1
|
Bogdanov AA, Solovyev ID, Savitsky AP. Sensors for Proteolytic Activity Visualization and Their Application in Animal Models of Human Diseases. BIOCHEMISTRY (MOSCOW) 2019; 84:S1-S18. [PMID: 31213192 DOI: 10.1134/s0006297919140013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Various sensors designed for optical and photo(opto)acoustic imaging in living systems are becoming essential components of basic and applied biomedical research. Some of them including those developed for determining enzyme activity in vivo are becoming commercially available. These sensors can be used for various fluorescent signal detection methods: from whole body tomography to endoscopy with miniature cameras. Sensor molecules including enzyme-cleavable macromolecules carrying multiple quenched near-infrared fluorophores are able to deliver their payload in vivo and have long circulation time in bloodstream enabling detection of enzyme activity for extended periods of time at low doses of these sensors. In the future, more effective "activated" probes are expected to become available with optimized sensitivity to enzymatic activity, spectral characteristics suitable for intraoperative imaging of surgical field, biocompatibility and lack of immunogenicity and toxicity. New in vivo optical imaging methods such as the fluorescence lifetime and photo(opto)acoustic imaging will contribute to early diagnosis of human diseases. The use of sensors for in vivo optical imaging will include more extensive preclinical applications of experimental therapies. At the same time, the ongoing development and improvement of optical signal detectors as well as the availability of biologically inert and highly specific fluorescent probes will further contribute to the introduction of fluorescence imaging into the clinic.
Collapse
Affiliation(s)
- A A Bogdanov
- University of Massachusetts Medical School, Department of Radiology, Laboratory of Molecular Imaging Probes, Worcester, MA 01655, USA. .,A. N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Laboratory of Molecular Imaging, Moscow, 119071, Russia.,Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, Moscow, 119991, Russia
| | - I D Solovyev
- A. N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Laboratory of Molecular Imaging, Moscow, 119071, Russia.,A. N. Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Laboratory of Physical Biochemistry, Moscow, 119071, Russia
| | - A P Savitsky
- A. N. Bach Institute of Biochemistry, Federal Research Center "Fundamentals of Biotechnology", Russian Academy of Sciences, Laboratory of Molecular Imaging, Moscow, 119071, Russia.,A. N. Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Laboratory of Physical Biochemistry, Moscow, 119071, Russia
| |
Collapse
|
2
|
Abdulla MH, Valli-Mohammed MA, Al-Khayal K, Al Shkieh A, Zubaidi A, Ahmad R, Al-Saleh K, Al-Obeed O, McKerrow J. Cathepsin B expression in colorectal cancer in a Middle East population: Potential value as a tumor biomarker for late disease stages. Oncol Rep 2017; 37:3175-3180. [PMID: 28440429 PMCID: PMC5442396 DOI: 10.3892/or.2017.5576] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/02/2017] [Indexed: 02/05/2023] Open
Abstract
Cathepsin B (CTSB), is a cysteine protease belonging to the cathepsin (Clan CA) family. The diagnostic and prognostic significance of increased CTSB in the serum of cancer patients have been evaluated for some tumor types. CTSB serum and protein levels have also been reported previously in colorectal cancer (CRC) with contradictory results. The aim of the present study was to investigate CTSB expression in CRC patients and the association of CTSB expression with various tumor stages in a Middle East population. Serum CTSB levels were evaluated in 70 patients and 20 healthy control subjects using enzyme-linked immunosorbant assay (ELISA) technique. CTSB expression was determined in 100 pairs of CRC tumor and adjacent normal colonic tissue using quantitative PCR for mRNA levels. Detection of CTSB protein expression in tissues was carried out using both immunohistochemistry and western blotting techniques. ELISA analysis showed that in sera obtained from CRC patients, the CTSB concentration was significantly higher in late stage patients with lymph node metastases when compared to early stage patients with values of 2.9 and 0.33 ng/ml, respectively (P=0.001). The majority of tumors studied had detectable CTSB protein expression with significant increased positive staining in tumors cells when compared with matched normal colon subjects (P=0.006). The mRNA expression in early stage CRC compared to late stage CRC was 0.04±0.01 and 0.07±0.02, respectively. Increased mRNA expression was more frequently observed in the advanced cancer stages with lymph node metastases when compared with the control (P=0.002). Mann-Whitney test and paired t-test were used to compare serum CTSB and mRNA levels in early and late tumor stage. A subset of four paired tissue extracts were analyzed by western blotting. The result confirmed a consistent increase in the CTSB protein expression level in tumor tissues compared with that noted in the adjacent normal mucosal cells. These findings indicate that CTSB may be an important prognostic biomarker for late stage CRC and cases with lymph node metastases in the Middle Eastern population. Monitoring serum CTSB in CRC patients may predict and/or diagnose cases with lymph node metastases.
Collapse
Affiliation(s)
- Maha-Hamadien Abdulla
- Colorectal Research Chair, Department of Surgery, King Khalid University Hospital, King Saud University, College of Medicine, Riyadh 11472, Kingdom of Saudi Arabia
| | - Mansoor-Ali Valli-Mohammed
- Colorectal Research Chair, Department of Surgery, King Khalid University Hospital, King Saud University, College of Medicine, Riyadh 11472, Kingdom of Saudi Arabia
| | - Khayal Al-Khayal
- Colorectal Research Chair, Department of Surgery, King Khalid University Hospital, King Saud University, College of Medicine, Riyadh 11472, Kingdom of Saudi Arabia
| | - Abdulmalik Al Shkieh
- Department of Pathology, King Khalid University Hospital, King Saud University, Riyadh 11472, Kingdom of Saudi Arabia
| | - Ahmad Zubaidi
- Colorectal Research Chair, Department of Surgery, King Khalid University Hospital, King Saud University, College of Medicine, Riyadh 11472, Kingdom of Saudi Arabia
| | - Rehan Ahmad
- Colorectal Research Chair, Department of Surgery, King Khalid University Hospital, King Saud University, College of Medicine, Riyadh 11472, Kingdom of Saudi Arabia
| | - Khalid Al-Saleh
- Medical Oncology Unit, Department of Medicine, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia
| | - Omar Al-Obeed
- Colorectal Research Chair, Department of Surgery, King Khalid University Hospital, King Saud University, College of Medicine, Riyadh 11472, Kingdom of Saudi Arabia
| | - James McKerrow
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
3
|
Abdollahi A, Getts LA, Sonoda G, Miller PD, Taguchi T, Godwin AK, Testa JR, Hamilton TC. Genome Scanning Detects Amplification of the Cathepsin B Gene (CtsB) in Transformed Rat Ovarian Surface Epithelial Cells. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155769900600108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | | | | | | | | | | | - Joseph R. Testa
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, and Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania
| | - Thomas C. Hamilton
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, and Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania
| |
Collapse
|
4
|
Blau R, Krivitsky A, Epshtein Y, Satchi-Fainaro R. Are nanotheranostics and nanodiagnostics-guided drug delivery stepping stones towards precision medicine? Drug Resist Updat 2016; 27:39-58. [PMID: 27449597 DOI: 10.1016/j.drup.2016.06.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/01/2016] [Accepted: 06/09/2016] [Indexed: 12/12/2022]
Abstract
The progress in medical research has led to the understanding that cancer is a large group of heterogeneous diseases, with high variability between and within individuals. This variability sprouted the ambitious goal to improve therapeutic outcomes, while minimizing drug adverse effects through stratification of patients by the differences in their disease markers, in a personalized manner, as opposed to the strategy of "one therapy fits all". Nanotheranostics, composed of nanoparticles (NPs) carrying therapeutic and/or diagnostics probes, have the potential to revolutionize personalized medicine. There are different modalities to combine these two distinct fields into one system for a synergistic outcome. The addition of a nanocarrier to a theranostic system holds great promise. Nanocarriers possess high surface area, enabling sophisticated functionalization with imaging agents, thus gaining enhanced diagnostic ability in real-time. Yet, most of the FDA-approved theranostic approaches are based on small molecules. The theranostic approaches that are reviewed herein are paving the road towards personalized medicine through all stages of patient care: starting from screening and diagnostics, proceeding to treatment and ending with treatment follow-up. Our current review provides a broad background and highlights new insights for the rational design of theranostic nanosystems for desired therapeutic niches, while summoning the hurdles on their way to become first-line diagnostics and therapeutics for cancer patients.
Collapse
Affiliation(s)
- Rachel Blau
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yana Epshtein
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
5
|
Xu ZZ, Xiu P, Lv JW, Wang FH, Dong XF, Liu F, Li T, Li J. Integrin αvβ3 is required for cathepsin B-induced hepatocellular carcinoma progression. Mol Med Rep 2014; 11:3499-504. [PMID: 25572981 DOI: 10.3892/mmr.2014.3140] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 08/14/2014] [Indexed: 11/05/2022] Open
Abstract
The cysteine protease cathepsin B (Cat B) is important in the progression of tumor cells, however, the function and molecular mechanisms of Cat B in hepatocellular carcinoma (HCC) remain to be elucidated. Our previous study demonstrated that integrin αvβ3 regulated the biological behavior of HCC. The present study demonstrated that Cat B was also important in cell proliferation and apoptosis in HCC. Notably, Cat B was observed to activate the phosphoinositide 3‑kinase (PI3K)/Akt signaling pathway to promote HCC proliferation. Furthermore, inhibition of integrin αvβ3 significantly prevented Cat B‑induced activation of PI3K/Akt and the progression of HCC. Thus, the results of the present study suggested the presence of a Cat B/integrin αvβ3/PI3K/Akt axis in the regulation of the progression of HCC.
