1
|
Paliwal P, Desai D, Datta M. Novel selective and sensitive electrochemical sensor for detection of ovarian cancer biomarker, Cathepsin L. Bioelectrochemistry 2025; 162:108856. [PMID: 39581047 DOI: 10.1016/j.bioelechem.2024.108856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Late detection of ovarian cancer is critical as it results in decreased life expectancy of patients. Screening methodology with a rapid turnover is required to ensure better patient outcomes. Till date, no point of care diagnostics exists capable of monitoring epithelial ovarian cancer. Cathepsin L is a classic early-stage biomarker demarcating the proliferation and transition of ovarian cancer. A novel label-free electrochemical diagnostic is proposed for detection of Cathepsin L in serum. Intrinsic fluorescence spectroscopy was used to confirm the interaction between Cathepsin L and probe, Cystatin C. Cystatin C was immobilized on screen-printed carbon electrode using 1-Ethyl-3-(3-dimethyl-aminopropyl carbodiimide and N-hydroxysuccinimide cross linker. Electrochemical fluctuations resulting from Cystatin-Cathepsin interaction was detected by cyclic voltammetry, differential voltammetry and electrochemical impedance spectroscopy within a timeframe of 20 min. The proposed diagnostic for Cathepsin L exhibited analytical performance with a limit of detection upto 70 pg mL-1 via electrochemical impedance spectroscopy in the dynamic detection range of 2 ng mL-1-20 ng mL-1. The platform indicated reliable reproducibility, selectivity, and stability. Furthermore, validation with spiked sera samples (n = 5) showed correlation of R2 = 0.98. The results indicate sensitive yet selective point of care diagnosis of Cathepsin L in serum.
Collapse
Affiliation(s)
- Priya Paliwal
- Amity Institute of Biotechnology, Amity University, Rajasthan 303002, India
| | - Dignya Desai
- Catalent Pharmasolutions, Somerset, NJ 008873, USA
| | - Manali Datta
- Amity Institute of Biotechnology, Amity University, Rajasthan 303002, India.
| |
Collapse
|
2
|
Lu J, Liang J, Xiao G, He Z, Yu G, Zhang L, Cai C, Yi G, Xie J. Cathepsin L in Lung Adenocarcinoma: Prognostic Significance and Immunotherapy Response Through a Multi Omics Perspective. Cancer Inform 2024; 23:11769351241307492. [PMID: 39687501 PMCID: PMC11648051 DOI: 10.1177/11769351241307492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Objectives Lung adenocarcinoma (LUAD), a predominant form of lung cancer, is characterized by a high rate of metastasis and recurrence, leading to a poor prognosis for LUAD patients. This study aimed to identify and rigorously validate a highly precise biomarker, Cathepsin L (CTSL), for the prognostic prediction of lung adenocarcinoma. Methods We employed a multicenter and omics-based approach, analyzing RNA sequencing data and mutation information from public databases such as The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). The DepMap portal with Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9) technology was used to assess the functional impact of CTSL. Immunohistochemistry (IHC) was conducted on a local cohort to validate the prognostic significance of CTSL at the protein expression level. Results Our findings revealed a significant correlation between elevated CTSL expression and advanced disease stage in LUAD patients. Kaplan-Meier survival analysis and Cox regression modeling revealed that high CTSL expression is associated with poor overall survival. The in vitro studies corroborated these findings, revealing notable suppression of tumor proliferation following CTSL knockout in cell lines, particularly in LUAD. Functional enrichment revealed that CTSL activated pathways associated with tumor progression, such as angiogenesis and Transforming growth factor beta (TGF-beta) signaling, and inhibited pathways such as apoptosis and DNA repair. Mutation analysis revealed distinct variations in the CTSL expression groups. Conclusion This study highlights the crucial role of CTSL as a prognostic biomarker in LUAD. This combined multicenter and omics-based analysis provides comprehensive insights into the biological role of CTSL, supporting its potential as a target for therapeutic intervention and a marker for prognosis in patients with LUAD.
Collapse
Affiliation(s)
- Jianming Lu
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jiaqi Liang
- Pneumology Department, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gang Xiao
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Thoracic Surgery, Guangzhou First People’s Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong, China
| | - Zitao He
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Guifang Yu
- Oncology Department, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Le Zhang
- Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA, USA
| | - Chao Cai
- Department of Urology, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Gao Yi
- Pneumology Department, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jianjiang Xie
- Center for Medical Research on Innovation and Translation, Institute of Clinical Medicine, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Thoracic Surgery, Guangzhou First People’s Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Khalid M, Alqarni MH, Foudah AI. Repurposed pharmacotherapy: targeting cathepsin L with repurposed drugs in virtual screening. Mol Divers 2024:10.1007/s11030-024-11022-4. [PMID: 39470912 DOI: 10.1007/s11030-024-11022-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024]
Abstract
Proteolytic enzymes are closely associated with cancer and are important in different phases, including tumor growth, angiogenesis, and metastasis. Despite efforts to target matrix metalloproteases (MMPs), clinical trials have often resulted in various side effects such as musculoskeletal pain, joint stiffness, and tendinitis, making them less optimal for chronic cancer treatment. Thus, there is a need for the identification of other protease targets that would provide different approaches towards the management of cancer. Of these targets, Cathepsin L (CatL) is a lysosomal cysteine protease that has been identified as a therapeutic target that is implicated in cancer development and metastasis. In this study, we performed an integrated approach of virtual screening and molecular dynamics (MD) simulations to identify the potential inhibitors of CatL from a library of drugs that have been used for different treatments. Towards this goal, we performed virtual screening of the DrugBank database and found two repurposed drugs, Irinotecan and Nilotinib, against CatL based on their docking profiles, favorable docking scores, and specific interaction with the CatL binding pocket. MD simulations of the Irinotecan and Nilotinib bound structures with CatL were carried out, and the analysis showed that both these compounds could function as CatL inhibitors as the protein-ligand interactions were stable for 300 ns. This study highlights the robustness of these drugs bound to CatL and indicates that they could be repurposed for the treatment of cancer. These findings endorse the use of computer-based approaches for the identification of new inhibitors, and the present study will be a useful resource for future experimental research towards the targeting of CatL in cancer therapeutics.
Collapse
Affiliation(s)
- Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Mohammed H Alqarni
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ahmed I Foudah
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
4
|
Choi JH, Jeong J, Kim J, You E, Keum S, Song S, Hwang YE, Ji M, Park KS, Rhee S. Genetic disruption of ATAT1 causes RhoA downregulation through abnormal truncation of C/EBPβ. BMB Rep 2024; 57:293-298. [PMID: 38835115 PMCID: PMC11214891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/03/2024] [Accepted: 02/01/2024] [Indexed: 06/06/2024] Open
Abstract
Microtubule acetylation has been shown to regulate actin filament dynamics by modulating signaling pathways that control actin organization, although the precise mechanisms remain unknown. In this study, we found that the downregulation of microtubule acetylation via the disruption ATAT1 (which encodes α-tubulin N-acetyltransferase 1) inhibited the expression of RhoA, a small GTPase involved in regulating the organization of actin filaments and the formation of stress fibers. Analysis of RHOA promoter and chromatin immunoprecipitation assays revealed that C/EBPβ is a major regulator of RHOA expression. Interestingly, the majority of C/EBPβ in ATAT1 knockout (KO) cells was found in the nucleus as a 27-kDa fragment (referred to as C/EBPβp27) lacking the N-terminus of C/EBPβ. Overexpression of a gene encoding a C/EBPβp27-mimicking protein via an N-terminal deletion in C/EBPβ led to competitive binding with wild-type C/EBPβ at the C/EBPβ binding site in the RHOA promoter, resulting in a significant decrease of RHOA expression. We also found that cathepsin L (CTSL), which is overexpressed in ATAT1 KO cells, is responsible for C/EBPβp27 formation in the nucleus. Treatment with a CTSL inhibitor led to the restoration of RHOA expression by downregulation of C/EBPβp27 and the invasive ability of ATAT1 KO MDA-MB-231 breast cancer cells. Collectively, our findings suggest that the downregulation of microtubule acetylation associated with ATAT1 deficiency suppresses RHOA expression by forming C/EBPβp27 in the nucleus through CTSL. We propose that CTSL and C/EBPβp27 may represent a novel therapeutic target for breast cancer treatment. [BMB Reports 2024; 57(6): 293-298].
Collapse
Affiliation(s)
- Jee-Hye Choi
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea, VA 22903, USA
| | - Jangho Jeong
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea, VA 22903, USA
| | - Jaegu Kim
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea, VA 22903, USA
| | - Eunae You
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea, VA 22903, USA
| | - Seula Keum
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea, VA 22903, USA
| | - Seongeun Song
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea, VA 22903, USA
| | - Ye Eun Hwang
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea, VA 22903, USA
| | - Minjoo Ji
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea, VA 22903, USA
| | - Kwon-Sik Park
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea, VA 22903, USA
| |
Collapse
|
5
|
Zhu L, Zeng Q, Wang J, Deng F, Jin S. Cathepsin V drives lung cancer progression by shaping the immunosuppressive environment and adhesion molecules cleavage. Aging (Albany NY) 2023; 15:13961-13979. [PMID: 38078882 PMCID: PMC10756122 DOI: 10.18632/aging.205278] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/16/2023] [Indexed: 12/21/2023]
Abstract
Cathepsin V (CTSV) is a cysteine cathepsin protease that plays a crucial role in extracellular matrix degradation. CTSV is correlated with poor prognosis in various cancers, but the underlying mechanism remains elusive. Here, we observed that CSTV is upregulated in lung cancer and is a poor prognosis factor for lung cancer. CTSV acts as a driver in the metastasis of lung cancer both in vitro and in vivo. CTSV promotes lung cancer metastasis by downregulating adhesion molecules, including fibronectin, E-cadherin, and N-cadherin. Our data revealed that CTSV functions by mediating the fragmentation of fibronectin, E-cadherin, and N-cadherin in cleavage, remodeling the extracellular matrix (ECM). The rationally designed antibody targeting CTSV blocks its cleaving ability towards fibronectin, E-cadherin, and N-cadherin, suppressing migration and invasion. Furthermore, we found that CTSV expression is negatively correlated with immune cell infiltration and immune scores and inhibits T cell activity. Targeting CTSV with specific antibodies effectively suppressed lung cancer metastasis in a mouse model. Our study demonstrates the critical role of CTSV in the immunity and metastasis of lung cancer, suggesting that the CTSV-targeting approach is a promising strategy for lung cancer.
