首页 | 本学科首页   官方微博 | 高级检索  
   检索      


Proteomics Analysis of Cancer Exosomes Using a Novel Modified Aptamer-based Array (SOMAscanTM) Platform
Authors:Jason Webber  Timothy C Stone  Evaldas Katilius  Breanna C Smith  Bridget Gordon  Malcolm D Mason  Zsuzsanna Tabi  Ian A Brewis  Aled Clayton
Institution:3. Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff CF14 2TL, United Kingdom;;5. Central Biotechnology Services and Institute of Translation, Innovation, Methodology and Engagement, Henry Wellcome Building, School of Medicine, Heath Park, Cardiff University, Cardiff CF14 4XN, United Kingdom;;6. Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, United Kingdom;
Abstract:We have used a novel affinity-based proteomics technology to examine the protein signature of small secreted extracellular vesicles called exosomes. The technology uses a new class of protein binding reagents called SOMAmers® (slow off-rate modified aptamers) and allows the simultaneous precise measurement of over 1000 proteins. Exosomes were highly purified from the Du145 prostate cancer cell line, by pooling selected fractions from a continuous sucrose gradient (within the density range of 1.1 to 1.2 g/ml), and examined under standard conditions or with additional detergent treatment by the SOMAscanTM array (version 3.0). Lysates of Du145 cells were also prepared, and the profiles were compared. Housekeeping proteins such as cyclophilin-A, LDH, and Hsp70 were present in exosomes, and we identified almost 100 proteins that were enriched in exosomes relative to cells. These included proteins of known association with cancer exosomes such as MFG-E8, integrins, and MET, and also those less widely reported as exosomally associated, such as ROR1 and ITIH4. Several proteins with no previously known exosomal association were confirmed as exosomally expressed in experiments using individual SOMAmer® reagents or antibodies in micro-plate assays. Western blotting confirmed the SOMAscanTM-identified enrichment of exosomal NOTCH-3, L1CAM, RAC1, and ADAM9. In conclusion, we describe here over 300 proteins of hitherto unknown association with prostate cancer exosomes and suggest that the SOMAmer®-based assay technology is an effective proteomics platform for exosome-associated biomarker discovery in diverse clinical settings.Prostate carcinoma is the most frequent male cancer, with an estimated 240,000 newly diagnosed individuals and 28,000 deaths in the United States during 2012 (National Cancer Institute (NIH)). Methods for detecting this cancer are based on a combination of physical examination through digital rectal examination, clinical imaging, quantification of circulating levels of prostate specific antigen (PSA),1 and transrectal ultrasound-guided biopsy. As a non-invasive test, PSA measurement is still widely used, but it remains insensitive, as around 15% of men with normal levels of PSA will have prostate cancer according to biopsy results (1), and 60% of men with elevated PSA levels may have other, noncancerous conditions but be subjected to further, unnecessary investigations and interventions (2). PSA may be of better utility in monitoring disease progression (2). An ability to diagnose the disease more specifically at an early stage is likely to save lives and alleviate the healthcare burden and morbidities arising from misdiagnosis. In addition, methods for monitoring the course of the disease in a non-invasive and perhaps predictive manner would offer increased patient benefit, enabling early detection of imminent relapse under hormone therapy, for example. Therefore there is a clinical need for improved molecular approaches for disease diagnosis and monitoring in these settings.Small vesicles termed exosomes are present in body fluids, including serum, plasma, urine, and seminal plasma (37), and their isolation and examiniation may prove useful as a minimally invasive means of obtaining a complex set of disease markers. Exosomes are secreted by most, if not all, cell types and are generally accepted as derived principally from multivesicular bodies of the late endocytic tract (8), although examples of plasma membrane budding nanovesicles of similar phenotype have also been described (9). Exosomes are particularly enriched in membrane proteins and in factors related to such endosomal compartments. They also contain proteins found in the cytosol, but they poorly represent components of organelles such as the mitochondria, nucleus, and endoplasmic reticulum (10). Exosomes also comprise an assortment of coding and noncoding RNA. There has been considerable global effort toward defining disease-related alterations in exosomal RNA. However, it is well established that aberrant alterations in cancer cells in response to metabolic, hypoxic, or other forms of stress are reflected in protein changes in the exosomes produced (1113). Thus exosomes from diseased origins can be distinguished from those of a normal phenotype based on their protein profiles alone.Proteomics studies using mass spectrometry (MS) have previously been conducted on prostate cancer exosomes/microvesicles obtained from cell lines (14, 15), xenotransplantation models (16), or ex vivo biofluids (17). Hundreds of proteins with putative associations with exosomes/microvesicles have been identified. These studies highlight several interesting candidate markers of potential biomarker utility that are currently being explored. However, global proteomic approaches of this nature can have two major limitations. Although the most abundant proteins are more likely to be identified by MS, it is difficult to infer information about relative abundances of proteins in complex samples when using these methods. Secondly, given the often exacting, difficult-to-reproduce, and time-consuming workflows involved, such technologies are poorly suited for the analysis of a large number of samples. Multiplex protein array methodologies have the potential to overcome such issues and offer quantification and options for more rapid sample throughput. However, most platforms are based on antibodies, and these arrays are typically limited to <100 proteins, principally because the cross-reactivity of secondary antibodies can negatively affect assay specificity (18).A recently developed proteomics platform, termed SOMAscanTM, provides a new generation of protein detection technologies. The platform is capable of the simultaneous quantitative analysis of 1129 proteins per sample in its current form. It is also an approach well suited to handling large numbers of specimens required for well-powered clinical studies (19). The key to this technology, which is described in detail by Gold et al. (20, 21), is the use of slow off-rate modified aptamers (SOMAmers) containing chemically modified nucleotides. This confers greater stability, expanded target range, and improved affinity for the target proteins. This multiplex platform has been applied successfully to small volumes (∼15 μl) of plasma specimens from chronic renal disease patients (20), serum specimens from mesothelioma (22) or lung cancer patients (19), tissue lysates (23), and cerebrospinal fluid (24). However, to date, the compatibility of this array technology with exosomes as the specimen has not been investigated.The purpose of the current study was to examine the utility of this evolving technology in profiling the protein repertoire of exosomes. Research was conducted using highly pure exosomes isolated from a prostate cancer cell line, and we compared this sample to the protein profile of the parent cells. By so doing, we obtained evidence of the compatibility of the platform with this difficult, membranous sample and identified several proteins of previously unknown association with exosomes. In summary, SOMAscanTM is a versatile tool for probing the composition of exosomes and is a suitable platform to provide a high-throughput approach for exosome-based biomarker discovery in prostate cancer and other clinical settings.
Keywords:
本文献已被 ScienceDirect 等数据库收录!
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号