Collapse
Affiliation(s)
- Zong-Zhen Xu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Peng Xiu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Ju-Wei Lv
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Fu-Hai Wang
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Xiao-Feng Dong
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Feng Liu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Tao Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Jie Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
6
|
Chen TP, Yang SF, Lin CW, Lee HL, Tsai CM, Weng CJ. A4383C and C76G SNP in Cathepsin B is respectively associated with the high risk and tumor size of hepatocarcinoma. Tumour Biol 2014; 35:11193-8. [PMID: 25106406 DOI: 10.1007/s13277-014-2004-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/22/2014] [Indexed: 12/16/2022] Open
Abstract
Single nucleotide polymorphism (SNP) in some genes is a candidate for having or developing a cancer. Cathepsin B (CTSB) is considered to be the biomarker of cancers. The study aimed to evaluate the impacts of three SNPs in CTSB gene on the risk and progress of hepatocellular carcinoma (HCC). The SNPs of CTSB C76G (rs12338), CTSB A4383C (rs13332), and CTSB A8422G (rs8898) from 135 patients with HCC and 520 control participants in Taiwan were determined by real-time PCR. Through analyzing by statistics, we found that the polymorphism of rs13332 was significantly associated to the risk of HCC cancer; a significantly high frequent tumor size development was observed in HCC patients carrying rs12338 polymorphic genotype than those carrying ancestral genotype. The SNPs of rs12338, rs13332, and rs8898 were irrelevant to the frequencies of HCC clinical status and the levels of HCC clinicopathological markers. In conclusions, CTSB A4383C SNP is observed modestly more often in patients who developed HCC than in healthy controls and might be associated with the risk of HCC. The association between CTSB C76G SNP and greater tumor size may warrant further study in regards to the biology of HCC.
Collapse
Affiliation(s)
- Tsung-Po Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
7
|
Bogdanov AA, Mazzanti ML. Fluorescent macromolecular sensors of enzymatic activity for in vivo imaging. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 113:349-87. [PMID: 23244795 DOI: 10.1016/b978-0-12-386932-6.00009-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Macromolecular imaging probes (or sensors) of enzymatic activity have a unique place in the armamentarium of modern optical imaging techniques. Such probes were initially developed by attaching optically "silent" fluorophores via enzyme-sensitive linkers to large copolymers of biocompatible poly(ethylene glycol) and poly(amino acids). In diseased tissue, where the concentration of enzymes is high, the fluorophores are freed from the macromolecular carrier and regain their initial ability to fluoresce, thus allowing in vivo optical localization of the diseased tissue. This chapter describes the design and application of these probes and their alternatives in various areas of experimental medicine and gives an overview of currently available techniques that allow imaging of animals using visible and near-infrared light.
Collapse
Affiliation(s)
- Alexei A Bogdanov
- Laboratory of Molecular Imaging Probes, Department of Radiology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
8
|
Braun K, Beining M, Wiessler M, Lammers T, Pipkorn R, Hennrich U, Nokihara K, Semmler W, Debus J, Waldeck W. BioShuttle mobility in living cells studied with high-resolution FCS & CLSM methodologies. Int J Med Sci 2012; 9:339-52. [PMID: 22811608 PMCID: PMC3399214 DOI: 10.7150/ijms.4414] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 06/18/2012] [Indexed: 01/04/2023] Open
Abstract
With the increase in molecular diagnostics and patient-specific therapeutic approaches, the delivery and targeting of imaging molecules and pharmacologically active agents gain increasing importance. The ideal delivery system does not exist yet. The realization of two features is indispensable: first, a locally high concentration of target-specific diagnostic and therapeutic molecules; second, the broad development of effective and safe carrier systems. Here we characterize the transport properties of the peptide-based BioShuttle transporter using FFM and CLSM methods. The modular design of BioShuttle-based formulations results in a multi-faceted field of applications, also as a theranostic tool.
Collapse
Affiliation(s)
- Klaus Braun
- Dept. of Imaging and Radiooncology, German Cancer Research Center, Im Neuenheimer Feld 280, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Xie BW, Mol IM, Keereweer S, van Beek ER, Que I, Snoeks TJA, Chan A, Kaijzel EL, Löwik CWGM. Dual-wavelength imaging of tumor progression by activatable and targeting near-infrared fluorescent probes in a bioluminescent breast cancer model. PLoS One 2012; 7:e31875. [PMID: 22348134 PMCID: PMC3278453 DOI: 10.1371/journal.pone.0031875] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 01/13/2012] [Indexed: 12/29/2022] Open
Abstract
Bioluminescence imaging (BLI) has shown its appeal as a sensitive technique for in vivo whole body optical imaging. However, the development of injectable tumor-specific near-infrared fluorescent (NIRF) probes makes fluorescence imaging (FLI) a promising alternative to BLI in situations where BLI cannot be used or is unwanted (e.g., spontaneous transgenic tumor models, or syngeneic mice to study immune effects). In this study, we addressed the questions whether it is possible to detect tumor progression using FLI with appropriate sensitivity and how FLI correlates with BLI measurements. In addition, we explored the possibility to simultaneously detect multiple tumor characteristics by dual-wavelength FLI (∼700 and ∼800 nm) in combination with spectral unmixing. Using a luciferase-expressing 4T1-luc2 mouse breast cancer model and combinations of activatable and targeting NIRF probes, we showed that the activatable NIRF probes (ProSense680 and MMPSense680) and the targeting NIRF probes (IRDye 800CW 2-DG and IRDye 800CW EGF) were either activated by or bound to 4T1-luc2 cells. In vivo, we implanted 4T1-luc2 cells orthotopically in nude mice and were able to follow tumor progression longitudinally both by BLI and dual-wavelength FLI. We were able to reveal different probe signals within the tumor, which co-localized with immuno-staining. Moreover, we observed a linear correlation between the internal BLI signals and the FLI signals obtained from the NIRF probes. Finally, we could detect pulmonary metastases both by BLI and FLI and confirmed their presence histologically. Taken together, these data suggest that dual-wavelength FLI is a feasible approach to simultaneously detect different features of one tumor and to follow tumor progression with appropriate specificity and sensitivity. This study may open up new perspectives for the detection of tumors and metastases in various experimental models and could also have clinical applications, such as image-guided surgery.
Collapse
Affiliation(s)
- Bang-Wen Xie
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Moin K, Sameni M, Victor BC, Rothberg JM, Mattingly RR, Sloane BF. 3D/4D functional imaging of tumor-associated proteolysis: impact of microenvironment. Methods Enzymol 2012; 506:175-94. [PMID: 22341225 PMCID: PMC3845223 DOI: 10.1016/b978-0-12-391856-7.00034-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proteases play causal roles in many aspects of the aggressive phenotype of tumors, yet many of the implicated proteases originate from tumor-associated cells or from responses of tumor cells to interactions with other cells. Therefore, to obtain a comprehensive view of tumor proteases, we need to be able to assess proteolysis in tumors that are interacting with their microenvironment. As this is difficult to do in vivo, we have developed functional live-cell optical imaging assays and 3D and 4D (i.e., 3D over time) coculture models. We present here a description of the probes used to measure proteolysis and protease activities, the methods used for imaging and analysis of proteolysis and the 3D and 4D models used in our laboratory. Of course, all assays have limitations; however, we suggest that the techniques discussed here will, with attention to their limitations, be useful as a screen for drugs to target the invasive phenotype of tumors.
Collapse
Affiliation(s)
- Kamiar Moin
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | | | | | |
Collapse
|
11
|
Cystatins – Extra- and intracellular cysteine protease inhibitors: High-level secretion and uptake of cystatin C in human neuroblastoma cells. Biochimie 2010; 92:1625-34. [DOI: 10.1016/j.biochi.2010.08.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Accepted: 08/09/2010] [Indexed: 02/01/2023]
|
12
|
Chan AT, Baba Y, Shima K, Nosho K, Chung DC, Hung KE, Mahmood U, Madden K, Poss K, Ranieri A, Shue D, Kucherlapati R, Fuchs CS, Ogino S. Cathepsin B expression and survival in colon cancer: implications for molecular detection of neoplasia. Cancer Epidemiol Biomarkers Prev 2010; 19:2777-85. [PMID: 20833970 DOI: 10.1158/1055-9965.epi-10-0529] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND AIMS Proteases play a critical role in tumorigenesis and are upregulated in colorectal cancer and neoplastic polyps. In animal models, cathepsin B (CTSB)-activatable imaging agents show high enzyme activity within intestinal tumors. METHODS We conducted a prospective cohort study of 558 men and women with colon cancer with tumors that were accessible for immunohistochemical assessment. We used Cox proportional hazards models, stratified by stage, to compute colon cancer-specific and overall mortality according to tumoral expression of CTSB. RESULTS Among 558 participants, 457 (82%) had tumors that expressed CTSB (CTSB positive) and 101 (18%) had tumors that did not express CTSB (CTSB negative). CTSB expression was not associated with disease stage (P = 0.19). After a median follow-up of 11.6 years, there were 254 total and 155 colon cancer-specific deaths. Compared with participants with CTSB-negative tumors, participants with CTSB-positive tumors experienced a multivariate hazard ratio for colon cancer-specific mortality of 1.99 (95% confidence interval, 1.19-3.34) and overall mortality of 1.71 (95% confidence interval, 1.16-2.50). CTSB expression was independently associated with KRAS (P = 0.01) and BRAF mutation (P = 0.04), but not microsatellite instability status, CpG island methylator phenotype status, PIK3CA mutation, LINE-1 methylation, TP53 expression, or PTGS2 (cyclooxygenase-2) expression. Among 123 individuals with adenomas, 91% expressed CTSB. CONCLUSIONS As assessed by immunohistochemistry, CTSB is expressed in the vast majority of colon cancers, independent of stage, and is significantly associated with higher risk of colon cancer-specific and overall mortality. IMPACT These results support the potential of CTSB a target for image detection of neoplastic lesions in humans.