Collapse
Affiliation(s)
- Lifei Zhu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Dermatology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qi Zeng
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Jinxiang Wang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Precision Medicine Center, Sun Yat-Sen University, Shenzhen 518107, China
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shi Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| |
Collapse
|
6
|
Luyapan J, Bossé Y, Li Z, Xiao X, Rosenberger A, Hung RJ, Lam S, Zienolddiny S, Liu G, Kiemeney LA, Chen C, McKay J, Johansson M, Johansson M, Tardon A, Fernandez-Tardon G, Brennan P, Field JK, Davies MP, Woll PJ, Cox A, Taylor F, Arnold SM, Lazarus P, Grankvist K, Landi MT, Christiani DC, MacKenzie TA, Amos CI. Candidate pathway analysis of surfactant proteins identifies CTSH and SFTA2 that influences lung cancer risk. Hum Mol Genet 2023; 32:2842-2855. [PMID: 37471639 PMCID: PMC10481107 DOI: 10.1093/hmg/ddad095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 07/22/2023] Open
Abstract
Pulmonary surfactant is a lipoprotein synthesized and secreted by alveolar type II cells in lung. We evaluated the associations between 200,139 single nucleotide polymorphisms (SNPs) of 40 surfactant-related genes and lung cancer risk using genotyped data from two independent lung cancer genome-wide association studies. Discovery data included 18,082 cases and 13,780 controls of European ancestry. Replication data included 1,914 cases and 3,065 controls of European descent. Using multivariate logistic regression, we found novel SNPs in surfactant-related genes CTSH [rs34577742 C > T, odds ratio (OR) = 0.90, 95% confidence interval (CI) = 0.89-0.93, P = 7.64 × 10-9] and SFTA2 (rs3095153 G > A, OR = 1.16, 95% CI = 1.10-1.21, P = 1.27 × 10-9) associated with overall lung cancer in the discovery data and validated in an independent replication data-CTSH (rs34577742 C > T, OR = 0.88, 95% CI = 0.80-0.96, P = 5.76 × 10-3) and SFTA2 (rs3095153 G > A, OR = 1.14, 95% CI = 1.01-1.28, P = 3.25 × 10-2). Among ever smokers, we found SNPs in CTSH (rs34577742 C > T, OR = 0.89, 95% CI = 0.85-0.92, P = 1.94 × 10-7) and SFTA2 (rs3095152 G > A, OR = 1.20, 95% CI = 1.14-1.27, P = 4.25 × 10-11) associated with overall lung cancer in the discovery data and validated in the replication data-CTSH (rs34577742 C > T, OR = 0.88, 95% CI = 0.79-0.97, P = 1.64 × 10-2) and SFTA2 (rs3095152 G > A, OR = 1.15, 95% CI = 1.01-1.30, P = 3.81 × 10-2). Subsequent transcriptome-wide association study using expression weights from a lung expression quantitative trait loci study revealed genes most strongly associated with lung cancer are CTSH (PTWAS = 2.44 × 10-4) and SFTA2 (PTWAS = 2.32 × 10-6).
Collapse
Affiliation(s)
- Jennifer Luyapan
- Quantitative Biomedical Science Program, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, G1V 0A6, Canada
- Department of Molecular Medicine, Laval University, Quebec City, G1V 0A6, Canada
| | - Zhonglin Li
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, G1V 0A6, Canada
| | - Xiangjun Xiao
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Albert Rosenberger
- Institut für Genetische Epidemiologie, Georg-August-Universität Göttingen, Gottingen, Niedersachsen, Germany
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbuaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Stephen Lam
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, BC, V5Z 4E6, Canada
| | - Shanbeh Zienolddiny
- Department of Toxicology, National Institute of Occupational Health, Oslo 0033, Norway
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, Princess Margaret Research Institute, Epidemiology Division,Toronto, ON, M5G 1L7, Canada
| | - Lambertus A Kiemeney
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, 6525 GA, the Netherlands
| | - Chu Chen
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - James McKay
- International Agency for Research on Cancer (IARC/WHO), Genomic Epidemiology Branch Lyon 69008, France
| | - Mattias Johansson
- International Agency for Research on Cancer (IARC/WHO), Genomic Epidemiology Branch Lyon 69008, France
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, 901 87, Sweden
| | - Adonina Tardon
- Health Research Institute of the Principality of Asturias, University of Oviedo and CIBERSP, Oviedo, Asturias, 33071, Spain
| | - Guillermo Fernandez-Tardon
- Health Research Institute of the Principality of Asturias, University of Oviedo and CIBERSP, Oviedo, Asturias, 33071, Spain
| | - Paul Brennan
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig Maximillians University, Munich, Bavaria, 80539, Germany
| | - John K Field
- Molecular and Clinical Cancer Medicine, Roy Castle Lung Cancer Research Programme, The University of Liverpool Institute of Translational Medicine, Liverpool, L69 7ZX, UK
| | - Michael P Davies
- Molecular and Clinical Cancer Medicine, Roy Castle Lung Cancer Research Programme, The University of Liverpool Institute of Translational Medicine, Liverpool, L69 7ZX, UK
| | - Penella J Woll
- Academic Unit of Clinical Oncology, University of Sheffield, Sheffield, S10 2AH, UK
| | - Angela Cox
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2AH, UK
| | - Fiona Taylor
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2AH, UK
| | - Susanne M Arnold
- Division of Medical Oncology, Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99163, USA
| | - Kjell Grankvist
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, 901 87, Sweden
| | - Maria T Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - David C Christiani
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02115, USA
| | - Todd A MacKenzie
- Quantitative Biomedical Science Program, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Christopher I Amos
- Quantitative Biomedical Science Program, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
7
|
Fujiwara T, Zhang L, Chandler A, Sung S, Yakoub M, Linkov I, Hameed M, Healey JH. Cathepsin protease expression in infiltrative soft tissue sarcomas: cathepsin-K correlates with infiltrative tumor growth and clinical outcomes. Hum Pathol 2023; 134:30-44. [PMID: 36565726 PMCID: PMC10748737 DOI: 10.1016/j.humpath.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/18/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Cathepsin proteases, activated in the lysosomes, are upregulated in many cancers. Intraoperative detection systems of microscopic residual tumor using cathepsin-mediated release of fluorescent nanoparticles may guide surgical excisions to improve local control. We sought to define the genetic and proteomic expression of cathepsins and their clinicopathological correlates in myxofibrosarcoma and undifferentiated pleomorphic sarcoma (UPS)-soft tissue sarcomas with high rates of positive resection margins and local recurrence-and to establish a cellular justification for cathepsin-dependent systems to identify residual cancer in the resection bed. Real-time quantitative polymerase chain reaction analysis of 58 fresh-frozen tumor specimens revealed that 56 (97%) had elevated mRNA expression of ≥1 cathepsin, including cathepsin-B (79%), cathepsin-K (59%), cathepsin-L (71%), and -S (71%). Immunohistochemical analysis of these fresh-frozen specimens revealed that 98% of tumors were positive for one or more of cathepsin-B (85%), cathepsin-K (50%), cathepsin-L (63%), and -S (10%). Strong cathepsin-K expression was associated with greater risks of local recurrence (hazard ratio, 3.78; p = 0.044) and disease-specific mortality (hazard ratio, 3.70; p = 0.025). Immunohistochemical analysis of 33 formalin-fixed paraffin-embedded block samples revealed that 97% were positive for cathepsin-B (88%), cathepsin-K (76%), cathepsin-L (52%), or -S (52%) at the tumor periphery; cathepsin-K positivity correlated with a radiographic tail-like sign (p = 0.004) and microscopic infiltrative growth (p = 0.020). We conclude that cathepsins are broadly overexpressed in myxofibrosarcoma and UPS, and cathepsin-K may be an immunohistochemical marker of local infiltration and poorer prognosis that could be used to guide precision surgery.
Collapse
Affiliation(s)
- Tomohiro Fujiwara
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, And Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Lingxin Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Andrew Chandler
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Shijun Sung
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Mohamed Yakoub
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Irina Linkov
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Meera Hameed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - John H Healey
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
8
|
Al-Bari AA. Inhibition of autolysosomes by repurposing drugs as a promising therapeutic strategy for the treatment of cancers. ALL LIFE 2022; 15:568-601. [DOI: 10.1080/26895293.2022.2078894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/02/2022] [Indexed: 10/18/2022] Open
Affiliation(s)
- Abdul Alim Al-Bari
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, Bangladesh
| |
Collapse
|
9
|
Combination of light and Ru(II) polypyridyl complexes: Recent advances in the development of new anticancer drugs. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
10
|
All Roads Lead to Cathepsins: The Role of Cathepsins in Non-Alcoholic Steatohepatitis-Induced Hepatocellular Carcinoma. Biomedicines 2022; 10:biomedicines10102351. [PMID: 36289617 PMCID: PMC9598942 DOI: 10.3390/biomedicines10102351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Cathepsins are lysosomal proteases that are essential to maintain cellular physiological homeostasis and are involved in multiple processes, such as immune and energy regulation. Predominantly, cathepsins reside in the lysosomal compartment; however, they can also be secreted by cells and enter the extracellular space. Extracellular cathepsins have been linked to several pathologies, including non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC). NASH is an increasingly important risk factor for the development of HCC, which is the third leading cause of cancer-related deaths and poses a great medical and economic burden. While information regarding the involvement of cathepsins in NASH-induced HCC (NASH-HCC) is limited, data to support the role of cathepsins in either NASH or HCC is accumulating. Since cathepsins play a role in both NASH and HCC, it is likely that the role of cathepsins is more significant in NASH-HCC compared to HCC derived from other etiologies. In the current review, we provide an overview on the available data regarding cathepsins in NASH and HCC, argue that cathepsins play a key role in the transition from NASH to HCC, and shed light on therapeutic options in this context.
