2101
|
Altadill T, Campoy I, Lanau L, Gill K, Rigau M, Gil-Moreno A, Reventos J, Byers S, Colas E, Cheema AK. Enabling Metabolomics Based Biomarker Discovery Studies Using Molecular Phenotyping of Exosome-Like Vesicles. PLoS One 2016; 11:e0151339. [PMID: 26974972 PMCID: PMC4790956 DOI: 10.1371/journal.pone.0151339] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/26/2016] [Indexed: 12/21/2022] Open
Abstract
Identification of sensitive and specific biomarkers with clinical and translational utility will require smart experimental strategies that would augment expanding the breadth and depth of molecular measurements within the constraints of currently available technologies. Exosomes represent an information rich matrix to discern novel disease mechanisms that are thought to contribute to pathologies such as dementia and cancer. Although proteomics and transcriptomic studies have been reported using Exosomes-Like Vesicles (ELVs) from different sources, exosomal metabolome characterization and its modulation in health and disease remains to be elucidated. Here we describe methodologies for UPLC-ESI-MS based small molecule profiling of ELVs from human plasma and cell culture media. In this study, we present evidence that indeed ELVs carry a rich metabolome that could not only augment the discovery of low abundance biomarkers but may also help explain the molecular basis of disease progression. This approach could be easily translated to other studies seeking to develop predictive biomarkers that can subsequently be used with simplified targeted approaches.
Collapse
Affiliation(s)
- Tatiana Altadill
- Biomedical Research Group in Ginecology, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irene Campoy
- Biomedical Research Group in Ginecology, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Lucia Lanau
- Biomedical Research Group in Ginecology, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Kirandeep Gill
- Departments of Oncology and Biochemistry, Molecular and Cellular Biology, Lombardi Comprehensive Cancer Center at Georgetown University Medical Center, Washington, D.C., United States of America
| | - Marina Rigau
- Institut d’Investigació Biomedica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Antonio Gil-Moreno
- Gynecological Department, Vall Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jaume Reventos
- Institut d’Investigació Biomedica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Stephen Byers
- Departments of Oncology and Biochemistry, Molecular and Cellular Biology, Lombardi Comprehensive Cancer Center at Georgetown University Medical Center, Washington, D.C., United States of America
| | - Eva Colas
- Biomedical Research Group in Ginecology, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pathology and Molecular Genetics/Oncologic Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, Lleida, Spain
| | - Amrita K. Cheema
- Departments of Oncology and Biochemistry, Molecular and Cellular Biology, Lombardi Comprehensive Cancer Center at Georgetown University Medical Center, Washington, D.C., United States of America
| |
Collapse
|
2102
|
Alix-Panabières C, Pantel K. Clinical Applications of Circulating Tumor Cells and Circulating Tumor DNA as Liquid Biopsy. Cancer Discov 2016; 6:479-91. [PMID: 26969689 DOI: 10.1158/2159-8290.cd-15-1483] [Citation(s) in RCA: 988] [Impact Index Per Article: 109.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/18/2016] [Indexed: 02/07/2023]
Abstract
UNLABELLED "Liquid biopsy" focusing on the analysis of circulating tumor cells (CTC) and circulating cell-free tumor DNA (ctDNA) in the blood of patients with cancer has received enormous attention because of its obvious clinical implications for personalized medicine. Analyses of CTCs and ctDNA have paved new diagnostic avenues and are, to date, the cornerstones of liquid biopsy diagnostics. The present review focuses on key areas of clinical applications of CTCs and ctDNA, including detection of cancer, prediction of prognosis in patients with curable disease, monitoring systemic therapies, and stratification of patients based on the detection of therapeutic targets or resistance mechanisms. SIGNIFICANCE The application of CTCs and ctDNA for the early detection of cancer is of high public interest, but it faces serious challenges regarding specificity and sensitivity of the current assays. Prediction of prognosis in patients with curable disease can already be achieved in several tumor entities, particularly in breast cancer. Monitoring the success or failure of systemic therapies (i.e., chemotherapy, hormonal therapy, or other targeted therapies) by sequential measurements of CTCs or ctDNA is also feasible. Interventional studies on treatment stratification based on the analysis of CTCs and ctDNA are needed to implement liquid biopsy into personalized medicine. Cancer Discov; 6(5); 479-91. ©2016 AACR.
