniques and shown some differences in the molecular signature profiles induced by exposure to either HDACis. In particular, in contrast to the more numerous and diversified changes induced by SAHA, JAHA has shown a more selective impact on expression of molecular signatures involved in antioxidant activity and DNA repair. Besides expanding the biological knowledge of the effect exerted by the modifications in compound structures on cell phenotype, the molecular elements put in evidence in our study may provide promising targets for therapeutic interventions on TNBCs.
Collapse
Affiliation(s)
- Mariangela Librizzi
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo , Viale delle Scienze, 90128 Palermo, Italy
| | - Fabio Caradonna
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo , Viale delle Scienze, 90128 Palermo, Italy
| | - Ilenia Cruciata
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo , Viale delle Scienze, 90128 Palermo, Italy
| | - Janusz Dębski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Pawinskiego 5a, 02-106 Warsaw, Poland
| | - Supojjanee Sansook
- Department of Chemistry, School of Life Sciences, University of Sussex , Falmer, Brighton BN1 9QJ, United Kingdom
| | - Michał Dadlez
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Pawinskiego 5a, 02-106 Warsaw, Poland
| | - John Spencer
- Department of Chemistry, School of Life Sciences, University of Sussex , Falmer, Brighton BN1 9QJ, United Kingdom
| | - Claudio Luparello
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo , Viale delle Scienze, 90128 Palermo, Italy
| |
Collapse
|
22
|
Duivenvoorden HM, Rautela J, Edgington‐Mitchell LE, Spurling A, Greening DW, Nowell CJ, Molloy TJ, Robbins E, Brockwell NK, Lee CS, Chen M, Holliday A, Selinger CI, Hu M, Britt KL, Stroud DA, Bogyo M, Möller A, Polyak K, Sloane BF, O'Toole SA, Parker BS. Myoepithelial cell‐specific expression of stefin A as a suppressor of early breast cancer invasion. J Pathol 2017; 243:496-509. [DOI: 10.1002/path.4990] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/21/2017] [Accepted: 09/18/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Hendrika M Duivenvoorden
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - Jai Rautela
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
- Sir Peter MacCallum Department of Oncology University of Melbourne VIC Australia
- The Walter and Eliza Hall Institute of Medical Research Melbourne VIC Australia
- Department of Medical Biology University of Melbourne VIC Australia
| | - Laura E Edgington‐Mitchell
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences Monash University Melbourne VIC Australia
| | - Alex Spurling
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - David W Greening
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences Monash University Melbourne VIC Australia
| | | | - Elizabeth Robbins
- Department of Tissue Pathology and Diagnostic Oncology Royal Prince Alfred Hospital Camperdown NSW Australia
| | - Natasha K Brockwell
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - Cheok Soon Lee
- Department of Tissue Pathology and Diagnostic Oncology Royal Prince Alfred Hospital Camperdown NSW Australia
- Sydney Medical School University of Sydney NSW Australia
- Cancer Pathology and Cell Biology Laboratory Ingham Institute for Applied Medical Research, and University of New South Wales NSW Australia
- Cancer Pathology, Bosch Institute University of Sydney NSW Australia
| | - Maoshan Chen
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| | - Anne Holliday
- Department of Tissue Pathology and Diagnostic Oncology Royal Prince Alfred Hospital Camperdown NSW Australia
| | - Cristina I Selinger
- Department of Tissue Pathology and Diagnostic Oncology Royal Prince Alfred Hospital Camperdown NSW Australia
| | - Min Hu
- Department of Medical Oncology Dana‐Farber Cancer Institute, Harvard Medical School Boston Massachusetts USA
| | - Kara L Britt
- Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - David A Stroud
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute Monash University Melbourne VIC Australia