Collapse
Affiliation(s)
- Andrew T Chan
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Shubin AV, Demidyuk IV, Kurinov AM, Demkin VV, Vinogradova TV, Zinovyeva MV, Sass AV, Zborovskaya IB, Kostrov SV. Cathepsin D messenger RNA is downregulated in human lung cancer. Biomarkers 2010; 15:608-13. [PMID: 20722505 DOI: 10.3109/1354750x.2010.504310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Lysosomal proteases cathepsins B and D (CB and CD) play a significant part in cancer progression. For many oncological diseases protein expression levels of CB and CD have been investigated and correlations with tumour characteristics revealed. Meanwhile, there is very little information concerning mRNA expression level. METHODS In the present work, data about mRNA levels of CB and CD in human lung cancer was obtained using reverse transcription followed by real-time polymerase chain reaction. RESULTS For the first time CD and CB mRNA in human lung cancer tumours was quantified. It was shown that CB and CD mRNA levels do not correlate with any tumour characteristics. However, in most analysed tumours, expression of CD mRNA was downregulated compared with adjacent normal tissue (p <0.0003). CONCLUSIONS The data obtained indicate CD mRNA as a potential lung cancer marker.
Collapse
Affiliation(s)
- Andrey V Shubin
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Green AH, Norris JR, Wang J, Xie Z, Zhang HF, La Riviere PJ. In vitro testing of a protease-sensitive contrast agent for optoacoustic imaging. JOURNAL OF BIOMEDICAL OPTICS 2010; 15:021315. [PMID: 20459237 DOI: 10.1117/1.3365935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
We have designed a protease-sensitive imaging probe for optoacoustic imaging whose absorption spectrum changes upon cleavage by a protease of interest. The probe comprises an active site, a derivative of chlorophyll or natural photosynthetic bacteriochlorophyll that absorbs in the near infrared, conjugated to a peptide backbone specific to the protease being imaged. The uncleaved molecules tend to aggregate in dimers and trimers, causing a change in the absorption spectrum relative to that of the monomer. Upon cleavage, the probe molecules deaggregate, giving rise to a spectrum characteristic of monomers. We show using photospectrometry that the two forms of the probe have markedly different absorption spectra, which could allow for in vivo optoacoustic identification using a multiwavelength imaging strategy. Optoacoustic measurements using a narrow-band dye laser find spectral peaks in the two forms of the probe at the expected location. The optoacoustic signal from the uncleaved probe is found to be considerably weaker than that of the cleaved probe, perhaps due to poor optical-acoustic coupling in the aggregated molecules. However, ultimately, it is detection of the cleaved probe that is of the greatest import, since it reports on the protease activity of interest.
Collapse
Affiliation(s)
- Anthony H Green
- The University of Chicago, Department of Chemistry, 5735 S. Ellis Avenue, Chicago, Illinois 60637, USA
| | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Ranger A, McDonald W, Moore E, Delmaestro R. The invasiveness of five medulloblastoma cell lines in collagen gels. J Neurooncol 2009; 96:181-9. [PMID: 19847623 DOI: 10.1007/s11060-009-9962-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Accepted: 07/06/2009] [Indexed: 10/20/2022]
Abstract
Local recurrence continues to limit survival in medulloblastoma patients, largely related to the persistence of invasive cells at the site of tumour resection and leptomeningeal dissemination. Given the relative dearth of understanding of causative mechanisms behind the invasiveness of medulloblastomas, and a general lack of validated in vitro models with which to study them, our objectives were (1) to obtain quantitative data on the invasiveness of five distinct medulloblastoma cell lines within a 3-dimensional in vitro collagen-based model; and (2) to characterize some of the mechanisms behind invasion, specifically striving to identify proteolytic processes that occur as medulloblastoma cells disrupt and thereby invade the normal tissue surrounding them, and specific inhibitors of these proteolytic enzymes. Five different medulloblastoma cell lines (UW228-1, 2 and 3; Daoy, and Madsen) were implanted onto a 3-dimensional, type I collagen gel assay to assess tumour invasion distance and mean doubling time over 5 days. Proteolytic activity was assessed against collagen types I and IV by measuring the degradation of 3H-collagen I and IV to products soluble in 100% w/v trichloroacetic acid; and general (neutral) proteolytic activity evaluated by measuring the degradation of 3H-albumin. In other experiments, cells were pre-exposed to a variety of protease inhibitors, including inhibitors of metalloproteinases and cysteine, serine and aspartic proteases, and then plated to identify any inhibition of invasion. Inter-group differences in mean invasion distance were assessed by means of Student's t-tests for non-paired subjects, with P < 0.05 set as the threshold for statistical significance. For the inhibitor studies, an inhibition index, called the inhibitory concentration 50, IC-50, was calculated by performing a regression analysis for each inhibitor tested over a range of concentrations, for each cell line. Within hours of implantation, individual cells readily detached from the surface of the cell aggregates and invaded the collagen matrix, to distances of up to 1,200 mum and at rates of up to 300-mum per day; the UW228-1 cell line clearly was less invasive than the other four cell lines. Proteolytic activity was identified against collagen type I, but not against collagen type IV or albumin; but there was no apparent correlation between invasion distance and either cell doubling time or the amount of collagen type I proteolytic activity. Both metalloproteinase inhibitors suppressed tumour invasion, as did one of two cysteine protease inhibitors; but there was no tumour suppression with either serine or aspartic protease inhibition. MMP-1 and 2, and TIMP-1 and 2 all were detectable by Western blot analysis. Medulloblastoma cell invasiveness within the 3-dimensional model used here appears to depend upon a combination of metalloproteinase and cysteine protease activity, a finding that may suggest areas for potential future clinical investigation and therapy.
Collapse
Affiliation(s)
- Adrianna Ranger
- Brain Research Laboratories, Experimental Research Unit, Division of Neurosurgery, London Health Sciences Centre, University of Western Ontario, London, ON N6A4G5, Canada.
| | | | | | | |
Collapse
|
17
|
Ekström U, Wallin H, Lorenzo J, Holmqvist B, Abrahamson M, Avilés FX. Internalization of cystatin C in human cell lines. FEBS J 2008; 275:4571-82. [PMID: 18699780 PMCID: PMC7163943 DOI: 10.1111/j.1742-4658.2008.06600.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Altered protease activity is considered important for tumour invasion and metastasis, processes in which the cysteine proteases cathepsin B and L are involved. Their natural inhibitor cystatin C is a secreted protein, suggesting that it functions to control extracellular protease activity. Because cystatins added to cell cultures can inhibit polio, herpes simplex and coronavirus replication, which are intracellular processes, the internalization and intracellular regulation of cysteine proteases by cystatin C should be considered. The extension, mechanism and biological importance of this hypothetical process are unknown. We investigated whether internalization of cystatin C occurs in a set of human cell lines. Demonstrated by flow cytometry and confocal microscopy, A‐431, MCF‐7, MDA‐MB‐453, MDA‐MB‐468 and Capan‐1 cells internalized fluorophore‐conjugated cystatin C when exposed to physiological concentrations (1 μm). During cystatin C incubation, intracellular cystatin C increased after 5 min and accumulated for at least 6 h, reaching four to six times the baseline level. Western blotting showed that the internalized inhibitor was not degraded. It was functionally intact and extracts of cells exposed to cystatin C showed a higher capacity to inhibit papain and cathepsin B than control cells (decrease in enzyme activity of 34% and 37%, respectively). The uptake of labelled cystatin C was inhibited by unlabelled inhibitor, suggesting a specific pathway for the internalization. We conclude that the cysteine protease inhibitor cystatin C is internalized in significant quantities in various cancer cell lines. This is a potentially important physiological phenomenon not previously described for this group of inhibitors.