Collapse
|
11
|
Wang T, Shi J, Li L, Zhou X, Zhang H, Zhang X, Wang Y, Liu L, Sheng L. Single-Cell Transcriptome Analysis Reveals Inter-Tumor Heterogeneity in Bilateral Papillary Thyroid Carcinoma. Front Immunol 2022; 13:840811. [PMID: 35515000 PMCID: PMC9065345 DOI: 10.3389/fimmu.2022.840811] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/23/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The tumor microenvironment (TME) plays a pivotal role in cancer progression in papillary thyroid carcinoma (PTC), yet the composition and the phenotype of cells within the TME in bilateral PTC are poorly understood. METHODS We performed unbiased transcriptome-wide single-cell RNA sequencing (scRNA-seq) analysis on 29,561 cells from 3 pairs of bilateral PTC and 1 non-tumor thyroid sample. The results of the analysis were validated by a large-scale bulk transcriptomic dataset deposited in The Cancer Genome Atlas (TCGA) database. RESULTS Our integrative analysis of thyroid follicular cells revealed 42 signaling pathways enriched in malignant follicular cells, including cytokine-cytokine receptor interaction, PI3K/Akt signaling pathway, mitogen-activated protein kinase (MAPK) signaling pathway, and tumor necrosis factor (TNF) signaling pathway. A 6-gene signature (CXCL3, CXCL1, IL1A, CCL5, TNFRSF12A, and IL18) in the cytokine-cytokine receptor interaction pathway was constructed to predict the prognosis of patients with PTC, with high risk scores being associated with decreased overall survival [hazard ratio (HR) = 3.863, 95% CI = 2.233-6.682, p < 0.001]. Gene set variation analysis (GSVA) indicated that the pathways enriched in bilateral PTC were significantly different, indicating great heterogeneity in bilateral PTC, even with the same BRAF V600E mutation. Comprehensive analysis of T cells revealed that the proportion of CD8+ tissue-resident memory T cells expressing IFNG decreased in tumor samples with advanced N stage. Within the myeloid compartment, the ratio of suppressive M2-like to pro-inflammatory M1-like macrophages increased with advanced disease stage, which was confirmed in the bulk dataset using transcriptomic profiles. In addition, we also identified numerous biologically critical interactions among myeloid cells, T cells, and follicular cells, which were related to T-cell recruitment, M2-like macrophage polarization, malignant follicular cell progression, and T-cell inhibitory signaling. CONCLUSION Our integrative analyses revealed great inter-tumor heterogeneity within the TME in bilateral PTC, which will offer assistance for precise diagnosis and treatment.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, China
| | - Jinyuan Shi
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
- Department of Thyroid Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Luchuan Li
- Department of Thyroid Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoming Zhou
- Department of Scientific Research, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hui Zhang
- Department of Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaofang Zhang
- Department of Pathology, Basic Medical College of Shandong University, Jinan, China
| | - Yong Wang
- Department of Thyroid Surgery, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, China
| | - Lian Liu
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Sheng
- Department of Thyroid Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
12
|
Gade A, Sharma A, Srivastava N, Flora SJS. Surface plasmon resonance: A promising approach for label-free early cancer diagnosis. Clin Chim Acta 2022; 527:79-88. [PMID: 35120900 DOI: 10.1016/j.cca.2022.01.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/12/2022] [Accepted: 01/28/2022] [Indexed: 12/18/2022]
Abstract
Cancer is the second leading cause of death worldwide after cardiovascular disease. The major cause of high mortality is delayed detection. Therefore, detection at an early stage followed by early treatment can mitigate morbidity as well as mortality. The utilization of biomarker-based detection tools helps in early-stage recognition. Fortunately, biomarkers indicating disease status are released in to the circulation. These include traditional marker proteins as well as exosomes, micro-RNA (miRNA) and circulating tumor DNA (ct-DNA). Biosensors are biological and chemical reaction devices that generate signals based on analyte concentration. Due to analyte binding, these devices demonstrate high sensitivity and specificity. This review examines the use of surface plasmon resonance (SPR)-based sensors in the diagnosis of various cancer including those of the breast, prostate, lung, ovary, cervix and pancreas. SPR is a label-free, real-time and non-invasive optical biosensing technology representing a novel diagnostic tool in cancer detection.
Collapse
Affiliation(s)
- Anushree Gade
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow 226002, India
| | - Ankita Sharma
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow 226002, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow 226002, India
| | - S J S Flora
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow 226002, India.
| |
Collapse
|
13
|
Yan C, Liu Q, Jia R. Construction and Validation of a Prognostic Risk Model for Triple-Negative Breast Cancer Based on Autophagy-Related Genes. Front Oncol 2022; 12:829045. [PMID: 35186763 PMCID: PMC8854264 DOI: 10.3389/fonc.2022.829045] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/10/2022] [Indexed: 01/14/2023] Open
Abstract
Background Autophagy plays an important role in triple-negative breast cancer (TNBC). However, the prognostic value of autophagy-related genes (ARGs) in TNBC remains unknown. In this study, we established a survival model to evaluate the prognosis of TNBC patients using ARGs signature. Methods A total of 222 autophagy-related genes were downloaded from The Human Autophagy Database. The RNA-sequencing data and corresponding clinical data of TNBC were obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed autophagy-related genes (DE-ARGs) between normal samples and TNBC samples were determined by the DESeq2 package. Then, univariate Cox, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses were performed. According to the LASSO regression results based on univariate Cox, we identified a prognostic signature for overall survival (OS), which was further validated by using the Gene Expression Omnibus (GEO) cohort. We also found an independent prognostic marker that can predict the clinicopathological features of TNBC. Furthermore, a nomogram was drawn to predict the survival probability of TNBC patients, which could help in clinical decision for TNBC treatment. Finally, we validated the requirement of an ARG in our model for TNBC cell survival and metastasis. Results There are 43 DE-ARGs identified between normal and tumor samples. A risk model for OS using CDKN1A, CTSD, CTSL, EIF4EBP1, TMEM74, and VAMP3 was established based on univariate Cox regression and LASSO regression analysis. Overall survival of TNBC patients was significantly shorter in the high-risk group than in the low-risk group for both the training and validation cohorts. Using the Kaplan–Meier curves and receiver operating characteristic (ROC) curves, we demonstrated the accuracy of the prognostic model. Multivariate Cox regression analysis was used to verify risk score as an independent predictor. Subsequently, a nomogram was proposed to predict 1-, 3-, and 5-year survival for TNBC patients. The calibration curves showed great accuracy of the model for survival prediction. Finally, we found that depletion of EIF4EBP1, one of the ARGs in our model, significantly reduced cell proliferation and metastasis of TNBC cells. Conclusion Based on six ARGs (CDKN1A, CTSD, CTSL, EIF4EBP1, TMEM74, and VAMP3), we developed a risk prediction model that can help clinical doctors effectively predict the survival status of TNBC patients. Our data suggested that EIF4EBP1 might promote the proliferation and migration in TNBC cell lines. These findings provided a novel insight into the vital role of the autophagy-related genes in TNBC and may provide new therapeutic targets for TNBC.
Collapse
Affiliation(s)
- Cheng Yan
- School of Pharmacy, Xinxiang University, Xinxiang, China
- Key Laboratory of Nano-Carbon Modified Film Technology of Henan Province, Xinxiang University, Xinxiang, China
- Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, China
| | - Qingling Liu
- School of Pharmacy, Xinxiang University, Xinxiang, China
| | - Ruoling Jia
- School of Pharmacy, Xinxiang University, Xinxiang, China
- *Correspondence: Ruoling Jia,
| |
Collapse
|
14
|
Huang Y, Li S, Huang S, Tu J, Chen X, Xiao L, Liu B, Yuan X. Comprehensive and Integrative Analysis of Two Novel SARS-CoV-2 Entry Associated Proteases CTSB and CTSL in Healthy Individuals and Cancer Patients. Front Bioeng Biotechnol 2022; 10:780751. [PMID: 35155389 PMCID: PMC8826559 DOI: 10.3389/fbioe.2022.780751] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/05/2022] [Indexed: 11/13/2022] Open
Abstract
More than 200 million people have been infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and 4 million deaths have been reported worldwide to date. Cathepsin B/cathepsin L (CTSB/L) are SARS-CoV-2 entry–associated proteases and facilitate SARS-CoV-2 to infect host cells. However, the expressions of CTSB/L in healthy individuals and cancer patients remain not fully elucidated yet. Here, we comprehensively profiled the expressions and distributions of CTSB/L in human normal tissues, cancer tissues, and cell lines. Moreover, we compared CTSB/L expressions between various cancers and matched normal tissues, and investigated their genetic alteration and prognostic values in pan-cancer. Finally, we also explored the correlation between CTSB/L expressions and immune infiltration. We found that CTSB was highly expressed in most tissues, and CTSL was highly expressed predominantly in the digestive, urinary, and respiratory systems, such as the lungs, liver and gallbladder, and kidney tissues in the translational level. Moreover, cancer patients may be more susceptible to SARS-CoV-2 infection. Our data suggested that CTSB/L are overexpressed in aerodigestive and genitourinary cancers when compared with that in matched normal tissues, and their expressions were closely related to the prognosis of some cancer types. Interestingly, CTSB/L expressions were significantly correlated with immune cell infiltration in manifold cancer tissues and their corresponding normal tissues. In conclusion, our study shows a comprehensive bioinformatic analysis of two important SARS-CoV-2 entry–related proteases, which could provide a potential indication on prevention of SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bo Liu
- *Correspondence: Xianglin Yuan, ; Bo Liu,
| | | |
Collapse
|
15
|
Bui S, Mejia I, Díaz B, Wang Y. Adaptation of the Golgi Apparatus in Cancer Cell Invasion and Metastasis. Front Cell Dev Biol 2021; 9:806482. [PMID: 34957124 PMCID: PMC8703019 DOI: 10.3389/fcell.2021.806482] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
The Golgi apparatus plays a central role in normal cell physiology by promoting cell survival, facilitating proliferation, and enabling cell-cell communication and migration. These roles are partially mediated by well-known Golgi functions, including post-translational modifications, lipid biosynthesis, intracellular trafficking, and protein secretion. In addition, accumulating evidence indicates that the Golgi plays a critical role in sensing and integrating external and internal cues to promote cellular homeostasis. Indeed, the unique structure of the mammalian Golgi can be fine-tuned to adapt different Golgi functions to specific cellular needs. This is particularly relevant in the context of cancer, where unrestrained proliferation and aberrant survival and migration increase the demands in Golgi functions, as well as the need for Golgi-dependent sensing and adaptation to intrinsic and extrinsic stressors. Here, we review and discuss current understanding of how the structure and function of the Golgi apparatus is influenced by oncogenic transformation, and how this adaptation may facilitate cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Sarah Bui
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Isabel Mejia
- Department of Internal Medicine, Division of Medical Hematology and Oncology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Begoña Díaz
- Department of Internal Medicine, Division of Medical Hematology and Oncology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States.,Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| |
Collapse
|
16
|
Identification of Key Genes Associated with Progression and Prognosis of Bladder Cancer through Integrated Bioinformatics Analysis. Cancers (Basel) 2021; 13:cancers13235931. [PMID: 34885040 PMCID: PMC8656554 DOI: 10.3390/cancers13235931] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary Bladder cancer is a heterogeneous disease with high recurrence rates. The current prognostication depends on tumor stage and grade and there is a need for predictive biomarkers that can distinguish between progressive versus non-progressive disease. We have identified a 3-gene signature panel having prognostic value in bladder cancer, which could aid in clinical decision making. Abstract Bladder cancer prognosis remains dismal due to lack of appropriate biomarkers that can predict its progression. The study aims to identify novel prognostic biomarkers associated with the progression of bladder cancer by utilizing three Gene Expression Omnibus (GEO) datasets to screen differentially expressed genes (DEGs). A total of 1516 DEGs were identified between non-muscle invasive and muscle invasive bladder cancer specimens. To identify genes of prognostic value, we performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. A total of seven genes, including CDKN2A, CDC20, CTSV, FOXM1, MAGEA6, KRT23, and S100A9 were confirmed with strong prognostic values in bladder cancer and validated by qRT-PCR conducted in various human bladder cancer cells representing stage-specific disease progression. ULCAN, human protein atlas and The Cancer Genome Atlas datasets were used to confirm the predictive value of these genes in bladder cancer progression. Moreover, Kaplan–Meier analysis and Cox hazard ratio analysis were performed to determine the prognostic role of these genes. Univariate analysis performed on a validation set identified a 3-panel gene set viz. CDKN2A, CTSV and FOXM1 with 95.5% sensitivity and 100% specificity in predicting bladder cancer progression. In summary, our study screened and confirmed a 3-panel biomarker that could accurately predict the progression and prognosis of bladder cancer.