| | - Matthew Bogyo
- Department of Pathology Stanford University School of Medicine California USA
| | - Andreas Möller
- Immunology Department QIMR Berghofer Medical Research Institute Brisbane QLD Australia
| | - Kornelia Polyak
- Department of Medical Oncology Dana‐Farber Cancer Institute, Harvard Medical School Boston Massachusetts USA
| | - Bonnie F Sloane
- Department of Pharmacology Wayne State University School of Medicine Detroit Michigan USA
- Barbara Ann Karmanos Cancer Institute Wayne State University School of Medicine Detroit Michigan USA
| | - Sandra A O'Toole
- Sydney Medical School University of Sydney NSW Australia
- Garvan Institute of Medical Research Darlinghurst NSW Australia
- Australian Clinical Labs Bella Vista NSW Australia
| | - Belinda S Parker
- Department of Biochemistry and Genetics La Trobe Institute for Molecular Science Melbourne VIC Australia
| |
Collapse
|
23
|
Bidovec K, Božič J, Dolenc I, Turk B, Turk V, Stoka V. Tumor Necrosis Factor‐α Induced Apoptosis in U937 Cells Promotes Cathepsin D‐Independent Stefin B Degradation. J Cell Biochem 2017; 118:4813-4820. [DOI: 10.1002/jcb.26152] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/19/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Katja Bidovec
- Department of Biochemistry and Molecular and Structural BiologyJožef Stefan InstituteLjubljana SI‐1000Slovenia
- Jožef Stefan International Postgraduate SchoolLjubljana SI‐1000Slovenia
| | - Janja Božič
- Department of Biochemistry and Molecular and Structural BiologyJožef Stefan InstituteLjubljana SI‐1000Slovenia
- Jožef Stefan International Postgraduate SchoolLjubljana SI‐1000Slovenia
| | - Iztok Dolenc
- Department of Biochemistry and Molecular and Structural BiologyJožef Stefan InstituteLjubljana SI‐1000Slovenia
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural BiologyJožef Stefan InstituteLjubljana SI‐1000Slovenia
- Center of Excellence for Integrated Approaches in Chemistry and Biology of ProteinsLjubljana SI‐1000Slovenia
- Faculty of Chemistry and Chemical TechnologyUniversity of LjubljanaLjubljana SI‐1000Slovenia
| | - Vito Turk
- Department of Biochemistry and Molecular and Structural BiologyJožef Stefan InstituteLjubljana SI‐1000Slovenia
- Jožef Stefan International Postgraduate SchoolLjubljana SI‐1000Slovenia
| | - Veronika Stoka
- Department of Biochemistry and Molecular and Structural BiologyJožef Stefan InstituteLjubljana SI‐1000Slovenia
- Jožef Stefan International Postgraduate SchoolLjubljana SI‐1000Slovenia
| |
Collapse
|
24
|
Cystatin C deficiency suppresses tumor growth in a breast cancer model through decreased proliferation of tumor cells. Oncotarget 2017; 8:73793-73809. [PMID: 29088746 PMCID: PMC5650301 DOI: 10.18632/oncotarget.17379] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/22/2017] [Indexed: 01/01/2023] Open
Abstract
Cysteine cathepsins are proteases that, in addition to their important physiological functions, have been associated with multiple pathologies, including cancer. Cystatin C (CstC) is a major endogenous inhibitor that regulates the extracellular activity of cysteine cathepsins. We investigated the role of cystatin C in mammary cancer using CstC knockout mice and a mouse model of breast cancer induced by expression of the polyoma middle T oncoprotein (PyMT) in the mammary epithelium. We showed that the ablation of CstC reduced the rate of mammary tumor growth. Notably, a decrease in the proliferation of CstC knockout PyMT tumor cells was demonstrated ex vivo and in vitro, indicating a role for this protease inhibitor in signaling pathways that control cell proliferation. An increase in phosphorylated p-38 was observed in CstC knockout tumors, suggesting a novel function for cystatin C in cancer development, independent of the TGF-β pathway. Moreover, proteomic analysis of the CstC wild-type and knockout PyMT primary cell secretomes revealed a decrease in the levels of 14-3-3 proteins in the secretome of knock-out cells, suggesting a novel link between cysteine cathepsins, cystatin C and 14-3-3 proteins in tumorigenesis, calling for further investigations.