Collapse
Affiliation(s)
- Ulf Ekström
- Department of Laboratory Medicine, Lund University, Sweden
| | | | | | | | | | | |
Collapse
|
18
|
Pandey RK, James N, Chen Y, Dobhal MP. Cyanine Dye-Based Compounds for Tumor Imaging With and Without Photodynamic Therapy. TOPICS IN HETEROCYCLIC CHEMISTRY 2008. [DOI: 10.1007/7081_2008_113] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
19
|
Ai L, Kim WJ, Kim TY, Fields CR, Massoll NA, Robertson KD, Brown KD. Epigenetic silencing of the tumor suppressor cystatin M occurs during breast cancer progression. Cancer Res 2007; 66:7899-909. [PMID: 16912163 DOI: 10.1158/0008-5472.can-06-0576] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cystatin M is a secreted inhibitor of lysosomal cysteine proteases. Several lines of evidence indicate that cystatin M is a tumor suppressor important in breast malignancy; however, the mechanism(s) that leads to inactivation of cystatin M during cancer progression is unknown. Inspection of the human cystatin M locus uncovered a large and dense CpG island within the 5' region of this gene (termed CST6). Analysis of cultured human breast tumor lines indicated that cystatin M expression is either undetectable or in low abundance in several lines; however, enhanced gene expression was measured in cells cultured on the DNA demethylating agent 5-aza-2'-deoxycytidine (5-aza-dC). Increased cystatin M expression does not correlate with a cytotoxic response to 5-aza-dC; rather, various molecular approaches indicated that the CST6 gene was aberrantly methylated in these tumor lines as well as in primary breast tumors. Moreover, 60% (12 of 20) of primary tumors analyzed displayed CST6 hypermethylation, indicating that this aberrant characteristic is common in breast malignancies. Finally, preinvasive and invasive breast tumor cells were microdissected from nine archival breast cancer specimens. Of the five tumors displaying CST6 gene methylation, four tumors displayed methylation in both ductal carcinoma in situ and invasive breast carcinoma lesions and reduced expression of cystatin M in these tumors was confirmed by immunohistochemistry. In summary, this study establishes that the tumor suppressor cystatin M is a novel target for epigenetic silencing during mammary tumorigenesis and that this aberrant event can occur before development of invasive breast cancer.
Collapse
Affiliation(s)
- Lingbao Ai
- Department of Biochemistry and Molecular Biology and University of Florida Shands Cancer Center Program in Cancer Genetics, Epigenetics, and Tumor Virology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
|
21
|
Kovar JL, Simpson MA, Schutz-Geschwender A, Olive DM. A systematic approach to the development of fluorescent contrast agents for optical imaging of mouse cancer models. Anal Biochem 2007; 367:1-12. [PMID: 17521598 DOI: 10.1016/j.ab.2007.04.011] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 03/26/2007] [Accepted: 04/06/2007] [Indexed: 11/25/2022]
|
22
|
Abstract
A new approach to selective photodynamic therapy (PDT) was developed by designing chlorin e6 (Ce6)-containing macromolecules, which are sensitive to tumor-associated proteases. The agents are nontoxic in their native state but become fluorescent and produce singlet oxygen on protease conversion. Coupled with optimized delivery systems, we show that (a) the agents efficiently accumulate in tumors due to the enhanced permeability and retention effect, (b) the agents are locally activated by proteases, (c) local drug concentrations can be measured by quantitative fluorescence tomography, and (d) light-treated tumors show reduced growth. A single low dose of PDT (0.125 mg Ce6 equivalent/kg) was sufficient to suppress tumor growth by >50%. Activatable singlet oxygen generation agents provide increased efficacy with reduced toxicity, and it could become a powerful PDT.
Collapse
Affiliation(s)
- Yongdoo Choi
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | |
Collapse
|
23
|
Mugherli L, Burchak ON, Chatelain F, Balakirev MY. Fluorogenic ester substrates to assess proteolytic activity. Bioorg Med Chem Lett 2006; 16:4488-91. [PMID: 16806926 DOI: 10.1016/j.bmcl.2006.06.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Revised: 06/09/2006] [Accepted: 06/10/2006] [Indexed: 10/24/2022]
Abstract
The synthesis of a new type of fluorogenic ester substrates is described. Prepared from fluorescein in three steps with common commercially available precursors, they all generate bright green fluorescence upon proteolysis. Their particular structure allows the same substrate be used to report enzymatic activity of various proteases from serine and cysteine superfamilies. The substrate cleavage is sensitive to specific protease inhibitors providing a tool for inhibitor screening.
Collapse
Affiliation(s)
- Laurent Mugherli
- Laboratoire Biopuces, Département Réponse et Dynamique Cellulaires, Commissariat à l'Energie Atomique, Grenoble, France
| | | | | | | |
Collapse
|
24
|
Pipkorn R, Waldeck W, Spring H, Jenne JW, Braun K. Delivery of substances and their target-specific topical activation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2006; 1758:606-10. [PMID: 16730647 DOI: 10.1016/j.bbamem.2006.03.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2005] [Revised: 03/17/2006] [Accepted: 03/23/2006] [Indexed: 10/24/2022]
Abstract
Goal in pharmaceutical research is achievement of necessary drug concentrations in the target organ, effective treatment with safe delivery of genetic agents, while sparing normal tissue and minimizing side effects. A new "BioShuttle"-delivery system harbouring a cathepsin B cutting site, a nuclear address sequence and a functional peptide was developed and tumor cells were treated. Transport and subcellular activation were determined by confocal laser scanning microscopy permitting the conclusion: BioShuttle-conjugates prove as efficient tools for genetic interventions by selective and topical activation of therapeutic peptide precursors by enzymatic cleavage. As shown here for glioma cells and the cathepsin B cleavable site, living cells can be treated with high specificity and selectivity for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Rüdiger Pipkorn
- Peptide Synthesis Facility, German Cancer Research Center, Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
25
|
Keppler D. Towards novel anti-cancer strategies based on cystatin function. Cancer Lett 2006; 235:159-76. [PMID: 15893421 DOI: 10.1016/j.canlet.2005.04.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2005] [Accepted: 04/01/2005] [Indexed: 02/04/2023]
Abstract
Cystatins have recently emerged as important players in a multitude of physiological and patho-physiological settings that range from cell survival and proliferation, to differentiation, cell signaling and immunomodulation. This group of cysteine protease inhibitors forms a large super-family of proteins composed of one, two, three, and, in some species, more than three cystatin domains. Over the last 20 years or so, members of the cystatin super-family have been primarily explored with respect to their capacity to inhibit intracellular cysteine proteases. Yet, this classical mode of action does not fully explain their remarkably diverse biological functions. Due to the space limitations, the author will discuss here the most recent findings that suggest that some of the single-domain, cytoplasmic and cell-secreted cystatins may play important roles in the promotion or suppression of tumor growth, invasion and metastasis. Based on the present understanding of cystatin function, novel avenues for anti-cancer strategies are proposed.
Collapse
Affiliation(s)
- Daniel Keppler
- Department of Cellular Biology and Anatomy and Feist-Weiller Cancer Center, School of Medicine, Louisiana State University Health Sciences Center in Shreveport, 1501 Kings Highway, P.O. Box 33932, Shreveport, LA 71130, USA.
| |
Collapse
|
26
|
Tardy C, Codogno P, Autefage H, Levade T, Andrieu-Abadie N. Lysosomes and lysosomal proteins in cancer cell death (new players of an old struggle). Biochim Biophys Acta Rev Cancer 2005; 1765:101-25. [PMID: 16412578 DOI: 10.1016/j.bbcan.2005.11.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2005] [Revised: 11/21/2005] [Accepted: 11/28/2005] [Indexed: 12/19/2022]
Abstract
Death of cancer cells influences tumor development and progression, as well as the response to anticancer therapies. This can occur through different cell death programmes which have recently been shown to implicate components of the acidic organelles, lysosomes. The role of lysosomes and lysosomal enzymes, including cathepsins and some lipid hydrolases, in programmed cell death associated with apoptotic or autophagic phenotypes is presented, as evidenced from observations on cultured cells and living animals. The possible molecular mechanisms that underlie the action of lysosomes during cell death are also described. Finally, the contribution of lysosomal proteins and lysosomes to tumor initiation and progression is discussed. Elucidation of this role and the underlying mechanisms will shed a new light on these 'old' organelles and hopefully pave the way for the development of novel anticancer strategies.
Collapse
Affiliation(s)
- Claudine Tardy
- INSERM U466, Laboratoire de Biochimie, Institut Louis Bugnard, Centre Hospitalier Universitaire de Rangueil, BP 84225, 31432 Toulouse, France
| | | | | | | | | |
Collapse
|
27
|
Abstract
Cystatins form a large superfamily of proteins with diverse biologic activities. All members of the cystatin superfamily share the presence of one, two or three cystatin domains. Cystatins were initially believed to act mainly as inhibitors of lysosomal cysteine proteases. In recent years, however, there has been increased awareness of additional or alternate biologic functions for these proteins. In this review, the authors will discuss the most recent findings and hypotheses that suggest that some members of the cystatin superfamily may play important roles during tumor progression. Special emphasis is given to their potential role as novel anti-angiogenic agents.
Collapse
Affiliation(s)
- Daniel Keppler
- Louisiana State University Health Sciences Center, Department of Cellular Biology & Anatomy and Feist-Weiller Cancer Center, School of Medicine, Shreveport, LA 71130, USA.
| | | |
Collapse
|
28
|
Sounni NE, Noel A. Membrane type-matrix metalloproteinases and tumor progression. Biochimie 2005; 87:329-42. [PMID: 15781320 DOI: 10.1016/j.biochi.2004.07.012] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2004] [Accepted: 07/16/2004] [Indexed: 01/30/2023]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc endopeptidases that process growth factors, growth factor binding proteins, cell surface proteins, degrade extracellular matrix (ECM) components and thereby play a central role in tissue remodeling and tumor progression. Membrane-type matrix metalloproteinases (MT-MMPs) are a recently discovered subgroup of intrinsic plasma membrane proteins. Their functions have been extended from pericellular proteolysis and control of cell migration to cell signaling, control of cell proliferation and regulation of multiple stages of tumor progression including growth and angiogenesis. This review sheds light on the new functions of MT-MMPs and their inhibitors in tumor development and angiogenesis, and presents recent investigations that document their influence on various cell functions.