Collapse
|
17
|
Construction and validation of a novel immune and tumor mutation burden-based prognostic model in lung adenocarcinoma. Cancer Immunol Immunother 2021; 71:1183-1197. [PMID: 34635925 DOI: 10.1007/s00262-021-03066-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022]
Abstract
Lung adenocarcinoma (LUAD), the most common type of cancer, is hard to diagnose and has an unfavorable prognosis. Tumor mutation burden (TMB) is a useful predictor and can also determine the efficacy of immunotherapy in various cancers. The present study focused on unraveling the association between immune infiltration and TMB and developing an immune- and TMB-related prognostic model to predict LUAD patients' prognosis. The results revealed that the immune-related prognostic model (IPM) based on TMB was capable of classifying LUAD patients in all cohorts into different risk groups. The IPM was useful and had a significant correlation with LUAD patients' overall survival (OS). Based on the multivariate Cox analysis results, the IPM was proved to be an independent predictive biomarker. Furthermore, the five hub genes and the immune-related model were related to different immune infiltrating cells. The IPM was related to immune checkpoints. At last, an effective nomogram was established to predict LUAD patients' prognosis. To conclude, our IPM is effective in predicting LUAD patients' prognosis and provides novel insights into immunotherapy for LUAD.
Collapse
|
18
|
Functional Implications of the Dynamic Regulation of EpCAM during Epithelial-to-Mesenchymal Transition. Biomolecules 2021; 11:biom11070956. [PMID: 34209658 PMCID: PMC8301972 DOI: 10.3390/biom11070956] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) is a transmembrane glycoprotein expressed in epithelial tissues. EpCAM forms intercellular, homophilic adhesions, modulates epithelial junctional protein complex formation, and promotes epithelial tissue homeostasis. EpCAM is a target of molecular therapies and plays a prominent role in tumor biology. In this review, we focus on the dynamic regulation of EpCAM expression during epithelial-to-mesenchymal transition (EMT) and the functional implications of EpCAM expression on the regulation of EMT. EpCAM is frequently and highly expressed in epithelial cancers, while silenced in mesenchymal cancers. During EMT, EpCAM expression is downregulated by extracellular signal-regulated kinases (ERK) and EMT transcription factors, as well as by regulated intramembrane proteolysis (RIP). The functional impact of EpCAM expression on tumor biology is frequently dependent on the cancer type and predominant oncogenic signaling pathways, suggesting that the role of EpCAM in tumor biology and EMT is multifunctional. Membrane EpCAM is cleaved in cancers and its intracellular domain (EpICD) is transported into the nucleus and binds β-catenin, FHL2, and LEF1. This stimulates gene transcription that promotes growth, cancer stem cell properties, and EMT. EpCAM is also regulated by epidermal growth factor receptor (EGFR) signaling and the EpCAM ectoderm (EpEX) is an EGFR ligand that affects EMT. EpCAM is expressed on circulating tumor and cancer stem cells undergoing EMT and modulates metastases and cancer treatment responses. Future research exploring EpCAM’s role in EMT may reveal additional therapeutic opportunities.
Collapse
|
19
|
Lange C, Machado Weber A, Schmidt R, Schroeder C, Strowitzki T, Germeyer A. Changes in protein expression due to metformin treatment and hyperinsulinemia in a human endometrial cancer cell line. PLoS One 2021; 16:e0248103. [PMID: 33690729 PMCID: PMC7943011 DOI: 10.1371/journal.pone.0248103] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/21/2021] [Indexed: 12/18/2022] Open
Abstract
The incidence of endometrial cancer (EC) has increased over the past years and mainly affects women above the age of 45 years. Metabolic diseases such as obesity and type II diabetes mellitus as well as associated conditions like polycystic ovary syndrome (PCOS), insulin resistance and hyperinsulinemia lead to elevated levels of circulating estrogens. Increased estrogen concentrations, in turn, further trigger the proliferation of endometrial cells and thus promote EC development and progression, especially in the absence of progesterone as seen in postmenopausal women. Elevated blood glucose levels in diabetic patients further contribute to the risk of EC development. Metformin is an insulin-sensitizing biguanide drug, commonly used in the treatment of type II diabetes mellitus, especially in obese patients. Besides its effects on glucose metabolism, metformin displayed anti-cancer effects in various cancer types, including EC. Direct anti-cancer effects of metformin target signaling pathways that are involved in cellular growth and proliferation, e.g. the AKT/PKB/mTOR pathway. Further proteins and pathways have been suggested as potential targets, but the underlying mechanism of action of metformin's anti-cancer activity is still not completely understood. In the present study, the effects of metformin on protein expression were investigated in the human EC cell line HEC-1A using an affinity proteomic approach. Cells were treated with 0.5 mmol/L metformin over a period of 7 days and changes in the expression pattern of 1,300 different proteins were compared to the expression in untreated control cells as well as insulin-treated cells. Insulin treatment (100 ng/mL) was incorporated into the study in order to implement a model for insulin resistance and associated hyperinsulinemia, conditions that are often observed in obese and diabetic patients. Furthermore, the culture medium was supplemented with 10 nmol/L ß-estradiol (E2) during treatments to mimic increased estrogen levels, a common risk factor for EC development. Based on the most prominent and significant changes in expression, a set of 80 proteins was selected and subjected to a more detailed analysis. The data revealed that metformin and insulin targeted similar pathways in the present study and mostly acted on proteins related to proliferation, migration and tumor immune response. These pathways may be affected in a tumor-promoting as well as a tumor-suppressing way by either metformin treatment or insulin supplementation. The consequences for the cells resulting from the detected expression changes were discussed in detail for several proteins. The presented data helps identify potential targets affected by metformin treatment in EC and allows for a better understanding of the mechanism of action of the biguanide drug's anti-cancer activity. However, further investigations are necessary to confirm the observations and conclusions drawn from the presented data after metformin administration, especially for proteins that were regulated in a favorable way, i.e. AKT3, CCND2, CD63, CD81, GFAP, IL5, IL17A, IRF4, PI3, and VTCN1. Further proteins might be of interest, where metformin counteracted unfavorable effects that have been induced by hyperinsulinemia.
Collapse
Affiliation(s)
- Carsten Lange
- Department of Gynecologic Endocrinology and Fertility Disorders, Women’s Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Amanda Machado Weber
- Department of Gynecologic Endocrinology and Fertility Disorders, Women’s Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | | | | | - Thomas Strowitzki
- Department of Gynecologic Endocrinology and Fertility Disorders, Women’s Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Ariane Germeyer
- Department of Gynecologic Endocrinology and Fertility Disorders, Women’s Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Hu L, Han Z, Cheng X, Wang S, Feng Y, Lin Z. Expression Profile Analysis Identifies a Novel Seven Immune-Related Gene Signature to Improve Prognosis Prediction of Glioblastoma. Front Genet 2021; 12:638458. [PMID: 33708242 PMCID: PMC7940837 DOI: 10.3389/fgene.2021.638458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/01/2021] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma multiform (GBM) is a malignant central nervous system cancer with dismal prognosis despite conventional therapies. Scientists have great interest in using immunotherapy for treating GBM because it has shown remarkable potential in many solid tumors, including melanoma, non-small cell lung cancer, and renal cell carcinoma. The gene expression patterns, clinical data of GBM individuals from the Cancer Genome Atlas database (TCGA), and immune-related genes (IRGs) from ImmPort were used to identify differentially expressed IRGs through the Wilcoxon rank-sum test. The association between each IRG and overall survival (OS) of patients was investigated by the univariate Cox regression analysis. LASSO Cox regression assessment was conducted to explore the prognostic potential of the IRGs of GBM and construct a risk score formula. A Kaplan–Meier curve was created to estimate the prognostic role of IRGs. The efficiency of the model was examined according to the area under the receiver operating characteristic (ROC) curve. The TCGA internal dataset and two GEO external datasets were used for model verification. We evaluated IRG expression in GBM and generated a risk model to estimate the prognosis of GBM individuals with seven optimal prognostic expressed IRGs. A landscape of 22 types of tumor-infiltrating immune cells (TIICs) in glioblastoma was identified, and we investigated the link between the seven IRGs and the immune checkpoints. Furthermore, there was a correlation between the IRGs and the infiltration level in GBM. Our data suggested that the seven IRGs identified in this study are not only significant prognostic predictors in GBM patients but can also be utilized to investigate the developmental mechanisms of GBM and in the design of personalized treatments for them.
Collapse
Affiliation(s)
- Li Hu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhibin Han
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xingbo Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Sida Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yumeng Feng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiguo Lin
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
21
|
Suarez-Martinez AD, Sole-Gras M, Dykes SS, Wakefield ZR, Bauer K, Majbour D, Bundy A, Pampo C, Burow ME, Siemann DW, Huang Y, Murfee WL. Bioprinting on Live Tissue for Investigating Cancer Cell Dynamics. Tissue Eng Part A 2020; 27:438-453. [PMID: 33059528 DOI: 10.1089/ten.tea.2020.0190] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A challenge in cancer research is the lack of physiologically responsive in vitro models that enable tracking of cancer cells in tissue-like environments. A model that enables real-time investigation of cancer cell migration, fate, and function during angiogenesis does not exist. Current models, such as 2D or 3D in vitro culturing, can contain multiple cell types, but they do not incorporate the complexity of intact microvascular networks. The objective of this study was to establish a tumor microvasculature model by demonstrating the feasibility of bioprinting cancer cells onto excised mouse tissue. Inkjet-printed DiI+ breast cancer cells on mesometrium tissues from C57Bl/6 mice demonstrated cancer cells' motility and proliferation through time-lapse imaging. Colocalization of DAPI+ nuclei confirmed that DiI+ cancer cells remained intact postprinting. Printed DiI+ 4T1 cells also remained viable after printing on Day 0 and after culture on Day 5. Time-lapse imaging over 5 days enabled tracking of cell migration and proliferation. The number of cells and cell area were significantly increased over time. After culture, cancer cell clusters were colocalized with angiogenic microvessels. The number of vascular islands, defined as disconnected endothelial cell segments, was increased for tissues with bioprinted cancer cells, which suggests that the early stages of angiogenesis were influenced by the presence of cancer cells. Bioprinting cathepsin L knockdown 4T1 cancer cells on wild-type tissues or nontarget 4T1 cells on NG2 knockout tissues served to validate the use of the model for probing tumor cell versus microenvironment changes. These results establish the potential for bioprinting cancer cells onto live mouse tissues to investigate cancer microvascular dynamics within a physiologically relevant microenvironment. Impact statement To keep advancing the cancer biology field, tissue engineering has been focusing on developing in vitro tumor biomimetic models that more closely resemble the native microenvironment. We introduce a novel methodology of bioprinting exogenous cancer cells onto mouse tissue that contains multiple cells and systems within native physiology to investigate cancer cell migration and interactions with nearby microvascular networks. This study corroborates the manipulation of different exogenous cells and host microenvironments that impact cancer cell dynamics in a physiologically relevant tissue. Overall, it is a new approach for delineating the effects of the microenvironment on cancer cells and vice versa.