Collapse
|
25
|
Kavčič N, Pegan K, Turk B. Lysosomes in programmed cell death pathways: from initiators to amplifiers. Biol Chem 2017; 398:289-301. [DOI: 10.1515/hsz-2016-0252] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 09/05/2016] [Indexed: 01/19/2023]
Abstract
Abstract
Lysosome is the central organelle for intracellular degradation of biological macromolecules and organelles. The material destined for degradation enters the lysosomes primarily via endocytosis, autophagy and phagocytosis, and is degraded through the concerted action of more than 50 lysosomal hydrolases. However, lysosomes are also linked with numerous other processes, including cell death, inflammasome activation and immune response, as well as with lysosomal secretion and cholesterol recycling. Among them programmed cell death pathways including apoptosis have received major attention. In most of these pathways, cell death was accompanied by lysosomal membrane permeabilization and release of lysosomal constituents with an involvement of lysosomal hydrolases, including the cathepsins. However, it is less clear, whether lysosomal membrane permeabilization is really critical for the initiation of cell death programme(s). Therefore, the role of lysosomal membrane permeabilization in various programmed cell death pathways is reviewed, as well as the mechanisms leading to it.
Collapse
|
26
|
Eatemadi A, Aiyelabegan HT, Negahdari B, Mazlomi MA, Daraee H, Daraee N, Eatemadi R, Sadroddiny E. Role of protease and protease inhibitors in cancer pathogenesis and treatment. Biomed Pharmacother 2016; 86:221-231. [PMID: 28006747 DOI: 10.1016/j.biopha.2016.12.021] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/26/2016] [Accepted: 12/05/2016] [Indexed: 10/20/2022] Open
Abstract
Cancer is the second cause of death in 2015, and it has been estimated to surpass heart diseases as the leading cause of death in the next few years. Several mechanisms are involved in cancer pathogenesis. Studies have indicated that proteases are also implicated in tumor growth and progression which is highly dependent on nutrient and oxygen supply. On the other hand, protease inhibitors could be considered as a potent strategy in cancer therapy. On the basis of the type of the key amino acid in the active site of the protease and the mechanism of peptide bond cleavage, proteases can be classified into six groups: cysteine, serine, threonine, glutamic acid, aspartate proteases, as well as matrix metalloproteases. In this review, we focus on the role of different types of proteases and protease inhibitors in cancer pathogenesis.
Collapse
Affiliation(s)
- Ali Eatemadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Lorestan, Iran.
| | - Hammed T Aiyelabegan
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Tehran University of Medical Sciences International Campus (TUMS-IC), Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Mazlomi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hadis Daraee
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Lorestan, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasim Daraee
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Lorestan, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Stoka V, Turk V, Turk B. Lysosomal cathepsins and their regulation in aging and neurodegeneration. Ageing Res Rev 2016; 32:22-37. [PMID: 27125852 DOI: 10.1016/j.arr.2016.04.010] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/12/2016] [Accepted: 04/23/2016] [Indexed: 02/07/2023]
Abstract
Lysosomes and lysosomal hydrolases, including the cathepsins, have been shown to change their properties with aging brain a long time ago, although their function was not really understood. The first biochemical and clinical studies were followed by a major expansion in the last 20 years with the development of animal disease models and new approaches leading to a major advancement of understanding of the role of physiological and degenerative processes in the brain at the molecular level. This includes the understanding of the major role of autophagy and the cathepsins in a number of diseases, including its critical role in the neuronal ceroid lipofuscinosis. Similarly, cathepsins and some other lysosomal proteases were shown to have important roles in processing and/or degradation of several important neuronal proteins, thereby having either neuroprotective or harmful roles. In this review, we discuss lysosomal cathepsins and their regulation with the focus on cysteine cathepsins and their endogenous inhibitors, as well as their role in several neurodegenerative diseases.