Collapse
Affiliation(s)
- N E Sounni
- Laboratory of Tumor and Development Biology, University of Liège, Sart-Tilman B23, B4000 Liège, Belgium
| | | |
Collapse
|
29
|
Büth H, Wolters B, Hartwig B, Meier-Bornheim R, Veith H, Hansen M, Sommerhoff CP, Schaschke N, Machleidt W, Fusenig NE, Boukamp P, Brix K. HaCaT keratinocytes secrete lysosomal cysteine proteinases during migration. Eur J Cell Biol 2005; 83:781-95. [PMID: 15679122 DOI: 10.1078/0171-9335-00428] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cathepsin B, a lysosomal cysteine proteinase, was detected within vesicles of cellular protrusions forming cell-cell contact sites between keratinocytes of the stratum spinosum of human skin. This observation suggested the possibility that secretion of the protease into the pericellular spaces could be involved in the dissociation of cell-cell contacts to enable intraepidermal keratinocyte migration. To determine whether cathepsin B is indeed secreted from migrating keratinocytes, we first used subconfluent HaCaT cells as a culture model to study spontaneous keratinocyte migration. A cathepsin B-specific fluorescent affinity label proved the association of mature cathepsin B with the surfaces of HaCaT cells at the leading edges of growing cells. Second, we used scratch-wounds of confluent HaCaT monolayers as a model of induced keratinocyte migration. Cathepsin B was detected within lysosomes, i.e. vesicles within the perinuclear region of non-wounded cells. Expression of cathepsin B was up-regulated and cathepsin B-positive vesicles showed a redistribution from perinuclear to peripheral regions of keratinocytes at the wound margins within 4 h after wounding. Enzyme cytochemistry further showed that cell surface-associated cathepsin B was proteolytically active at the leading fronts of migrating keratinocytes. In addition, increased amounts of mature forms of cathepsin B were detected within the conditioned media of HaCaT cells during the first 4 h after scratch-wounding. In contrast, and as a control, the activity of the cytosolic enzyme lactate dehydrogenase was not significantly higher in media of wounded cells as compared with non-wounded controls, arguing for a specific induction of cathepsin B secretion upon wounding and migration of the cells. This was further substantiated by applying various cathepsin B-specific inhibitors after wounding. These experiments showed that the migration ability of keratinocytes was reduced due to the blockage of functional cathepsin B. Thus, our results strongly suggest that cell surface-associated cathepsin B is a protease that contributes to the remodelling of the extracellular matrix and thereby promotes keratinocyte migration during wound healing.
Collapse
Affiliation(s)
- Heiko Büth
- School of Engineering and Science, International University Bremen, Bremen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Im E, Venkatakrishnan A, Kazlauskas A. Cathepsin B regulates the intrinsic angiogenic threshold of endothelial cells. Mol Biol Cell 2005; 16:3488-500. [PMID: 15901832 PMCID: PMC1182291 DOI: 10.1091/mbc.e04-11-1029] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The lysosomal protease cathepsin B has been implicated in a variety of pathologies including pancreatitis, tumor angiogenesis, and neuronal diseases. We used a tube formation assay to investigate the role of cathepsin B in angiogenesis. When cultured between two layers of collagen I, primary endothelial cells formed tubes in response to exogenously added VEGF. Overexpressing cathepsin B reduced the VEGF-dependent tube response, whereas pharmacologically or molecularly suppressing cathepsin B eliminated the dependence on exogenous VEGF. However, tube formation still required VEGF receptor activity, which suggested that endothelial cells generated VEGF. Indeed, VEGF mRNA and protein was detectable in cells treated with cathepsin B inhibitor, which correlated with a rise in the level of HIF-1alpha. In addition to boosting the level of proangiogenic factors, blocking cathepsin B activity reduced the amount of the antiangiogenic protein endostatin. Thus endothelial cells have the intrinsic capacity to generate pro- and antiangiogenic agents. These observations complement and expand our appreciation of how endothelial cell-derived proteases regulate angiogenesis.
Collapse
Affiliation(s)
- Eunok Im
- Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|
31
|
Freitas ZFO, Rodrigues EG, Oliveira V, Carmona AK, Travassos LR. Melanoma heterogeneity: differential, invasive, metastatic properties and profiles of cathepsin B, D and L activities in subclones of the B16F10-NEX2 cell line. Melanoma Res 2005; 14:333-44. [PMID: 15457088 DOI: 10.1097/00008390-200410000-00002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Tumour cell lines and in vivo growing tumours are heterogeneous, comprising different cell clones. To understand why some cells primarily invade a tissue, while others are more apt to metastasize, several clones from the established B16F10-Nex2 cell line were isolated and 10 viable cells of each clone were injected intravenously into C57Bl/6 and Balb/c mice. Two cell clones (Nex2B and Nex2D) showed contrasting metastatic abilities. Clone 2D rather than clone 2B colonized the lungs of both mice after intravenous injection. Surprisingly, clone 2B grew more rapidly than 2D after subcutaneous implantation, significantly reducing the survival of injected mice. Clearly, dissociation between subcutaneous growth and metastatic ability was observed in clones from the same tumour cell lineage. Clone Nex2B continuously released proteolytic activity, including cathepsin B, and showed a greater capacity to invade Matrigel than clone Nex2D. Clone Nex2D accumulated cathepsins B, D and L intracellularly and released a moderate proteolytic activity in vitro that was inhibited with the time of incubation. E-64-treated Nex2B cells injected subcutaneously showed a significant delay in tumour development and increased survival of challenged animals. A similar result was obtained on treatment of clone 2B with chagasin, a cysteine proteinase inhibitor from Trypanosoma cruzi, even at 2 microM. Clone Nex2D was less sensitive to pretreatment with inhibitors of cysteine proteases for tumour development in vivo. Our results suggest that, in a tumour cell population, cells dissociate into metastatic and non-metastatic subtypes, and that release or accumulation of cathepsins can be a differential trait of these cells.
Collapse
Affiliation(s)
- Zenilda F O Freitas
- Experimental Oncology Unit (UNONEX), Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | | | | | | | | |
Collapse
|
32
|
|
33
|
Graber S, Maiti S, Halpain S. Cathepsin B-like proteolysis and MARCKS degradation in sub-lethal NMDA-induced collapse of dendritic spines. Neuropharmacology 2005; 47:706-13. [PMID: 15458842 DOI: 10.1016/j.neuropharm.2004.08.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2004] [Revised: 07/29/2004] [Accepted: 08/02/2004] [Indexed: 11/19/2022]
Abstract
Sub-lethal excitotoxic injury to dendrites can elicit loss or shrinkage of dendritic spines. Here, we used a cell culture model of sub-lethal NMDA-induced injury to investigate a role for proteolysis in spine collapse. Transient incubation with NMDA-induced spine collapse and spine F-actin loss within 10 min, an effect not mimicked by the actin assembly inhibitor latrunculin A. NMDA-induced spine collapse was significantly attenuated by preincubation with broad-spectrum cysteine protease inhibitors. Results obtained using several class-specific protease inhibitors suggested that this protective effect was due to specific blockade of cathepsin B/L type protease activity, since selective inhibitors of only these proteases significantly attenuated spine loss. Cathepsin B-like immunoreactivity was observed at synaptic sites, but lysosomes were not. Immunoblot analysis showed that MARCKS (myristoylated-alanine-rich C-kinase substrate), a known substrate of cathepsin B, was specifically degraded in response to intense NMDA receptor stimulation. This effect was blocked by preincubation with a cathepsin B-selective inhibitor. Together these data suggest a model in which NMDA-induced spine collapse involves cathepsin B-like proteolysis of MARCKS, and possibly other proteins that regulate the actin-based cytoskeleton.
Collapse
Affiliation(s)
- S Graber
- Department of Cell Biology and Institute for Childhood and Neurological Diseases, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
34
|
Caserman S, Lah TT. Comparison of expression of cathepsins B and L and MMP2 in endothelial cells and in capillary sprouting in collagen gel. Int J Biol Markers 2005; 19:120-9. [PMID: 15255544 DOI: 10.1177/172460080401900206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The lysosomal cysteine proteinases cathepsins B and L are known to play an important role in the invasive growth of tumor cells, but their association with angiogenesis has been less well studied. The aim of this study was to determine the possible role of endothelial cell-associated cathepsins B and L in induced capillary growth in the aorta ring model of angiogenesis. Specific inhibitors of cysteine proteinases did not inhibit capillary growth in aorta ring culture and only slightly inhibited the degradation of surrounding collagen. In contrast, strong inhibition of both processes by the matrix metalloproteinase inhibitor BB-94 was observed, indicating the importance of endogenous MMP production in angiogenesis. In support of this finding, we demonstrated a significant increase in endogenous endothelial mRNA of MMP2, but not of cathepsins B and L, in proliferating primary human dermal microvascular endothelial cells (HMVEC-d) in culture. However, MMP2 mRNA expression was increased only when the cells were embedded in collagen but not when they were grown on plastic, regardless of the addition of the growth factors VEGF or bFGF. Moreover, on plastic the impairment of MMP2 induction by growth factors was observed. The differential effect of growth factors implies the crosstalk with integrin signaling as a consequence of binding to the different matrix. This study suggests that endothelial cell-associated cathepsins B and L are not involved in the invasive growth of capillaries from existing blood vessels and that the presence of collagen is necessary for MMP2 expression in endothelial cells.