Collapse
Affiliation(s)
- Ariana D Suarez-Martinez
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Marc Sole-Gras
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida, USA
| | - Samantha S Dykes
- Department of Radiation Oncology, University of Florida, Gainesville, Florida, USA
| | - Zachary R Wakefield
- Department of Radiation Oncology, University of Florida, Gainesville, Florida, USA
| | - Kevin Bauer
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Dima Majbour
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Angela Bundy
- Department of Radiation Oncology, University of Florida, Gainesville, Florida, USA
| | - Christine Pampo
- Department of Radiation Oncology, University of Florida, Gainesville, Florida, USA
| | - Matthew E Burow
- Department of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Dietmar W Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, Florida, USA
| | - Yong Huang
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida, USA
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
22
|
Pranjol MZI, Gutowski NJ, Hannemann M, Whatmore JL. Cathepsin L Induces Proangiogenic Changes in Human Omental Microvascular Endothelial Cells via Activation of the ERK1/2 Pathway. Curr Cancer Drug Targets 2020; 19:231-242. [PMID: 30173647 DOI: 10.2174/1568009618666180831123951] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/09/2018] [Accepted: 08/16/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Metastasis still remains the major cause of therapeutic failure, poor prognosis and high mortality in epithelial ovarian cancer (EOC) patients. Previously, we showed that EOC cells secrete a range of factors with potential pro-angiogenic activity, in disease-relevant human omental microvascular endothelial cells (HOMECs), including the lysosomal protease cathepsin L (CathL). Thus, the aim of this study was to examine potential pro-proliferative and pro-migratory effects of CathL in HOMECs and the activated signalling pathways, and whether these proangiogenic responses are dependent on CathL-catalytic activity. METHODS HOMECs proliferation was investigated using WST-1, BrdU and CyQUANT assays. Cell migration was examined using a Cultrex Cell 96 transwell migration assay. Enzyme activity was assayed at various pHs using the CathL-specific fluorogenic substrate FY-CHO. Activation of cell signalling pathways was tested using a commercially available phosphokinase array and intact cellbased ELISAs. RESULTS We showed for the first time that CathL has a potent pro-proliferative and pro-migratory effect on HOMECs. For instance, CathL significantly increases HOMEC proliferation (134.8±14.7% vs control 100%) and migration (146.6±17.3% vs control 100%). Our data strongly suggest that these proangiogenic effects of CathL are mediated via a non-proteolytic mechanism. Finally, we show that CathL-induced activation of the ERK1/2 pathway is involved in inducing these cellular effects in HOMECs. CONCLUSION These data suggest that CathL acts as an extracellular ligand and plays an important pro-angiogenic, and thus pro-metastatic, role during EOC metastasis to the omentum, by activating the omental microvasculature, and thus can potentially be targeted therapeutically in the future.
Collapse
Affiliation(s)
- Md Zahidul I Pranjol
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, Devon EX1 2LU, United Kingdom.,William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Nicholas J Gutowski
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, Devon EX1 2LU, United Kingdom.,Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon EX2 7JU, United Kingdom
| | - Michael Hannemann
- Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon EX2 7JU, United Kingdom
| | - Jacqueline L Whatmore
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, Devon EX1 2LU, United Kingdom
| |
Collapse
|
23
|
Tabish TA, Pranjol MZI, Whatmore JL, Zhang S. Status and Future Directions of Anti-metastatic Cancer Nanomedicines for the Inhibition of Cathepsin L. FRONTIERS IN NANOTECHNOLOGY 2020. [DOI: 10.3389/fnano.2020.00001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
24
|
Hadad EH, Ahmadzadeh A, Abooali A, Saki Malehi A, Shokouhian M, Saki N. Prognostic role and therapeutic susceptibility of cathepsin in various types of solid tumor and leukemia: A systematic review. J Cell Physiol 2020; 235:7709-7730. [PMID: 32324258 DOI: 10.1002/jcp.29710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/31/2020] [Indexed: 12/18/2022]
Abstract
Cathepsins (CTSs) are multifunctional proteins that can play prominent roles in cancer progression and metastasis. In this systematic review, we compared the prognosis of CTS subtypes overexpression in leukemia and solid tumors, and investigated the effect of different factors on CTS prognosis. We systematically searched published articles indexed in PubMed, Scopus, Cochrane library, ISI Web of Science, and EmBase databases from February 2000 until January 2020. Among the selected leukemia and solid tumors studies, overexpression of CTS subtypes in newly diagnosed and treated patients were with poor prognosis in 43 studies (79.6%) and with good prognosis in 9 studies (16.6%). However, there were 2 studies (3.8%) with either good or poor prognosis, depending on conditions and caner stage and host cell. The relation between CTS and human leukocyte antigen (HLA) in leukemia and solid tumors was mentioned in 7 studies (13%). Overexpression of CTS subtypes in all new case patients had contributed to the induction of poor prognosis. It seems that CTS subtypes, based on the type of cancer and its stage, the type of host cells, and the probable relation with HLA, breed good or poor prognosis in patients with cancer. Therefore, monitoring the overexpression of CTS subtypes and determining the effect of each of these factors on CTS prognosis could be helpful in predicting cancer prognosis both in newly diagnosed or under treatment patients. They could also be useful in finding ways for improving the efficiency of contemporary therapeutic strategies in various types of leukemia and solid tumors.
Collapse
Affiliation(s)
- Elham Homaei Hadad
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ahmad Ahmadzadeh
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Abooali
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amal Saki Malehi
- Department of Biostatistics and Epidemiology, School of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Shokouhian
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
25
|
Hu B, Zhu X, Lu J. Cathepsin A knockdown decreases the proliferation and invasion of A549 lung adenocarcinoma cells. Mol Med Rep 2020; 21:2553-2559. [PMID: 32323791 PMCID: PMC7185279 DOI: 10.3892/mmr.2020.11068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/13/2020] [Indexed: 02/07/2023] Open
Abstract
Cathepsin A (CTSA) is a lysosomal protease that is abnormally expressed in various types of cancer; however, the function of CTSA in lung adenocarcinoma (LUAD) is unknown. The aim of the present study was to investigate the role of CTSA during LUAD development in vitro. The Cancer Genome Atlas (TCGA) database was used to analyze the expression of CTSA mRNA in LUAD tissues. CTSA was significantly upregulated in LUAD tissues compared with normal lung tissues. To explore the effect of CTSA on LUAD in vitro, LUAD A549 cells were transfected with CTSA small interfering RNA and the hallmarks of tumorigenesis were investigated using cell proliferation, cell cycle, wound healing, invasion and western blot assays. Following CTSA knockdown, proliferation of LUAD cells was decreased and an increased proportion of LUAD cells were arrested at the G0/G1 phase, with altered expression of critical cell cycle and proliferative marker proteins, including p53, p21 and proliferating cell nuclear antigen. Moreover, CTSA knockdown decreased the migration and invasion of A549 cells, as determined by wound healing, invasion, and western blotting assays. The expression levels of key proteins involved in epithelial-mesenchymal transition were analyzed by western blotting. CTSA knockdown enhanced the expression of E-cadherin, but decreased the expression of N-cadherin and β-catenin in A549 cells. To the best of our knowledge, the present study suggested for the first time it has been identified that CTSA may serve as a tumor promoter in LUAD, enhancing the malignant progression of LUAD cells by promoting cell proliferation, migration and invasion. The results suggested that CTSA may serve as a novel therapeutic target for LUAD.
Collapse
Affiliation(s)
- Bo Hu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xike Zhu
- Department of Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jibin Lu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
26
|
Mehra S, Panwar R, Thakur B, Yadav R, Kumar M, Singh R, Dash NR, Sahni P, Chauhan SS. Expression and Clinical Implications of Cysteine Cathepsins in Gallbladder Carcinoma. Front Oncol 2019; 9:1239. [PMID: 31824841 PMCID: PMC6883407 DOI: 10.3389/fonc.2019.01239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Gallbladder carcinoma (GBC) exhibits poor prognosis due to its detection at an advanced stage. Upregulation of lysosomal cysteine proteases cathepsin L (CTSL) and cathepsin B (CTSB) has been implicated in several tumorigenic processes. However, no such information in GBC was available. Therefore, the present study was planned to investigate the expression and clinical significance of these cathepsins in GBC. Methods: Activities of CTSL and CTSB were assayed in the gallbladder (GB) tissues obtained from GBC patients (n = 43) and control subjects (n = 69). Protein and mRNA levels were quantified using immunohistochemistry and real-time PCR (qPCR), respectively. Finally, serum levels of CTSL and CTSB were estimated by ELISA. Receiver operating characteristic (ROC) curve analysis was used for the assessment of sensitivity, specificity, and diagnostic accuracy of these cysteine cathepsins in GBC. The association of combined CTSL and CTSB activity with overall survival was assessed using Kaplan Meier survival analysis. Results: The expression and activity of both CTSL and CTSB were significantly increased (p < 0.050) in tumors of GBC patients as compared to controls. Enzymatic activity of CTSL+B and CTSB exhibited a strong positive association with tumor stage and lymph node involvement in GBC (p < 0.050). Interestingly, the elevated activity of combined CTSL+B was also associated with increased mortality in these patients. Furthermore, significantly enhanced levels of serum CTSL and CTSB were also observed in GBC (p < 0.050) as compared to controls. ROC analysis revealed high diagnostic significance of serum CTSB and CTSL for distinguishing GBC patients from controls with an area under the curve (AUC) of 82 and 77%, respectively. Conclusion: This study, for the first time, demonstrates the clinical significance of CTSL and CTSB overexpression in GBC. Our findings may help improve the clinical management of this carcinoma.