Collapse
Affiliation(s)
- Veronika Stoka
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, Sl-1000 Ljubljana, Slovenia; J. Stefan International Postgraduate School, Jamova 39, Sl-1000 Ljubljana, Slovenia.
| | - Vito Turk
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, Sl-1000 Ljubljana, Slovenia; J. Stefan International Postgraduate School, Jamova 39, Sl-1000 Ljubljana, Slovenia
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, Sl-1000 Ljubljana, Slovenia; Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Jamova 39, Sl-1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, Sl-1000 Ljubljana, Slovenia.
| |
Collapse
|
28
|
Gómez-Sintes R, Ledesma MD, Boya P. Lysosomal cell death mechanisms in aging. Ageing Res Rev 2016; 32:150-168. [PMID: 26947122 DOI: 10.1016/j.arr.2016.02.009] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 02/22/2016] [Accepted: 02/29/2016] [Indexed: 12/14/2022]
Abstract
Lysosomes are degradative organelles essential for cell homeostasis that regulate a variety of processes, from calcium signaling and nutrient responses to autophagic degradation of intracellular components. Lysosomal cell death is mediated by the lethal effects of cathepsins, which are released into the cytoplasm following lysosomal damage. This process of lysosomal membrane permeabilization and cathepsin release is observed in several physiopathological conditions and plays a role in tissue remodeling, the immune response to intracellular pathogens and neurodegenerative diseases. Many evidences indicate that aging strongly influences lysosomal activity by altering the physical and chemical properties of these organelles, rendering them more sensitive to stress. In this review we focus on how aging alters lysosomal function and increases cell sensitivity to lysosomal membrane permeabilization and lysosomal cell death, both in physiological conditions and age-related pathologies.
Collapse
Affiliation(s)
- Raquel Gómez-Sintes
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biologicas, CIB-CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - María Dolores Ledesma
- Department of Molecular Neurobiology, Centro Biologia Molecular Severo Ochoa, CSIC-UAM, C/Nicolás Cabrera 1, 28049 Madrid, Spain
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biologicas, CIB-CSIC, C/Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
29
|
Khaki PSS, Feroz A, Amin F, Rehman MT, Bhat WF, Bano B. Structural and functional studies on a variant of cystatin purified from brain of Capra hircus. J Biomol Struct Dyn 2016; 35:1693-1709. [DOI: 10.1080/07391102.2016.1191375] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
| | - Anna Feroz
- Faculty of Life Sciences, Department of Biochemistry, AMU, Aligarh, Uttar Pradesh 202002, India
| | - Fakhra Amin
- Faculty of Life Sciences, Department of Zoology, AMU, Aligarh, Uttar Pradesh 202002, India
| | - Md Tabish Rehman
- Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary Biotechnology Unit, AMU, Aligarh, Uttar Pradesh 202002, India
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Waseem Feeroze Bhat
- Faculty of Life Sciences, Department of Biochemistry, AMU, Aligarh, Uttar Pradesh 202002, India
| | - Bilqees Bano
- Faculty of Life Sciences, Department of Biochemistry, AMU, Aligarh, Uttar Pradesh 202002, India
| |
Collapse
|
30
|
Lin YY, Chen ZW, Lin ZP, Lin LB, Yang XM, Xu LY, Xie Q. Tissue Levels of Stefin A and Stefin B in Hepatocellular Carcinoma. Anat Rec (Hoboken) 2016; 299:428-38. [DOI: 10.1002/ar.23311] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 10/16/2015] [Accepted: 11/12/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Yang-Yuan Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Science; Putian University; Putian Fujian China
| | - Zhi-Wei Chen
- Department of Pathology; the Affiliated Hospital of Putian University; Putian Fujian China
| | - Zhi-Ping Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Science; Putian University; Putian Fujian China
| | - Li-Bin Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Science; Putian University; Putian Fujian China
| | - Xue-Ming Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Science; Putian University; Putian Fujian China
| | - Li-Yan Xu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Science; Putian University; Putian Fujian China
| | - Qun Xie
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Science; Putian University; Putian Fujian China
| |
Collapse
|
31
|
Chauhan S, Tomar RS. Efficient expression and purification of biologically active human cystatin proteins. Protein Expr Purif 2016; 118:10-7. [DOI: 10.1016/j.pep.2015.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 10/07/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022]
|
32
|
Chung C, Puthanveetil P, Ory DS, Lieberman AP. Genetic and pharmacological evidence implicates cathepsins in Niemann-Pick C cerebellar degeneration. Hum Mol Genet 2016; 25:1434-46. [PMID: 26908626 DOI: 10.1093/hmg/ddw025] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2016] [Indexed: 11/13/2022] Open