Collapse
Affiliation(s)
- S Caserman
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | | |
Collapse
|
35
|
Koppert LB, Wijnhoven BPL, van Dekken H, Tilanus HW, Dinjens WNM. The molecular biology of esophageal adenocarcinoma. J Surg Oncol 2005; 92:169-90. [PMID: 16299787 DOI: 10.1002/jso.20359] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Barrett's esophagus is an acquired metaplastic change that occurs in the distal esophagus secondary to chronic gastroesophageal reflux. This premalignant condition forms the most important risk factor for developing esophageal adenocarcinoma, which is an extremely aggressive tumor with a 5-year survival rate of less than 25%. Carcinomas that arise in the setting of Barrett's esophagus are thought to develop as part of the metaplasia-dysplasia-carcinoma sequence. OBJECTIVE To review the current knowledge on the genomic alterations involved in the development of Barrett's esophagus and its progression to dysplasia and/or cancer. RESULTS Several changes in gene structure, gene expression, and protein structure are associated with the progression of Barrett's esophagus to adenocarcinoma. Accumulation of these changes seems to be essential, rather than the exact sequence of these changes. Multiple molecular pathways are involved and interact with each other. Alterations in tumor suppressor genes, amongst which p53 and p16, are early events in the metaplasia-dysplasia-adenocarcinoma sequence, followed by loss of cell cycle checkpoints. Ongoing genomic instability leads to cumulative genetic errors and thereby the generation of multiple clones of transformed cells. CONCLUSIONS Within the multistep process of esophageal adenocarcinogenesis, to date no single molecular marker came forward able to predict who will and who will not develop cancer in the setting of Barrett's esophagus. Instead, panels of markers need to be developed in the future allowing to indicate disease progression. Identification of crucial molecular pathways involved in esophageal adenocarcinogenesis would ultimately improve therapy and facilitate development of new treatment strategies.
Collapse
Affiliation(s)
- Linetta B Koppert
- Department of Surgery, Erasmus MC, University Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
36
|
Gondi CS, Lakka SS, Dinh DH, Olivero WC, Gujrati M, Rao JS. RNAi-mediated inhibition of cathepsin B and uPAR leads to decreased cell invasion, angiogenesis and tumor growth in gliomas. Oncogene 2004; 23:8486-96. [PMID: 15378018 DOI: 10.1038/sj.onc.1207879] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2003] [Revised: 04/16/2004] [Accepted: 05/06/2004] [Indexed: 11/09/2022]
Abstract
RNA interference (RNAi) provides a powerful method for gene silencing in eukaryotic cells, including proliferating mammalian cells. Here, we determined whether RNAi could be utilized to inhibit the expression of proteases implicated in the extracellular matrix degradation, which is characteristic of tumor progression. We have previously shown that antisense stable clones of uPAR and cathepsin B were less invasive and did not form tumors when injected intracranially ex vivo. Since antisense-mediated gene silencing does not completely inhibit the translation of target mRNA and high molar concentrations of antisense molecules are required to achieve gene silencing, we used the RNAi approach to silence uPAR and cathepsin B in this study. We found that the expression of double-stranded RNA leads to the efficient and specific inhibition of endogenous uPAR and cathepsin B protein expression in glioma cell lines as determined by Western blotting. We also found the RNAi of uPAR and cathepsin B reduces glioma cell invasion and angiogenesis in in vitro and in vivo models. Intratumoral injections of plasmid vectors expressing hpRNA for uPAR and cathepsin B resulted in the regression of pre-established intracranial tumors. Further, RNAi for uPAR and cathepsin B inhibited cell proliferation and reduced the levels of pERK and pFAK compared to controls. Taken together, our findings indicate for the first time that RNAi operates in human glioma cells with potential application for cancer gene therapy.
Collapse
Affiliation(s)
- Christopher S Gondi
- Program of Cancer Biology, Department of Biomedical and Therapeutic Sciences, University of Illinois College of Medicine-Peoria, IL 61656, USA
| | | | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- Umar Mahmood
- MGH Center for Molecular Imaging Research, Charlestown, MA 02129, USA.
| |
Collapse
|
38
|
Gondi CS, Lakka SS, Yanamandra N, Olivero WC, Dinh DH, Gujrati M, Tung CH, Weissleder R, Rao JS. Adenovirus-Mediated Expression of Antisense Urokinase Plasminogen Activator Receptor and Antisense Cathepsin B Inhibits Tumor Growth, Invasion, and Angiogenesis in Gliomas. Cancer Res 2004; 64:4069-77. [PMID: 15205313 DOI: 10.1158/0008-5472.can-04-1243] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have shown previously that urokinase plasminogen activator receptor (uPAR) and cathepsin B are overexpressed during glioma progression, particularly at the leading edge of the tumor. In the present study, we simultaneously down-regulated uPAR and cathepsin B in SNB19 glioma cell monolayer or SNB19 spheroids using an adenoviral vector carrying antisense uPAR and antisense cathepsin B and a combination of these genes as determined by Western blot analysis. The Ad-uPAR-Cath B-infected cells revealed a marked reduction in tumor growth and invasiveness as compared with the parental and vector controls. In vitro and in vivo angiogenic assays demonstrated inhibition of capillary-like structure formation and microvessel formation after Ad-uPAR-Cath B infection of SNB19 cells when compared with Ad-cytomegalovirus (CMV)-infected or mock-infected controls. Furthermore, using a near infrared fluorescence probe, in vivo imaging for cathepsin B indicated low/undetectable levels of fluorescence after injection of the Ad-uPAR-Cath B construct into pre-established s.c. tumors as compared with Ad-CMV-treated and untreated tumors. The effect with bicistronic construct (Ad-uPAR-Cath B) was much higher than with single (Ad-uPAR/Ad-Cath B) constructs. These results indicate that the down-regulation of cathepsin B and uPAR plays a significant role in inhibiting tumor growth, invasion, and angiogenesis. Hence, the targeting of these two proteases may be a potential therapy for brain tumors and other cancers.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Cathepsin B/antagonists & inhibitors
- Cathepsin B/genetics
- Cell Division/genetics
- Cell Movement/genetics
- DNA, Antisense/administration & dosage
- DNA, Antisense/genetics
- Female
- Genetic Therapy/methods
- Glioblastoma/blood supply
- Glioblastoma/genetics
- Glioblastoma/therapy
- Glioblastoma/virology
- Humans
- Male
- Mice
- Mice, Nude
- Neoplasm Invasiveness
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/therapy
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/genetics
- Receptors, Urokinase Plasminogen Activator
- Spheroids, Cellular
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Christopher S Gondi
- Program of Cancer Biology and Department of Biomedical and Therapeutic Sciences, University of Illinois College of Medicine, Peoria, 61656, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Witmer AN, van Blijswijk BC, van Noorden CJF, Vrensen GFJM, Schlingemann RO. In vivo angiogenic phenotype of endothelial cells and pericytes induced by vascular endothelial growth factor-A. J Histochem Cytochem 2004; 52:39-52. [PMID: 14688216 DOI: 10.1177/002215540405200105] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
VEGF-A is a major angiogenesis and permeability factor. Its cellular effects, which can be used as targets in anti-angiogenesis therapy, have mainly been studied in vitro using endothelial cell cultures. The purpose of the present study was to further characterize these effects in vivo in vascular endothelial cells and pericytes, in an experimental monkey model of VEGF-A-induced iris neovascularization. Two cynomolgus monkeys (Macaca fascicularis) received four injections of 0.5 microg VEGF-A in the vitreous of one eye and PBS in the other eye. After sacrifice at day 9, eyes were enucleated and iris samples were snap-frozen for immunohistochemistry (IHC) and stained with a panel of antibodies recognizing endothelial and pericyte determinants related to angiogenesis and permeability. After VEGF-A treatment, the pre-existing iris vasculature showed increased permeability, hypertrophy, and activation, as demonstrated by increased staining of CD31, PAL-E, tPA, uPA, uPAR, Glut-1, and alphavbeta3 and alphavbeta5 integrins, VEGF receptors VEGFR-1, -2 and -3, and Tie-2 in endothelial cells, and of NG2 proteoglycan, uPA, uPAR, integrins and VEGFR-1 in pericytes. Vascular sprouts at the anterior surface of the iris were positive for the same antigens except for tPA, Glut-1, and Tie-2, which were notably absent. Moreover, in these sprouts VEGFR-2 and VEGFR-3 expression was very high in endothelial cells, whereas many pericytes were present that were positive for PDGFR-beta, VEGFR-1, and NG2 proteoglycan and negative for alpha-SMA. In conclusion, proteins that play a role in angiogenesis are upregulated in both pre-existing and newly formed iris vasculature after treatment with VEGF-A. VEGF-A induces hypertrophy and loss of barrier function in pre-existing vessels, and induces angiogenic sprouting, characterized by marked expression of VEGFR-3 and lack of expression of tPA and Tie-2 in endothelial cells, and lack of alpha-SMA in pericytes. Our in vivo study indicates a role for alpha-SMA-negative pericytes in early stages of angiogenesis. Therefore, our findings shed new light on the temporal and spatial role of several proteins in the angiogenic cascade in vivo.