Collapse
Affiliation(s)
- Siddharth Mehra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Rajesh Panwar
- Department of G.I. Surgery and Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Bhaskar Thakur
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Rajni Yadav
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Manish Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ratnakar Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Nihar Ranjan Dash
- Department of G.I. Surgery and Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Peush Sahni
- Department of G.I. Surgery and Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Shyam S Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
27
|
Rudzińska M, Parodi A, Soond SM, Vinarov AZ, Korolev DO, Morozov AO, Daglioglu C, Tutar Y, Zamyatnin AA. The Role of Cysteine Cathepsins in Cancer Progression and Drug Resistance. Int J Mol Sci 2019; 20:3602. [PMID: 31340550 PMCID: PMC6678516 DOI: 10.3390/ijms20143602] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022] Open
Abstract
Cysteine cathepsins are lysosomal enzymes belonging to the papain family. Their expression is misregulated in a wide variety of tumors, and ample data prove their involvement in cancer progression, angiogenesis, metastasis, and in the occurrence of drug resistance. However, while their overexpression is usually associated with highly aggressive tumor phenotypes, their mechanistic role in cancer progression is still to be determined to develop new therapeutic strategies. In this review, we highlight the literature related to the role of the cysteine cathepsins in cancer biology, with particular emphasis on their input into tumor biology.
Collapse
Affiliation(s)
- Magdalena Rudzińska
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Alessandro Parodi
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Surinder M Soond
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Andrey Z Vinarov
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia
| | - Dmitry O Korolev
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia
| | - Andrey O Morozov
- Institute for Urology and Reproductive Health, Sechenov University, 119992 Moscow, Russia
| | - Cenk Daglioglu
- Izmir Institute of Technology, Faculty of Science, Department of Molecular Biology and Genetics, 35430 Urla/Izmir, Turkey
| | - Yusuf Tutar
- Faculty of Pharmacy, University of Health Sciences, 34668 Istanbul, Turkey
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia.
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
| |
Collapse
|
28
|
Tenora L, Alt J, Dash RP, Gadiano AJ, Novotná K, Veeravalli V, Lam J, Kirkpatrick QR, Lemberg KM, Majer P, Rais R, Slusher BS. Tumor-Targeted Delivery of 6-Diazo-5-oxo-l-norleucine (DON) Using Substituted Acetylated Lysine Prodrugs. J Med Chem 2019; 62:3524-3538. [PMID: 30892035 DOI: 10.1021/acs.jmedchem.8b02009] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
6-Diazo-5-oxo-l-norleucine (DON) is a glutamine antagonist with robust anticancer efficacy; however, its therapeutic potential was hampered by its biodistribution and toxicity to normal tissues, specifically gastrointestinal (GI) tissues. To circumvent DON's toxicity, we synthesized a series of tumor-targeted DON prodrugs designed to circulate inert in plasma and preferentially activate over DON in tumor. Our best prodrug 6 (isopropyl 2-(6-acetamido-2-(adamantane-1-carboxamido)hexanamido)-6-diazo-5-oxohexanoate) showed stability in plasma, liver, and intestinal homogenates yet was readily cleaved to DON in P493B lymphoma cells, exhibiting a 55-fold enhanced tumor cell-to-plasma ratio versus that of DON and resulting in a dose-dependent inhibition of cell proliferation. Using carboxylesterase 1 knockout mice that were shown to mimic human prodrug metabolism, systemic administration of 6 delivered 11-fold higher DON exposure to tumor (target tissue; AUC0- t = 5.1 nmol h/g) versus GI tissues (toxicity tissue; AUC0- t = 0.45 nmol h/g). In summary, these studies describe the discovery of a glutamine antagonist prodrug that provides selective tumor exposure.
Collapse
Affiliation(s)
- Lukáš Tenora
- Institute of Organic Chemistry and Biochemistry , Academy of Sciences of the Czech Republic v.v.i. , Prague 166 10 , Czech Republic
| | | | | | | | - Kateřina Novotná
- Institute of Organic Chemistry and Biochemistry , Academy of Sciences of the Czech Republic v.v.i. , Prague 166 10 , Czech Republic
| | | | | | | | | | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry , Academy of Sciences of the Czech Republic v.v.i. , Prague 166 10 , Czech Republic
| | | | | |
Collapse
|
29
|
Wang W, Xiong Y, Ding X, Wang L, Zhao Y, Fei Y, Zhu Y, Shen X, Tan C, Liang Z. Cathepsin L activated by mutant p53 and Egr-1 promotes ionizing radiation-induced EMT in human NSCLC. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:61. [PMID: 30732622 PMCID: PMC6367810 DOI: 10.1186/s13046-019-1054-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/23/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Ionizing radiation (IR) is one of the major clinical therapies of cancer, although it increases the epithelial-mesenchymal transition (EMT) of non-small cell lung cancer (NSCLC), unexpectedly. The cellular and molecular mechanisms underlying this role are not completely understood. METHODS We used NSCLC cell lines as well as tumor specimens from 78 patients with NSCLC to evaluate p53, Cathepsin L (CTSL) and EMT phenotypic changes. Xenograft models was also utilized to examine the roles of mutant p53 (mut-p53) and CTSL in regulating IR-induced EMT of NSCLC. RESULTS Expression of CTSL was markedly increased in human NSCLC tissues with mutant p53 (mut-p53), and p53 mutation positively correlated with metastasis of NSCLC patients. In human non-small cell lung cancer cell line, H1299 cells transfected with various p53 lentivirus vectors, mut-p53 could promote the invasion and motility of cells under IR, mainly through the EMT. This EMT process was induced by elevating intranuclear CTSL which was regulated by mut-p53 depending on Early growth response protein-1 (Egr-1) activation. In the subcutaneous tumor xenograft model, IR promoted the EMT of the cancer cells in the presence of mut-p53, owing to increase expression and nuclear transition of its downstream protein CTSL. CONCLUSION Taken together, these data reveal the role of the mut-p53/Egr-1/CTSL axis in regulating the signaling pathway responsible for IR-induced EMT.
Collapse
Affiliation(s)
- Wenjuan Wang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, China.,Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Yajie Xiong
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, China
| | - Xinyuan Ding
- Institutes for Translational Medicine, Soochow University, Suzhou, 215000, China.,Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215000, China
| | - Long Wang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, China
| | - Yifan Zhao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, China
| | - Yao Fei
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, China
| | - Ying Zhu
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, China
| | - Xiao Shen
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, China
| | - Caihong Tan
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, China. .,Department of Pharmacy, Affiliated Hospital of Jiangsu University, Zhenjiang, 212000, China.
| | - Zhongqin Liang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
30
|
Abstract
Proteases drive the life cycle of all proteins, ensuring the transportation and activation of newly minted, would-be proteins into their functional form while recycling spent or unneeded proteins. Far from their image as engines of protein digestion, proteases play fundamental roles in basic physiology and regulation at multiple levels of systems biology. Proteases are intimately associated with disease and modulation of proteolytic activity is the presumed target for successful therapeutics. "Proteases: Pivot Points in Functional Proteomics" examines the crucial roles of proteolysis across a wide range of physiological processes and diseases. The existing and potential impacts of proteolysis-related activity on drug and biomarker development are presented in detail. All told the decisive roles of proteases in four major categories comprising 23 separate subcategories are addressed. Within this construct, 15 sets of subject-specific, tabulated data are presented that include identification of proteases, protease inhibitors, substrates, and their actions. Said data are derived from and confirmed by over 300 references. Cross comparison of datasets indicates that proteases, their inhibitors/promoters and substrates intersect over a range of physiological processes and diseases, both chronic and pathogenic. Indeed, "Proteases: Pivot Points …" closes by dramatizing this very point through association of (pro)Thrombin and Fibrin(ogen) with: hemostasis, innate immunity, cardiovascular and metabolic disease, cancer, neurodegeneration, and bacterial self-defense.
Collapse
Affiliation(s)
- Ingrid M Verhamme
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Sarah E Leonard
- Chemical and Biomolecular Engineering, University of Illinois Champaign-Urbana School of Chemical Sciences, Champaign, IL, USA
| | - Ray C Perkins
- New Liberty Proteomics Corporation, New Liberty, KY, USA.
| |
Collapse
|
31
|
Pišlar A, Jewett A, Kos J. Cysteine cathepsins: Their biological and molecular significance in cancer stem cells. Semin Cancer Biol 2018; 53:168-177. [DOI: 10.1016/j.semcancer.2018.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022]
|
32
|
Down-regulation of cathepsin S and matrix metalloproteinase-9 via Src, a non-receptor tyrosine kinase, suppresses triple-negative breast cancer growth and metastasis. Exp Mol Med 2018; 50:1-14. [PMID: 30185799 PMCID: PMC6123788 DOI: 10.1038/s12276-018-0135-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/28/2018] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly metastatic breast cancer with poor prognosis. In the present study, we demonstrated that Src, a non-receptor tyrosine kinase, might provide an effective therapeutic strategy to overcome TNBC invasion and metastasis, which are mediated via the synergistic action of the lysosomal enzyme cathepsin S (CTSS) and gelatinase MMP-9. Knock-down of MMP-9 and CTSS using siRNAs resulted in a synergistic suppression of MDA-MB-231 cell invasion, which was similarly observed with pharmacological inhibitors. During the screening of new drug candidates that suppress both CTSS and MMP-9, BJ-2302, a novel 7-azaindolin-2-one derivative, was discovered. Src, an upstream activator of both pathways (PI3K/Akt and Ras/Raf/ERK) responsible for the expression of CTSS and MMP-9, was identified as a high-affinity target of BJ-2302 (IC90: 3.23 µM) through a Src kinase assay and a drug affinity responsive target stability (DARTS) assay. BJ-2302 effectively suppressed MDA-MB-231 cell invasion (Matrigel invasion assay) and metastasis (chorioallantoic membrane assay xenografted with MDA-MB-231-luc2-tdTomato cancer cells). Unlike Z-FL-COCHO (potent CTSS inhibitor), BJ-2302 did not induce any cytotoxicity in MCF-10A normal breast epithelial cells. Additionally, BJ-2302 (1 mg/kg) strongly suppressed TNBC cell proliferation in vitro and tumor growth in a xenograft mouse tumor model. The anti-metastatic and anti-tumor effects of BJ-2302 were superior to those of Z-FL-COCHO (1 mg/kg) or batimastat (30 mg/kg), a pan-MMP inhibitor. In summary, inhibition of Src kinase suppressed TNBC tumor growth and metastasis, and Src inhibitors such as BJ-2302 may constitute a novel therapeutic tool to treat breast cancer that expresses high levels of CTSS and MMP-9.