Abstract
Niemann-Pick C1 (NPC) disease, an autosomal recessive lipid trafficking disorder caused by loss-of-function mutations in the NPC1 gene, is characterized by progressive neurodegeneration resulting in cognitive impairment, ataxia and early death. Little is known about the cellular pathways leading to neuron loss. Here, we studied the effects of diminishing expression of cystatin B, an endogenous inhibitor of cathepsins B, H and L, on the development of NPC neuropathology. We show that decreased expression of cystatin B in patient fibroblasts enhances cathepsin activity. Deletion of the encoding Cstb gene in Npc1-deficient mice resulted in striking deleterious effects, particularly within the cerebellum where diffuse loss of Purkinje cells was observed in young mice. This severe pathology occurred through cell autonomous mechanisms that triggered Purkinje cell death. Moreover, our analyses demonstrated the mislocalization of lysosomal cathepsins within the cytosol of Npc1-deficient Purkinje cells. We provide evidence that this may be a consequence of damage to lysosomal membranes by reactive oxygen species (ROS), leading to the leakage of lysosomal contents that culminates in apoptotic cell death. Consistent with this notion, toxicity from ROS was attenuated in an NPC cell model by cystatin B over-expression or pharmacological inhibition of cathepsin B. The observation that Npc1 and Cstb deletion genetically interact to potently enhance the degenerative phenotype of the NPC cerebellum provides strong support for the notion that lysosomal membrane permeabilization contributes to cerebellar degeneration in NPC disease.
Collapse
Affiliation(s)
- Chan Chung
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA and
| | - Prasanth Puthanveetil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA and
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center and Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA and
| |
Collapse
|
33
|
Olson OC, Joyce JA. Cysteine cathepsin proteases: regulators of cancer progression and therapeutic response. Nat Rev Cancer 2015; 15:712-29. [PMID: 26597527 DOI: 10.1038/nrc4027] [Citation(s) in RCA: 484] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cysteine cathepsin protease activity is frequently dysregulated in the context of neoplastic transformation. Increased activity and aberrant localization of proteases within the tumour microenvironment have a potent role in driving cancer progression, proliferation, invasion and metastasis. Recent studies have also uncovered functions for cathepsins in the suppression of the response to therapeutic intervention in various malignancies. However, cathepsins can be either tumour promoting or tumour suppressive depending on the context, which emphasizes the importance of rigorous in vivo analyses to ascertain function. Here, we review the basic research and clinical findings that underlie the roles of cathepsins in cancer, and provide a roadmap for the rational integration of cathepsin-targeting agents into clinical treatment.
Collapse
Affiliation(s)
- Oakley C Olson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center
- Gerstner Sloan Kettering Graduate School of Biomedical Science, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center
- Department of Oncology, University of Lausanne
- Ludwig Institute for Cancer Research, University of Lausanne, CH-1066 Lausanne, Switzerland
| |
Collapse
|
34
|
Železnik TZ, Kadin A, Turk V, Dolenc I. Aspartic cathepsin D degrades the cytosolic cysteine cathepsin inhibitor stefin B in the cells. Biochem Biophys Res Commun 2015; 465:213-7. [PMID: 26239660 DOI: 10.1016/j.bbrc.2015.07.155] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 07/30/2015] [Indexed: 10/23/2022]
Abstract
Stefin B is the major general cytosolic protein inhibitor of cysteine cathepsins. Its main function is to protect the organism against the activity of endogenous potentially hazardous proteases accidentally released from lysosomes. In this study, we investigated the possible effect of endosomal/lysosomal aspartic cathepsins D and E on stefin B after membrane permeabilization. Loss of membrane integrity of lysosomes and endosomes was induced by a lysosomotropic agent L-Leucyl-L-leucine methyl ester (Leu-Leu-OMe). The rat thyroid cell line FRTL-5 was selected as a model cell line owing to its high levels of proteases, including cathepsin D and E. Permeabilization of acid vesicles from FRTL-5 cells induced degradation of stefin B. The process was inhibited by pepstatin A, a potent inhibitor of aspartic proteases. However, degradation of stefin B was prevented by siRNA-mediated silencing of cathepsin D expression. In contrast, cathepsin E silencing had no effect on stefin B degradation. These results showed that cathepsin D and not cathepsin E degrades stefin B. It can be concluded that the presence of cathepsin D in the cytosol affects the inhibitory potency of stefin B, thus preventing the regulation of cysteine cathepsin activities in various biological processes.