Collapse
Affiliation(s)
- Antonella N Witmer
- Ocular Angiogenesis Group, Department of Ophthalmology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
40
|
Shridhar R, Zhang J, Song J, Booth BA, Kevil CG, Sotiropoulou G, Sloane BF, Keppler D. Cystatin M suppresses the malignant phenotype of human MDA-MB-435S cells. Oncogene 2003; 23:2206-15. [PMID: 14676833 DOI: 10.1038/sj.onc.1207340] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Proteases are involved in many aspects of tumor progression, including cell survival and proliferation, escape from immune surveillance, cell adhesion and migration, remodeling and invasion of the extracellular matrix. Several lysosomal cysteine proteases have been cloned and shown to be overexpressed in cancer; yet, despite the great potential for development of novel therapeutics, we still know little about the regulation of their proteolytic activity. Cystatins such as cystatin M are potent endogenous protein inhibitors of lysosomal cysteine proteases. Cystatin M is expressed in normal and premalignant human epithelial cells, but not in many cancer cell lines. Here, we examined the effects of cystatin M expression on malignant properties of human breast carcinoma MDA-MB-435S cells. Cystatin M was found to significantly reduce in vitro: cell proliferation, migration, Matrigel invasion, and adhesion to endothelial cells. Reduction of cell proliferation and adhesion to an endothelial cell monolayer were both independent of the inhibition of lysosomal cysteine proteases. In contrast, cell migration and matrix invasion seemed to rely on lysosomal cysteine proteases, as both recombinant cystatin M and E64 were able to block these processes. This study provides the first evidence that cystatin M may play important roles in safeguarding against human breast cancer.
Collapse
Affiliation(s)
- Ravi Shridhar
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Cao X, Deng X, May WS. Cleavage of Bax to p18 Bax accelerates stress-induced apoptosis, and a cathepsin-like protease may rapidly degrade p18 Bax. Blood 2003; 102:2605-14. [PMID: 12816867 DOI: 10.1182/blood-2003-01-0211] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bax is cleaved by calpain at aspartate 33 (Asp33) to yield p18 Bax during stress-induced apoptosis. To assess the role of p18 Bax in apoptosis, an ecdysone-inducible expression system was generated. Similar levels of wild-type (WT) and noncleavable Asp33Ala (Asp-->Ala) Bax are induced in 293 cells while expression of N-terminal-deleted p18 (Delta1-33) Bax remains low (20% of full-length p21 Bax) due to a reduced half-life (2 hours versus 12 hours for p21 Bax) resulting from increased sensitivity to cathepsin-like proteolytic degradation. Expression of p18 Bax is enhanced to levels comparable to p21 Bax when induction is carried out in the presence of cathepsin inhibitors, Z-Phe-Gly-NHO-Bz or N-Acetyl-Leu-Leu-Met-CHO. Compared with WT Bax, expression of similar levels of p18 Bax and, surprisingly, Asp33Ala Bax more potently induces apoptosis as indicated by increased cytochrome c release, caspase-9/-3 activation, and DNA fragmentation, potentially due to their increased homo-oligomerization in mitochondrial membranes. Studies in A-549, U-937, K-562, and HL-60 cells confirm that inhibition of Bax cleavage results in 25% to 35% reduction of drug-induced apoptosis, while inhibition of p18 Bax degradation enhances apoptosis by 25% to 40%. Results indicate that although cleavage to p18 Bax is not required for Bax to initiate apoptosis, p18 Bax potently accelerates the apoptotic process.
Collapse
Affiliation(s)
- Xuefang Cao
- University of Florida Shands Cancer Center, UF Shands Cancer Center, Gainesville, USA
| | | | | |
Collapse
|
42
|
Fan X, Kopitar-Jerala N, Premzl A, Bestagno M, Burrone O, Kos J. Molecular cloning and chimerisation of an inhibitory anti-cathepsin B antibody and its expression in Chinese hamster ovary cells. Biol Chem 2002; 383:1817-20. [PMID: 12530548 DOI: 10.1515/bc.2002.204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The lysosomal cysteine protease cathepsin B is one of several proteases that have been linked to tumour progression. Its increased expression and secretion in tumour cells may facilitate the degradation of extracellular matrix proteins, leading to tumour cell invasion and metastasis. Specific inhibitory monoclonal antibodies are a possible alternative to synthetic inhibitors as a therapeutic tool for cancer treatment. An inhibitory monoclonal antibody, which binds to an epitope near the active site of cathepsin B and inhibits its proteolytic activity, was prepared and its effect on invasion of ras-transformed MCF-10A neoT cells was tested in vitro. Here we present the nucleotide sequences of the heavy and light chains of the inhibitory antibody and compare them to the murine immunoglobulin germline sequences for possible somatic hypermutations. Since no harmful mutations were found, a mouse/human chimeric antibody was constructed by fusing murine V(H) and V(L) variable regions of the inhibitory antibody with human gamma 1 and K constant regions, respectively. Chinese hamster ovary K1 cells were co-transfected with expression vectors pcD-NA3L and pcDNA3H and the reactivity of the isolated chimeric antibody was tested by ELISA and Western blotting. We could demonstrate an inhibitory effect of the chimeric antibody.
Collapse
Affiliation(s)
- Xiaohui Fan
- Department of Biochemistry and Molecular Biology, Jozef Stefan Institute, Jamova 39, SI-1 000 Ljubljana, Slovenia
| | | | | | | | | | | |
Collapse
|
43
|
Cathers BE, Barrett C, Palmer JT, Rydzewski RM. pH Dependence of inhibitors targeting the occluding loop of cathepsin B. Bioorg Chem 2002; 30:264-75. [PMID: 12392705 DOI: 10.1016/s0045-2068(02)00009-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Potent and selective cathepsin B inhibitors have previously been synthesized based upon the natural product cysteine protease inhibitor E-64. X-ray crystal data indicates that these compounds interact through their free carboxylate with the positively charged histidine residues located on the prime-side of the active site within the occluding loop of cathepsin B. Herein, we examine the pH dependence of two prime-side-binding compounds. In each case there is a dramatic decrease in k(inact)/K(I) as the pH is raised from 4 to 7.8 corresponding to a single ionization of pK(a) 4.4. These results suggest that targeting of the occluding loop of cathepsin B may be a poor inhibitor design strategy if the enzyme environment has a pH greater than 5.5. However, this type of inhibitor may be a useful tool to help elucidate the role and the environment of cathepsin B in invading tumors.
Collapse
Affiliation(s)
- Brian E Cathers
- Axys Pharmaceuticals Inc, 180 Kimball Way, South San Francisco, CA 94080, USA.
| | | | | | | |
Collapse
|
44
|
Arakawa K, Endo Y, Kimura M, Yoshida T, Kitaoka T, Inakazu T, Nonami Y, Abe M, Masuyama A, Nojima M, Sasaki T. Multifunctional anti-angiogenic activity of the cyclic peroxide ANO-2 with antitumor activity. Int J Cancer 2002; 100:220-7. [PMID: 12115573 DOI: 10.1002/ijc.10469] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Our focus was to develop an anti-angiogenic drug possessing the inhibitory activity of urokinase-type plasminogen activator (u-PA) production. During preliminary screening, the effects of 13 ozonides on the inhibition of u-PA production in human fibrosarcoma HT-1080 cells and on the inhibition of angiogenesis on chicken embryonic chorioallantoic membranes were determined. Of the ozonides tested, 9 inhibited in vitro u-PA production of HT-1080 cells and 7 of these 9 exhibited strong anti-angiogenic activity. Interestingly, 6 of the 13 ozonides also inhibited cathepsin B activity. 1-Phenyl-1, 4-epoxy-1H,4H-naphtho[1,8-de][1, 2]dioxepin (ANO-2) potently inhibited cathepsin B (IC(50) = 0.47 microM) as well as u-PA production. Consequently, ANO-2 was selected for further study. ANO-2 inhibited tube formation by human umbilical vein endothelial cells cultured on Matrigel while exhibiting no cytotoxicity. Additionally, in vivo administration of ANO-2 inhibited angiogenesis induced by mouse Sarcoma-180 cells tested using the mouse dorsal air sac assay. Moreover, ANO-2 also suppressed primary tumor growth and reduced the number of pulmonary metastases caused by Lewis lung carcinoma cells in mice. These in vitro and in vivo activities indicate that ANO-2 has considerable potential as a new and potent anti-angiogenic drug that inhibits both u-PA production and enzymatic activity of cathepsins, indicating that ANO-2 may be a multifunctional inhibitor of angiogenesis.
Collapse
Affiliation(s)
- Kazuhito Arakawa
- Department of Experimental Therapeutics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mohanam S, Jasti SL, Kondraganti SR, Chandrasekar N, Lakka SS, Kin Y, Fuller GN, Yung AW, Kyritsis AP, Dinh DH, Olivero WC, Gujrati M, Ali-Osman F, Rao JS. Down-regulation of cathepsin B expression impairs the invasive and tumorigenic potential of human glioblastoma cells. Oncogene 2001; 20:3665-73. [PMID: 11439329 DOI: 10.1038/sj.onc.1204480] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2000] [Revised: 03/07/2001] [Accepted: 03/21/2001] [Indexed: 11/08/2022]
Abstract
Increases in abundance of cathepsin B transcript and protein correlate with increases in tumor grade and alterations in subcellular localization and activity of cathepsin B. The enzyme is able to degrade the components of the extracellular matrix (ECM) and activate other proteases capable of degrading ECM. To investigate the role played by this protease in the invasion of brain tumor cells, we transfected SNB19 human glioblastoma cells with a plasmid containing cathepsin B cDNA in antisense orientation. Control cells were transfected with vector alone. Clones expressing antisense cathepsin B cDNA exhibited significant reductions in cathepsin B mRNA, enzyme activity and protein compared to controls. Matrigel Invasion assay showed that the antisense-transfected cells had a markedly diminished invasiveness compared with controls. When tumor spheroids containing antisense transfected SNB19 cells expressing reduced cathepsin B were co-cultured with fetal rat brain aggregates, invasion of fetal rat brain aggregates was significantly reduced. Green Fluorescent Protein (GFP) expressing parental cells and antisense transfectants were generated for detection in mouse brain tissue without any post-chemical treatment. Intracerebral injection of SNB19 stable antisense transfectants resulted in reduced tumor formation in nude mice. These results strongly support a role for cathepsin B in the invasiveness of human glioblastoma cells and suggest cathepsin B antisense may prove useful in cancer therapy.