Collapse
Affiliation(s)
- Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), Department of Cellular and Tissular Biopathology of Tumors, University Medical Centre, Montpellier, France. EA2415 - Help for Personalized Decision, Methodological Aspects, University Institute of Clinical Research (IURC), Montpellier University, Montpellier, France
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
2103
|
Angeloni NL, McMahon KM, Swaminathan S, Plebanek MP, Osman I, Volpert OV, Thaxton CS. Pathways for Modulating Exosome Lipids Identified By High-Density Lipoprotein-Like Nanoparticle Binding to Scavenger Receptor Type B-1. Sci Rep 2016; 6:22915. [PMID: 26964503 PMCID: PMC4786789 DOI: 10.1038/srep22915] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/24/2016] [Indexed: 01/03/2023] Open
Abstract
Exosomes are produced by cells to mediate intercellular communication, and have been shown to perpetuate diseases, including cancer. New tools are needed to understand exosome biology, detect exosomes from specific cell types in complex biological media, and to modify exosomes. Our data demonstrate a cellular pathway whereby membrane-bound scavenger receptor type B-1 (SR-B1) in parent cells becomes incorporated into exosomes. We tailored synthetic HDL-like nanoparticles (HDL NP), high-affinity ligands for SR-B1, to carry a fluorescently labeled phospholipid. Data show SR-B1-dependent transfer of the fluorescent phospholipid from HDL NPs to exosomes. Modified exosomes are stable in serum and can be directly detected using flow cytometry. As proof-of-concept, human serum exosomes were found to express SR-B1, and HDL NPs can be used to label and isolate them. Ultimately, we discovered a natural cellular pathway and nanoparticle-receptor pair that enables exosome modulation, detection, and isolation.
Collapse
Affiliation(s)
- Nicholas L. Angeloni
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Northwestern University Feinberg School of Medicine, Chicago, IL United States
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Kaylin M. McMahon
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Northwestern University Feinberg School of Medicine, Chicago, IL United States
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, United States
| | - Suchitra Swaminathan
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Michael P. Plebanek
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Northwestern University Feinberg School of Medicine, Chicago, IL United States
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, United States
| | - Iman Osman
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY, United States
- Laura and Isaac Perlmutter Cancer Center, New York University, Langone Medical Center, New York, NY, United States
| | - Olga V. Volpert
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - C. Shad Thaxton
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Simpson Querrey Institute for BioNanotechnology, Northwestern University Feinberg School of Medicine, Chicago, IL United States
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- International Institute for Nanotechnology (IIN), Northwestern University, Evanston, IL, United States
| |
Collapse
|
2104
|
Mereiter S, Balmaña M, Gomes J, Magalhães A, Reis CA. Glycomic Approaches for the Discovery of Targets in Gastrointestinal Cancer. Front Oncol 2016; 6:55. [PMID: 27014630 PMCID: PMC4783390 DOI: 10.3389/fonc.2016.00055] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 02/24/2016] [Indexed: 12/22/2022] Open
Abstract
Gastrointestinal (GI) cancer is the most common group of malignancies and many of its types are among the most deadly. Various glycoconjugates have been used in clinical practice as serum biomarker for several GI tumors, however, with limited diagnose application. Despite the good accessibility by endoscopy of many GI organs, the lack of reliable serum biomarkers often leads to late diagnosis of malignancy and consequently low 5-year survival rates. Recent advances in analytical techniques have provided novel glycoproteomic and glycomic data and generated functional information and putative biomarker targets in oncology. Glycosylation alterations have been demonstrated in a series of glycoconjugates (glycoproteins, proteoglycans, and glycosphingolipids) that are involved in cancer cell adhesion, signaling, invasion, and metastasis formation. In this review, we present an overview on the major glycosylation alterations in GI cancer and the current serological biomarkers used in the clinical oncology setting. We further describe recent glycomic studies in GI cancer, namely gastric, colorectal, and pancreatic cancer. Moreover, we discuss the role of glycosylation as a modulator of the function of several key players in cancer cell biology. Finally, we address several state-of-the-art techniques currently applied in this field, such as glycomic and glycoproteomic analyses, the application of glycoengineered cell line models, microarray and proximity ligation assay, and imaging mass spectrometry, and provide an outlook to future perspectives and clinical applications.