Collapse
|
33
|
Cysteine cathepsins as a prospective target for anticancer therapies-current progress and prospects. Biochimie 2018; 151:85-106. [PMID: 29870804 DOI: 10.1016/j.biochi.2018.05.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/31/2018] [Indexed: 02/08/2023]
Abstract
Cysteine cathepsins (CTS), being involved in both physiological and pathological processes, play an important role in the human body. During the last 30 years, it has been shown that CTS are highly upregulated in a wide variety of cancer types although they have received a little attention as a potential therapeutic target as compared to serine or metalloproteinases. Studies on the increasing problem of neoplastic progression have revealed that secretion of cell-surface- and intracellular cysteine proteases is aberrant in tumor cells and has an impact on their growth, invasion, and metastasis by taking part in tumor angiogenesis, in apoptosis, and in events of inflammatory and immune responses. Considering the role of CTS in carcinogenesis, inhibition of these enzymes becomes an attractive strategy for cancer therapy. The downregulation of natural CTS inhibitors (CTSsis), such as cystatins, observed in various types of cancer, supports this claim. The intention of this review is to highlight the relationship of CTS with cancer and to present illustrations that explain how some of their inhibitors affect processes related to neoplastic progression.
Collapse
|
34
|
Leto G, Crescimanno M, Flandina C. On the role of cystatin C in cancer progression. Life Sci 2018; 202:152-160. [DOI: 10.1016/j.lfs.2018.04.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/17/2018] [Accepted: 04/11/2018] [Indexed: 02/06/2023]
|
35
|
Wang X, He Y, Ye Y, Zhao X, Deng S, He G, Zhu H, Xu N, Liang S. SILAC-based quantitative MS approach for real-time recording protein-mediated cell-cell interactions. Sci Rep 2018; 8:8441. [PMID: 29855483 PMCID: PMC5981645 DOI: 10.1038/s41598-018-26262-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/04/2018] [Indexed: 02/05/2023] Open
Abstract
In tumor microenvironment, interactions among multiple cell types are critical for cancer progression. To understand the molecular mechanisms of these complex interplays, the secreted protein analysis between malignant cancer cells and the surrounding nonmalignant stroma is a good viewpoint to investigate cell-cell interactions. Here, we developed two stable isotope labeling of amino acids in cell culture (SILAC)-based mass spectrometry (MS)/MS approaches termed spike-in SILAC and triple-SILAC to quantify changes of protein secretion level in a cell co-cultured system. Within the co-culture system of CT26 and Ana-1 cells, the spike-in SILAC and triple-SILAC MS approaches are sensitive to quantitatively measure protein secretion changes. Three representative quantified proteins (Galectin-1, Cathepsin L1 and Thrombospondin-1) by two SILAC-based MS methods were further validated by Western blotting, and the coming result matched well with SILACs’. We further applied these two SILACs to human cell lines, NCM460 and HT29 co-culture system, for evaluating the feasibility, which confirmed the spike-in and triple SILAC were capable of monitoring the changed secreted proteins of human cell lines. Considering these two strategies in time consuming, sample complexity and proteome coverage, the triple-SILAC way shows more efficiency and economy for real-time recording secreted protein levels in tumor microenvironment.
Collapse
Affiliation(s)
- Xixi Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China.,Chengdu Center for Disease Control and Prevention, Chengdu, 610041, P. R. China
| | - Yu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Yang Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Xinyu Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Shi Deng
- Department of Urinary Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, P. R. China
| | - Gu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100021, P. R. China
| | - Ningzhi Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China.,Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, 100021, P. R. China
| | - Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and National Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P. R. China.
| |
Collapse
|
36
|
Pandey G, Bakhshi S, Thakur B, Jain P, Chauhan SS. Prognostic significance of cathepsin L expression in pediatric acute myeloid leukemia. Leuk Lymphoma 2018; 59:2175-2187. [PMID: 29345177 DOI: 10.1080/10428194.2017.1422865] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Overexpression of cathepsin L (CTSL), an endolysosomal cysteine protease, is associated with inferior survival of patients with various human malignancies. We evaluated the expression/activity of CTSL in peripheral blood mononuclear cells (PBMCs) and bone marrow mononuclear cells (BMMCs) of 103 pediatric acute myeloid leukemia (AML) patients to assess its prognostic significance in this malignancy. Thirty-five healthy siblings of patients served as controls. Our results revealed significantly higher CTSL activity (p < .0001), protein (p < .05), and mRNA levels (p < .01) in both PBMCs and BMMCs of patients as compared with controls. BMMCs displayed higher activity of CTSL than PBMCs (p < .01). A dramatic reduction in CTSL activity was recorded after chemotherapy in a significant proportion (74%) of patients (p < .0001). By multivariate analysis, CTSL in BMMCs emerged as a strong independent prognostic marker for overall survival (OS) (p = .004). Thus, our results suggest the potential utility of CTSL in predicting the outcome of pediatric AML.
Collapse
Affiliation(s)
- Garima Pandey
- a Department of Biochemistry , AIIMS , New Delhi , India
| | - Sameer Bakhshi
- b Department of Medical Oncology , AIIMS , New Delhi , India
| | - Bhaskar Thakur
- c Department of Biostatistics , AIIMS , New Delhi , India
| | - Prerna Jain
- a Department of Biochemistry , AIIMS , New Delhi , India
| | | |
Collapse
|
37
|
Sui H, Shi C, Yan Z, Wu M. Overexpression of Cathepsin L is associated with chemoresistance and invasion of epithelial ovarian cancer. Oncotarget 2018; 7:45995-46001. [PMID: 27351223 PMCID: PMC5216776 DOI: 10.18632/oncotarget.10276] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/03/2016] [Indexed: 01/07/2023] Open
Abstract
Paclitaxel is recommended as a first-line chemotherapeutic agent against, ovarian cancer, however, the development of chemoresistance is a major obstacle in patients with aggressive ovarian cancer and results in recurrence after conventional therapy. The key molecule or mechanism associated with paclitaxel resistance in ovarian cancer still remains unclear. Cathepsin L (CTSL) is overexpressed in various cancers, however, the association between CTSL expression and paclitaxel resistance remains unclear. In the present study, we investigated the role of CTSL in paclitaxel-resistant SKOV3/TAX cells by CTSL silencing. Expression of CTSL was examined by immunohistochemistry and qRT-PCR in 58 clinical samples, and in SKOV3 cells and SKOV3/TAX cells. Effects of CTSL knockdown on ovarian cancer cell proliferation, apoptosis, migration, and invasion were also studied. The IHC and real-time PCR results showed that the difference of CTSL expression between ovarian cancer and the adjacent non-tumourous ovarian tissues was statistically significant. Western blot analysis showed that the CTSL was overexpressed in SKOV3/TAX cells and weakly detectable in paclitaxel-sensitive SKOV3 cells. Knocking-down of CTSL in ovarian cancer cells could decrease cell proliferation, migration, and invasion, and potentiate apoptosis induced by paclitaxel, suggesting CTSL may contribute to Paclitaxel resistance in ovarian cancer.
Collapse
Affiliation(s)
- Hongying Sui
- Department of Gynecological Oncology, Hunan cancer hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan, China
| | - Caixia Shi
- Department of Gynecological Oncology, Hunan cancer hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan, China
| | - Zhipeng Yan
- Department of Gynecological Oncology, Hunan cancer hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan, China
| | - Mei Wu
- Department of Gynecological Oncology, Hunan cancer hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan, China
| |
Collapse
|
38
|
Sigloch FC, Tholen M, Gomez-Auli A, Biniossek ML, Reinheckel T, Schilling O. Proteomic analysis of lung metastases in a murine breast cancer model reveals divergent influence of CTSB and CTSL overexpression. J Cancer 2017; 8:4065-4074. [PMID: 29187882 PMCID: PMC5706009 DOI: 10.7150/jca.21401] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/09/2017] [Indexed: 12/26/2022] Open
Abstract
Studies in the MMTV-PyMT (PyMT) breast cancer mouse model have shown a strong influence of the lysosomal cysteine cathepsins B or L on lung metastasis formation. Transgenic expression of human CTSB (tgCTSB) or CTSL (tgCTSL) both led to similar metastatic phenotypes with increased metastatic burden in the PyMT mice. However, recent studies in other tumor models proved marked differences in effects of either cathepsin on the proteome composition. We sought to analyze and compare proteome changes in the metastatic proteome of PyMT mice expressing either tgCTSB or tgCTSL to evaluate similarities and differences in those models. Performing an explorative, quantitative proteome comparison based on LC-MS/MS, we identified up to 3,000 proteins from murine lung metastases in three independent biological replicates per genotype. In both cases, when compared to wild-type (WT) mice, we noticed a pronounced impact of transgene cathepsin expression on the metastasis proteome. Highlights include increased moesin, integrin beta 1 and vinexin levels in the tgCTSB dataset and increased saposin and granulin levels in the tgCTSL dataset. Importantly, non-supervised hierarchical clustering clearly separated tgCTSB vs. tgCTSL induced proteome changes. In summary, tgCTSB and tgCTSL both display a strong and distinct impact on proteome composition of lung macrometastases in the PyMT model. Our observations suggest that they impact malignant behavior in distinct ways, thus further emphasizing interest into their tumor-contextual functionality.
Collapse
Affiliation(s)
- Florian Christoph Sigloch
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, D-79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, D-79104 Freiburg, Germany
| | - Martina Tholen
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, D-79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, D-79104 Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, D-79104 Freiburg, Germany.,Present address: Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Alejandro Gomez-Auli
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, D-79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, D-79104 Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, D-79104 Freiburg, Germany
| | - Martin Lothar Biniossek
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, D-79104 Freiburg, Germany
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, D-79104 Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, D-79104 Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
39
|
Guo S, Xue Y, He Q, He X, Guo K, Dong P, Yao K, Yang G, Chen D, Li Z, Li X, Qin Z, Liu Z, Cheng W, Guo C, Zhang M, Han H, Zhou F. Preoperative serum cystatin-C as a potential biomarker for prognosis of renal cell carcinoma. PLoS One 2017; 12:e0178823. [PMID: 28586363 PMCID: PMC5460820 DOI: 10.1371/journal.pone.0178823] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 05/21/2017] [Indexed: 11/18/2022] Open
Abstract
PURPOSE The prognostic value of serum cystatin-C (Cys-C) in renal cell carcinoma (RCC) remains unknown. The purpose of this study is to explore the prognostic value of Cys-C for RCC patients. PATIENTS AND METHODS The levels of preoperative Cys-C, creatinine (CRE) and estimated glomerular filtration rate (e-GFR) were retrospectively collected in 325 RCC patients undergoing surgery. The cutoff values of Cys-C, CRE and e-GFR were determined by the standardized Cutoff Finder algorithm. The receiver operating characteristic (ROC) curve and pairwise comparison were performed to compare the three variables. Univariate and multivariate Cox regression analyses were performed to investigate the prognostic value of serum Cys-C in RCC. RESULTS Based on the analysis of Cutoff Finder algorithm, ROC curve and pairwise comparison, the preoperative Cys-C was superior to CRE and e-GFR as a predictive factor in RCC. Multivariate Cox regression analyses showed that high preoperative Cys-C (>1.09 mg/L) was significantly associated with shorter overall survival (OS) in all RCC patients (hazard ratio [HR], 1.59; P = 0.012), patients at pT1-2 (P<0.001), pN0 (P<0.001) and pM0 stages (P<0.001). Moreover, Multivariate Cox regression analyses also showed that in the 306 patients without metastasis, high preoperative Cys-C was also associated with shorter disease-free survival (DFS) (HR, 3.50; P = 0.013). CONCLUSIONS An elevated preoperative Cys-C level was demonstrated to be related with worse survival in patients with RCC. Measuring preoperative serum Cys-C might be a simple way for finding poor prognostic patients and patients with elevated preoperative Cys-C level should be more closely followed up.