Collapse
Affiliation(s)
- S Mohanam
- Division of Cancer Biology, Department of Biomedical and Therapeutic Sciences, University of Illinois College of Medicine at Peoria, 61656, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pallini R, Pierconti F, Falchetti ML, D'Arcangelo D, Fernandez E, Maira G, D'Ambrosio E, Larocca LM. Evidence for telomerase involvement in the angiogenesis of astrocytic tumors: expression of human telomerase reverse transcriptase messenger RNA by vascular endothelial cells. J Neurosurg 2001; 94:961-71. [PMID: 11409526 DOI: 10.3171/jns.2001.94.6.0961] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECT Evidence from recent in vitro studies indicates that reactivation of telomerase, the enzyme that synthesizes the telomere ends of chromosomes, is a crucial event in the unlimited clonal expansion of endothelial cells that precedes the neoplastic conversion of these cells. It is known that high-grade gliomas express telomerase and that, in these neoplasms, proliferating endothelial cells may undergo transformational changes with development of sarcomatous components within the primitive tumor. To assess whether telomerase is involved in the endothelial cell proliferation that characterizes brain tumor angiogenesis, the authors investigated at the single-cell level the expression of messenger (m)RNA for the human telomerase catalytic subunit human telomerase reverse transcriptase (hTERT) by vascular cells of astrocytic tumors. METHODS The in situ hybridization (ISH) method was performed by processing histological sections with specific riboprobes for hTERT and for c-myc, an oncogene that is known to upregulate hTERT. Results of the ISH studies were compared with proliferative activity, as estimated by Ki-67 immunostaining. The expression of hTERT mRNA by vascular endothelial cells was related to the histological grade of the tumor because it was detected in five (29%) of 17 low-grade astrocytomas, nine (56%) of 16 anaplastic astrocytomas, and 19 (100%) of 19 glioblastomas multiforme (GBMs). Expression of c-myc mRNA was strictly correlated with that of hTERT mRNA. In low-grade astrocytomas and anaplastic astrocytomas, a dissociation was noted between hTERT mRNA expression and the proliferation rate of endothelial cells. Conversely, GBMs displayed a significant correlation between the level of hTERT mRNA expression and endothelial cell proliferation. Data from an in vitro assay in which human umbilical vein endothelial cells were stimulated to proliferate by adding vascular endothelial growth factor and an ISH study of newly formed vessels surrounding brain infarcts confirmed that expression of hTERT mRNA does not merely reflect the proliferative status of endothelial cells but represents a specific feature of brain tumor neovascularization. CONCLUSIONS The results of this study are consistent with a role of telomerase in the angiogenesis of astrocytic tumors. Expression of hTERT mRNA by tumor vascular cells is an early event during the progression of astrocytic tumors, which precedes endothelial cell proliferation and may represent a first sign of dedifferentiation. Other than elucidating the mechanisms of tumor angiogenesis, these results encourage research on antitelomerase drugs for the treatment of malignant gliomas.
Collapse
Affiliation(s)
- R Pallini
- Istituto di Neurochirurgia, Università Cattolica, Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
OBJECTIVE To review the current knowledge on the genetic alterations involved in the development and progression of Barrett's esophagus-associated neoplastic lesions. SUMMARY BACKGROUND DATA Barrett's esophagus (BE) is a premalignant condition in which the normal squamous epithelium of the esophagus is replaced by metaplastic columnar epithelium. BE predisposes patients to the development of esophageal adenocarcinoma. Endoscopic surveillance can detect esophageal adenocarcinomas when they are early and curable, but most of the adenocarcinomas are detected at an advanced stage. Despite advances in multimodal therapy, the prognosis for invasive esophageal adenocarcinoma is poor. A better understanding of the molecular evolution of the Barrett's metaplasia to dysplasia to adenocarcinoma sequence may allow improved diagnosis, therapy, and prognosis. METHODS The authors reviewed data from the published literature to address what is known about the molecular changes thought to be important in the pathogenesis of BE-associated neoplastic lesions. RESULTS The progression of Barrett's metaplasia to adenocarcinoma is associated with several changes in gene structure, gene expression, and protein structure. Some of the molecular alterations already showed promise as markers for early cancer detection or prognostication. Among these, alterations in the p53 and p16 genes and cell cycle abnormalities or aneuploidy appear to be the most important and well-characterized molecular changes. However, the exact sequence of events is not known, and probably multiple molecular pathways interact and are involved in the progression of BE to adenocarcinoma. CONCLUSIONS Further research into the molecular biology of BE-associated adenocarcinoma will enhance our understanding of the genetic events critical for the initiation and progression of Barrett's adenocarcinoma, leading to more effective surveillance and treatment.
Collapse
Affiliation(s)
- B P Wijnhoven
- Department of Surgery, University Hospital Rotterdam, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | | | | |
Collapse
|
48
|
Hazen LG, Bleeker FE, Lauritzen B, Bahns S, Song J, Jonker A, Van Driel BE, Lyon H, Hansen U, Köhler A, Van Noorden CJ. Comparative localization of cathepsin B protein and activity in colorectal cancer. J Histochem Cytochem 2000; 48:1421-30. [PMID: 10990495 DOI: 10.1177/002215540004801012] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cathepsin B is a lysosomal cysteine proteinase that may participate in cancer progression. We compared localization of its protein and activity during progression of human colorectal cancer. In adenomas and carcinomas, protein expression and, particularly, activity were elevated compared with those in normal colorectal mucosa. In normal mucosa, cathepsin B protein expression was moderate in stroma and variable in epithelium, whereas activity was mainly present in distinct areas of stroma directly underneath the surface of the colon and in epithelium at the surface of the colon. Stroma in adenomas and carcinomas contained moderate to high protein levels but little activity except for areas of angiogenesis, inflammation, and necrosis, in which activity was high. In adenomas and the majority of well-differentiated carcinomas and moderately differentiated carcinomas, cathepsin B protein and activity were found in granular form in the epithelium, close to the basement membrane. Protein and activity levels were low and diffusely distributed in cancer cells in the remainder of the well-differentiated and moderately differentiated carcinomas and in all poorly differentiated carcinomas. Invasive fronts in most cancers contained moderate protein levels but high activity. We conclude that (a) activity localization is essential to understand the role of cathepsin B in cancer progression, and (b) cathepsin B activity in human colon is associated with invasion of cancer cells, endothelial cells, and inflammatory cells, and in cell death, both apoptotic and necrotic.
Collapse
Affiliation(s)
- L G Hazen
- Academic Medical Center, University of Amsterdam, Department of Cell Biology and Histology, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Yamashima T. Implication of cysteine proteases calpain, cathepsin and caspase in ischemic neuronal death of primates. Prog Neurobiol 2000; 62:273-95. [PMID: 10840150 DOI: 10.1016/s0301-0082(00)00006-x] [Citation(s) in RCA: 253] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Although more than 8000 papers of apoptosis are published annually, there are very few reports concerning necrosis in the past few years. A number of recent studies using lower species animals have suggested that the cornu Ammonis (CA) 1 neuronal death after brief global cerebral ischemia occurs by apoptosis, an active and genetically controlled cell suicide process. However, the studies of monkeys and humans rather support necrosis, the calpain-mediated release of lysosomal enzyme cathepsin after ischemia conceivably contributes to the cell degeneration of CA1 neurons. This paper provides an overview of recent developments in ischemic neuronal death, presents the cascade of the primate neuronal death with particular attentions to the cysteine proteases, and also indicates selective cathepsin inhibitors as a novel neuroprotectant. Furthermore, the possible interaction of calpain, cathepsin, and caspase in the cascade of ischemic neuronal death is discussed.
Collapse
Affiliation(s)
- T Yamashima
- Department of Neurosurgery, Kanazawa University School of Medicine, 920-8641, Kanazawa, Japan.
| |
Collapse
|
50
|
Zhu DM, Uckun FM. Cathepsin inhibition induces apoptotic death in human leukemia and lymphoma cells. Leuk Lymphoma 2000; 39:343-54. [PMID: 11342315 DOI: 10.3109/10428190009065834] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We examined the effects of cathepsin inhibitor 1 (CATI-1), a selective inhibitor of cysteine cathepsins, on human leukemia and lymphoma cells. CATI-1 induced apoptosis in all 12 cell lines tested. Apoptosis of CATI-1-treated leukemia/lymphoma cells was caspase-independent, p53-independent, BAX-independent as well as MAP kinase-independent. Our findings provide unprecedented experimental evidence that cathepsins play a pivotal role for the survival of human leukemia/lymphoma cells. Therefore, cathepsin inhibitors may provide the basis for new treatment programs against leukemia and lymphoma.
Collapse
Affiliation(s)
- D M Zhu
- Parker Hughes Cancer Center; Parker Hughes Institute, 2665 Long Lake Road, Roseville, MN 55113, USA
| | | |
Collapse
|