Collapse
Affiliation(s)
- Tajana Zajc Železnik
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Andrey Kadin
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Vito Turk
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Iztok Dolenc
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
35
|
Brix K, McInnes J, Al-Hashimi A, Rehders M, Tamhane T, Haugen MH. Proteolysis mediated by cysteine cathepsins and legumain-recent advances and cell biological challenges. PROTOPLASMA 2015; 252:755-774. [PMID: 25398648 DOI: 10.1007/s00709-014-0730-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 11/04/2014] [Indexed: 06/04/2023]
Abstract
Proteases play essential roles in protein degradation, protein processing, and extracellular matrix remodeling in all cell types and tissues. They are also involved in protein turnover for maintenance of homeostasis and protein activation or inactivation for cell signaling. Proteases range in function and specificity, with some performing distinct substrate cleavages, while others accomplish proteolysis of a wide range of substrates. As such, different cell types use specialized molecular mechanisms to regulate the localization of proteases and their function within the compartments to which they are destined. Here, we focus on the cysteine family of cathepsin proteases and legumain, which act predominately within the endo-lysosomal pathway. In particular, recent knowledge on cysteine cathepsins and their primary regulator legumain is scrutinized in terms of their trafficking to endo-lysosomal compartments and other less recognized cellular locations. We further explore the mechanisms that regulate these processes and point to pathological cases which arise from detours taken by these proteases. Moreover, the emerging biological roles of specific forms and variants of cysteine cathepsins and legumain are discussed. These may be decisive, pathogenic, or even deadly when localizing to unusual cellular compartments in their enzymatically active form, because they may exert unexpected effects by alternative substrate cleavage. Hence, we propose future perspectives for addressing the actions of cysteine cathepsins and legumain as well as their specific forms and variants. The increasing knowledge in non-canonical aspects of cysteine cathepsin- and legumain-mediated proteolysis may prove valuable for developing new strategies to utilize these versatile proteases in therapeutic approaches.
Collapse
Affiliation(s)
- Klaudia Brix
- Research Area HEALTH, Research Center MOLIFE-Molecular Life Sciences, Jacobs University Bremen, Campus Ring 1, 28759, Bremen, Germany,
| | | | | | | | | | | |
Collapse
|
36
|
Maher K, Jerič Kokelj B, Butinar M, Mikhaylov G, Manček-Keber M, Stoka V, Vasiljeva O, Turk B, Grigoryev SA, Kopitar-Jerala N. A role for stefin B (cystatin B) in inflammation and endotoxemia. J Biol Chem 2014; 289:31736-31750. [PMID: 25288807 DOI: 10.1074/jbc.m114.609396] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Stefin B (cystatin B) is an endogenous cysteine cathepsin inhibitor, and the loss-of-function mutations in the stefin B gene were reported in patients with Unverricht-Lundborg disease (EPM1). In this study we demonstrated that stefin B-deficient (StB KO) mice were significantly more sensitive to the lethal LPS-induced sepsis and secreted higher amounts of pro-inflammatory cytokines IL-1β and IL-18 in the serum. We further showed that increased caspase-11 gene expression and better pro-inflammatory caspase-1 and -11 activation determined in StB KO bone marrow-derived macrophages resulted in enhanced IL-1β processing. Pretreatment of macrophages with the cathepsin inhibitor E-64d did not affect secretion of IL-1β, suggesting that the increased cathepsin activity determined in StB KO bone marrow-derived macrophages is not essential for inflammasome activation. Upon LPS stimulation, stefin B was targeted into the mitochondria, and the lack of stefin B resulted in the increased destabilization of mitochondrial membrane potential and mitochondrial superoxide generation. Collectively, our study demonstrates that the LPS-induced sepsis in StB KO mice is dependent on caspase-11 and mitochondrial reactive oxygen species but is not associated with the lysosomal destabilization and increased cathepsin activity in the cytosol.