Collapse
Affiliation(s)
- Shengjie Guo
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yunfei Xue
- Medicine school of Sun Yat-Sen University, Guangzhou, China
| | - Qiuming He
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiaobo He
- Department of Medical Oncology, the Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Kunbin Guo
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pei Dong
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Kai Yao
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Guangwei Yang
- Medicine school of Sun Yat-Sen University, Guangzhou, China
| | - Dong Chen
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zaishang Li
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiangdong Li
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zike Qin
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhuowei Liu
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenjie Cheng
- Department of Urology, the Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Chao Guo
- The Jiangcheng Branch of Yangjiang People’s Hospital, Yangjiang, Guangdong, China
| | - Meng Zhang
- Medicine school of Sun Yat-Sen University, Guangzhou, China
| | - Hui Han
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- * E-mail: (HH); (FZ)
| | - Fangjian Zhou
- Department of Urology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- * E-mail: (HH); (FZ)
| |
Collapse
|
40
|
Tian L, Xie K, Sheng D, Wan X, Zhu G. Antiangiogenic effects of oridonin. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:192. [PMID: 28376864 PMCID: PMC5379751 DOI: 10.1186/s12906-017-1706-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 03/24/2017] [Indexed: 12/18/2022]
Abstract
Background Oridonin, the major terpene found in Rabdosia rubescens (Henmsl.) Hara, is widely used as a dietary supplement and therapeutic drug. Oridonin has been proven to possess good anti-tumour activity, but little is known about its effect on angiogenesis. The aim of this study was to investigate the antiangiogenic effects of oridonin in vivo and in vitro and prove that oridonin anti-tumour activity is based on suppressing angiogenesis. Methods In vitro, the antiangiogenesis effect was studied by proliferation, apoptosis, migration, invasion, and tube formation experiments on human umbilical vascular endothelial cells (HUVECs). In vivo, using the Tg (fli1: GFP) zebrafish model, the embryonic vasculogenesis and postnatal regeneration were evaluated. The vascular endothelial growth factor (VEGF) signalling pathway gene expressions were assessed by reverse transcription-polymerase chain reaction (RT-PCR). Furthermore, the inhibition effects on tumour growth and metastasis were observed using a xenograft zebrafish tumour model and xenograft nude mouse tumour model. Angiogenesis was assayed by immunostaining with cluster of differentiation 31. Importantly, the proteins were identified as being differentially expressed in an in vivo model by two-dimensional electrophoresis-mass spectrometry (2D–MS) and western blot (WB). Results The results indicated that oridonin inhibited HUVEC proliferation, migration, invasion, and tube formation and induced cell apoptosis. Oridonin inhibited zebrafish angiogenesis during embryonic development and tail fin regeneration. RT-PCR showed that oridonin decreased the VEGFA, VEGFR2, and VEGFR3 expressions in zebrafish, while the TP53 expression increased. Moreover, oridonin had strong effects on tumour growth and metastasis in vivo. 2D–MS identified a total of 50 proteins differentially expressed (17 up-expressed, 28 down-expressed). Lastly, WB showed that Claudin 1, Claudin 4, and Claudin 7 were closely related to tumour growth and metastasis. Conclusion This study demonstrated that oridonin could inhibit tumour growth and metastasis, which mainly based on oridonin antiangiogenic effects. Claudin 1, Claudin 4, and Claudin 7 were the main contributors to the mechanism.
Collapse
|
41
|
Tumor microenvironment: The culprit for ovarian cancer metastasis? Cancer Lett 2016; 377:174-82. [DOI: 10.1016/j.canlet.2016.04.038] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 04/23/2016] [Accepted: 04/25/2016] [Indexed: 01/08/2023]
|
42
|
Knockdown of cathepsin L sensitizes ovarian cancer cells to chemotherapy. Oncol Lett 2016; 11:4235-4239. [PMID: 27313771 DOI: 10.3892/ol.2016.4494] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/16/2016] [Indexed: 01/06/2023] Open
Abstract
Ovarian cancer is a leading gynecological malignancy associated with high mortality. The development of acquired drug resistance is the primary cause of chemotherapy failure in the treatment of ovarian cancer. To examine the mechanism underlying paclitaxel resistance in ovarian cancer and attempt to reverse it, the present study induced a TAX-resistant ovarian cancer cell line, SKOV3/TAX. Cathepsin L (CTSL) has been found to be overexpressed in ovarian cancer. The aim of the present study was to investigate the possible involvement of CTSL in the development of TAX resistance in ovarian cancer. CTSL expression was knocked down in SKOV3 ovarian cancer cells and their phenotypic changes were analyzed. The effects of silenced CTSL on the resistant cell line were investigated by proliferation and apoptosis analysis compared with control SKOV3 cells. CTSL was more highly expressed in SKOV3/TAX cells compared with SKOV3 cells. Paclitaxel treatment downregulated the expression of CTSL in SKOV-3 but not in the paclitaxel-resistant SKOV3/TAX cells. CTSL small hairpin RNA (shRNA) knockdown significantly potentiated apoptosis induced by paclitaxel compared with SKOV3/TAX cells transfected with control shRNA, suggesting that CTSL contributes to paclitaxel resistance in ovarian cancer cells and that CTSL silencing can enhance paclitaxel-mediated cell apoptosis. Thus, CTSL should be explored as a candidate of therapeutic target for modulating paclitaxel sensitivity in ovarian cancer.
Collapse
|
43
|
Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, Soma M, Okamoto H, Oitate M, Arakawa S, Hirai T, Atsumi R, Nakada T, Hayakawa I, Abe Y, Agatsuma T. DS-8201a, A Novel HER2-Targeting ADC with a Novel DNA Topoisomerase I Inhibitor, Demonstrates a Promising Antitumor Efficacy with Differentiation from T-DM1. Clin Cancer Res 2016; 22:5097-5108. [DOI: 10.1158/1078-0432.ccr-15-2822] [Citation(s) in RCA: 353] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/20/2016] [Indexed: 12/31/2022]
|
44
|
Ha Y, Choi HK. Recent conjugation strategies of small organic fluorophores and ligands for cancer-specific bioimaging. Chem Biol Interact 2016; 248:36-51. [DOI: 10.1016/j.cbi.2016.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/02/2016] [Accepted: 02/08/2016] [Indexed: 01/03/2023]
|
45
|
Minchenko OH, Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine, Kyiv. GLUCOSE DEPRIVATION AFFECTS THE EXPRESSION OF LONP1 AND CATHEPSINS IN IRE1 KNOCKDOWN U87 GLIOMA CELLS. BIOTECHNOLOGIA ACTA 2016. [DOI: 10.15407/biotech9.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
46
|
Yu F, Chen Z, Wang B, Jin Z, Hou Y, Ma S, Liu X. The role of lysosome in cell death regulation. Tumour Biol 2015; 37:1427-36. [DOI: 10.1007/s13277-015-4516-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/25/2015] [Indexed: 02/01/2023] Open
|
47
|
Lysosomal cysteine peptidases – Molecules signaling tumor cell death and survival. Semin Cancer Biol 2015; 35:168-79. [DOI: 10.1016/j.semcancer.2015.08.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 07/31/2015] [Accepted: 08/03/2015] [Indexed: 12/18/2022]
|
48
|
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: 479] [Impact Index Per Article: 47.9] [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
|
49
|
The Potential Role of the Proteases Cathepsin D and Cathepsin L in the Progression and Metastasis of Epithelial Ovarian Cancer. Biomolecules 2015; 5:3260-79. [PMID: 26610586 PMCID: PMC4693277 DOI: 10.3390/biom5043260] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/13/2015] [Indexed: 02/08/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the leading cause of death from gynecologic malignancies and has a poor prognosis due to relatively unspecific early symptoms, and thus often advanced stage, metastasized cancer at presentation. Metastasis of EOC occurs primarily through the transcoelomic route whereby exfoliated tumor cells disseminate within the abdominal cavity, particularly to the omentum. Primary and metastatic tumor growth requires a pool of proangiogenic factors in the microenvironment which propagate new vasculature in the growing cancer. Recent evidence suggests that proangiogenic factors other than the widely known, potent angiogenic factor vascular endothelial growth factor may mediate growth and metastasis of ovarian cancer. In this review we examine the role of some of these alternative factors, specifically cathepsin D and cathepsin L.
Collapse
|
50
|
Lęgowska M, Wysocka M, Burster T, Pikuła M, Rolka K, Lesner A. Ultrasensitive internally quenched substrates of human cathepsin L. Anal Biochem 2014; 466:30-7. [PMID: 25151941 DOI: 10.1016/j.ab.2014.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/08/2014] [Accepted: 08/12/2014] [Indexed: 10/24/2022]
Abstract
Internally quenched cathepsin L (Cat L) substrate ABZ-Bip-Arg-Ala-Gln-Tyr(3-NO2)-NH2 with high specificity constant (kcat/KM=2.6×10(7)M(-1)s(-1)) was synthesized. The resultant compound displayed high selectivity over other members of the cathepsin family (B, S, X, V, C, K, H, F, D, and A). Activity of Cat L at picomolar (pM) concentrations was found using this substrate. Moreover, it was established that the presence of the selective Cat L inhibitor suppressed the proteolysis of the substrate to a non-detectable level. Incubation of the synthesized compound with a cell lysate of healthy and cancer cell lines indicated significant differences in Cat L activity. Based on the obtained results, it is proposed that this substrate could be used for selective monitoring of Cat L activity in biological systems.
Collapse
Affiliation(s)
- Monika Lęgowska
- Department of Biochemistry, Faculty of Chemistry, University of Gdansk, 80-952 Gdansk, Poland
| | - Magdalena Wysocka
- Department of Biochemistry, Faculty of Chemistry, University of Gdansk, 80-952 Gdansk, Poland
| | - Timo Burster
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| | - Michał Pikuła
- Department of Clinical Immunology and Transplantology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Krzysztof Rolka
- Department of Biochemistry, Faculty of Chemistry, University of Gdansk, 80-952 Gdansk, Poland
| | - Adam Lesner
- Department of Biochemistry, Faculty of Chemistry, University of Gdansk, 80-952 Gdansk, Poland.
| |
Collapse
|