Collapse
Affiliation(s)
- Katarina Maher
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia,; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana SI-1000, Slovenia,; Department of Biochemistry and Molecular Biology, College of Medicine, Penn State University, Hershey, Pennsylvania 17033
| | - Barbara Jerič Kokelj
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia,; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana SI-1000, Slovenia
| | - Miha Butinar
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia,; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana SI-1000, Slovenia
| | - Georgy Mikhaylov
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia,; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana SI-1000, Slovenia
| | - Mateja Manček-Keber
- Department of Biotechnology, National Institute of Chemistry, Hajdrihova 19, Ljubljana SI-1000, Slovenia,; EN-FIST Centre of Excellence, SI-1000 Ljubljana, Slovenia
| | - Veronika Stoka
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia,; Jožef Stefan International Postgraduate School, Jamova 39, Ljubljana SI-1000, Slovenia
| | - Olga Vasiljeva
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia
| | - Boris Turk
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia,; Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins (CIPKeBiP), Jamova 39, SI-1000 Ljubljana, Slovenia,; Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Cesta v Mestni log 88A, SI-1000 Ljubljana, Slovenia, and
| | - Sergei A Grigoryev
- Department of Biochemistry and Molecular Biology, College of Medicine, Penn State University, Hershey, Pennsylvania 17033
| | - Nataša Kopitar-Jerala
- Department of Biochemistry, Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana SI-1000, Slovenia,.
| |
Collapse
|
37
|
Polajnar M, Zavašnik-Bergant T, Škerget K, Vizovišek M, Vidmar R, Fonović M, Kopitar-Jerala N, Petrovič U, Navarro S, Ventura S, Žerovnik E. Human stefin B role in cell's response to misfolded proteins and autophagy. PLoS One 2014; 9:e102500. [PMID: 25047918 PMCID: PMC4105463 DOI: 10.1371/journal.pone.0102500] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/18/2014] [Indexed: 11/30/2022] Open
Abstract
Alternative functions, apart from cathepsins inhibition, are being discovered for stefin B. Here, we investigate its role in vesicular trafficking and autophagy. Astrocytes isolated from stefin B knock-out (KO) mice exhibited an increased level of protein aggregates scattered throughout the cytoplasm. Addition of stefin B monomers or small oligomers to the cell medium reverted this phenotype, as imaged by confocal microscopy. To monitor the identity of proteins embedded within aggregates in wild type (wt) and KO cells, the insoluble cell lysate fractions were isolated and analyzed by mass spectrometry. Chaperones, tubulins, dyneins, and proteosomal components were detected in the insoluble fraction of wt cells but not in KO aggregates. In contrast, the insoluble fraction of KO cells exhibited increased levels of apolipoprotein E, fibronectin, clusterin, major prion protein, and serpins H1 and I2 and some proteins of lysosomal origin, such as cathepsin D and CD63, relative to wt astrocytes. Analysis of autophagy activity demonstrated that this pathway was less functional in KO astrocytes. In addition, synthetic dosage lethality (SDL) gene interactions analysis in Saccharomyces cerevisiae expressing human stefin B suggests a role in transport of vesicles and vacuoles These activities would contribute, directly or indirectly to completion of autophagy in wt astrocytes and would account for the accumulation of protein aggregates in KO cells, since autophagy is a key pathway for the clearance of intracellular protein aggregates.
Collapse
Affiliation(s)
- Mira Polajnar
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
- Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Tina Zavašnik-Bergant
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Katja Škerget
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
- Department of Molecular and Biomedical Science, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Matej Vizovišek
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
- Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Robert Vidmar
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
- Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Marko Fonović
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
- Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Nataša Kopitar-Jerala
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Uroš Petrovič
- Department of Molecular and Biomedical Science, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Susanna Navarro
- Institute of Biotechnology and Biomedicine, Autonomous University of Barcelona, Bellaterra, Spain
| | - Salvador Ventura
- Institute of Biotechnology and Biomedicine, Autonomous University of Barcelona, Bellaterra, Spain
| | - Eva Žerovnik
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
- Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
- CipKeBip - Center of Excellence for integrated approaches in chemistry and biology of proteins, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
38
|
The current stage of cathepsin B inhibitors as potential anticancer agents. Future Med Chem 2014; 6:1355-71. [DOI: 10.4155/fmc.14.73] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cathepsin B is a lysosomal cysteine peptidase, with an important role in the development and progression of cancer. It is involved in the degradation of extracellular matrix proteins, a process promoting invasion and metastasis of tumor cells and tumor angiogenesis. Cathepsin B is unique among cathepsins in possessing both carboxypeptidase and endopeptidase activities. While the former is associated with its physiological role, the latter is involved in pathological degradation of the extracellular matrix. Its activities are regulated by different means, the most important being its endogenous inhibitors, the cystatins. In cancer this peptidase/inhibitor balance is altered, leading to harmful cathepsin B activity. The latter can be prevented by exogenous inhibitors. They differ in modes of inhibition, size, structure, binding affinity, selectivity, toxicity and bioavailability. In this article, we review the properties and function of endogenous and exogenous cathepsin B inhibitors and indicate their application as possible anticancer agents.
Collapse
|
39
|
WANG XINGXING, GUI LU, ZHANG YOUYUAN, ZHANG JIHONG, SHI JIMIN, XU GUOXIONG. Cystatin B is a progression marker of human epithelial ovarian tumors mediated by the TGF-β signaling pathway. Int J Oncol 2014; 44:1099-1106. [PMID: 24452274 PMCID: PMC3977810 DOI: 10.3892/ijo.2014.2261] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 12/24/2013] [Indexed: 01/23/2023] Open
Abstract
Advanced ovarian cancer is a devastating disease. Gaining biomarkers of early detection during ovarian tumorigenesis may lead to earlier diagnosis and better therapeutic strategies. Cystatin B (CSTB) functions as an inhibitor to suppress intracellular cysteine proteases and has been implicated in several types of cancers. The present study explored the expression of CSTB in human ovarian tumors, to investigate CSTB expression associated with clinicopathological features, and to examine the effect of transforming growth factor-β (TGF-β), which plays a key role in ovarian tumorigenesis, on CSTB expression in ovarian cancer cells. The ovarian tissue samples from 33 patients were retrieved. The expression of CSTB in ovarian tissue was examined by immunohistochemistry. We found that CSTB was over-expressed in human ovarian surface epithelial tumors, including serous, mucinous and clear cell tumors. The immunoreactive staining of CSTB was strong in borderline and malignant tumors, weak in benign tumors, and negative in normal tissue counterparts, but was not correlated with the clinicopathological features of patients with ovarian tumors, such as age, histological types, tumor size, lymph node metastasis and clinical stages. The CSTB at mRNA and protein levels in two types of epithelial ovarian cancer cells, OVCAR-3 and SK-OV-3, was decreased after TGF-β1 treatment detected by quantitative PCR and western blot analysis, respectively. The inhibitory effect of TGF-β1 on CSTB expression was abolished in the presence of SB-431542, a TGF-β type I receptor kinase inhibitor. Our data suggest that CSTB is tumor tissue-specific and overexpressed in ovarian borderline and malignant tumors. The increased CSTB expression in ovarian tissue represents tumor progression and is dysregulated by the TGF-β signaling pathway. CSTB may become a novel diagnostic intracellular biomarker for the early detection of ovarian cancer.
Collapse
Affiliation(s)
- XINGXING WANG
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai 201508
| | - LU GUI
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai 201508
| | - YOUYUAN ZHANG
- Department of Pathology, Jinshan Hospital, Fudan University, Shanghai 201508
| | - JIHONG ZHANG
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508
| | - JIMIN SHI
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508
| | - GUOXIONG XU
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032,
P.R. China
| |
Collapse
|