Collapse
Affiliation(s)
- Stefan Mereiter
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Meritxell Balmaña
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona , Girona , Spain
| | - Joana Gomes
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Medical Faculty, University of Porto, Porto, Portugal
| |
Collapse
|
2105
|
Flynn AD, Yin H. Lipid-Targeting Peptide Probes for Extracellular Vesicles. J Cell Physiol 2016; 231:2327-32. [PMID: 26909741 DOI: 10.1002/jcp.25354] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 02/19/2016] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles released from cells are under intense investigation for their roles in cell-cell communication and cancer progression. However, individual vesicles have been difficult to probe as their small size renders them invisible by conventional light microscopy. However, as a consequence of their small size these vesicles possess highly curved lipid membranes that offer an unconventional target for curvature-sensing probes. In this article, we present a strategy for using peptide-based biosensors to detect highly curved membranes and the negatively charged membrane lipid phosphatidylserine, we delineate several assays used to validate curvature- and lipid-targeting mechanisms, and we explore potential applications in probing extracellular vesicles released from sources such as apoptotic cells, cancer cells, or activated platelets. J. Cell. Physiol. 231: 2327-2332, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aaron D Flynn
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado.,BioFrontiers Institute, University of Colorado, Boulder, Colorado
| | - Hang Yin
- BioFrontiers Institute, University of Colorado, Boulder, Colorado.,Department of Chemistry and Biochemistry, University of Colorado, Boulder, Colorado
| |
Collapse
|
2106
|
Yang Q, Diamond MP, Al-Hendy A. Early Life Adverse Environmental Exposures Increase the Risk of Uterine Fibroid Development: Role of Epigenetic Regulation. Front Pharmacol 2016; 7:40. [PMID: 26973527 PMCID: PMC4772696 DOI: 10.3389/fphar.2016.00040] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/12/2016] [Indexed: 12/21/2022] Open
Abstract
Uterine Fibroids [UF(s), AKA: leiomyoma] are the most important benign neoplastic threat to women's health. They are the most common cause of hysterectomy imposing untold personal consequences and 100s of billions of healthcare dollars, worldwide. Currently, there is no long term effective FDA-approved medical treatment available, and surgery is the mainstay. The etiology of UFs is not fully understood. In this regard, we and others have recently reported that somatic mutations in the gene encoding the transcriptional mediator subunit Med12 are found to occur at a high frequency (∼85%) in UFs. UFs likely originate when a Med12 mutation occurs in a myometrial stem cell converting it into a tumor-forming stem cell leading to a clonal fibroid lesion. Although the molecular attributes underlying the mechanistic formation of UFs is largely unknown, a growing body of literature implicates unfavorable early life environmental exposures as potentially important contributors. Early life exposure to EDCs during sensitive windows of development can reprogram normal physiological responses and alter disease susceptibility later in life. Neonatal exposure to the EDCs such as diethylstilbestrol (DES) and genistein during reproductive tract development has been shown to increase the incidence, multiplicity and overall size of UFs in the Eker rat model, concomitantly reprogramming estrogen-responsive gene expression. Importantly, EDC exposure represses enhancer of zeste 2 (EZH2) and reduces levels of histone 3 lysine 27 trimethylation (H3K27me3) repressive mark through Estrogen receptor/Phosphatidylinositide 3-kinases/Protein kinase B non-genomic signaling in the developing uterus. Considering the fact that distinct Mediator Complex Subunit 12 (Med12) mutations are detected in different fibroid lesions in the same uterus, the emergence of each Med12 mutation is likely an independent event in an altered myometrial stem cell. It is therefore possible that a chronic reduction in DNA repair capacity eventually causes the emergence of mutations such as Med12 in myometrial stem cells converting them into fibroid tumor-forming stem cells, and thereby leads to the development of UFs. Advancing our understanding of the mechanistic role epigenetic regulation of stem cells plays in mediating risk and tumorigenesis will help in pointing the way toward the development of novel therapeutic options.
Collapse
Affiliation(s)
- Qiwei Yang
- Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta GA, USA
| | - Michael P Diamond
- Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta GA, USA
| | - Ayman Al-Hendy
- Division of Translation Research, Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta GA, USA
| |
Collapse
|
2107
|
Eskander MF, Bliss LA, Tseng JF. Pancreatic adenocarcinoma. Curr Probl Surg 2016; 53:107-54. [DOI: 10.1067/j.cpsurg.2016.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 01/04/2016] [Indexed: 12/17/2022]
|
2108
|
Abstract
Tumor-derived exosomes (TEX) are harbingers of tumor-induced immune suppression: they carry immunosuppressive molecules and factors known to interfere with immune cell functions. By delivering suppressive cargos consisting of proteins similar to those in parent tumor cells to immune cells, TEX directly or indirectly influence the development, maturation, and antitumor activities of immune cells. TEX also deliver genomic DNA, mRNA, and microRNAs to immune cells, thereby reprogramming functions of responder cells to promote tumor progression. TEX carrying tumor-associated antigens can interfere with antitumor immunotherapies. TEX also have the potential to serve as noninvasive biomarkers of tumor progression. In the tumor microenvironment, TEX may be involved in operating numerous signaling pathways responsible for the downregulation of antitumor immunity.
Collapse
|
2109
|
Jeong S, Park J, Pathania D, Castro CM, Weissleder R, Lee H. Integrated Magneto-Electrochemical Sensor for Exosome Analysis. ACS NANO 2016; 10:1802-9. [PMID: 26808216 PMCID: PMC4802494 DOI: 10.1021/acsnano.5b07584] [Citation(s) in RCA: 346] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Extracellular vesicles, including exosomes, are nanoscale membrane particles that carry molecular information on parental cells. They are being pursued as biomarkers of cancers that are difficult to detect or serially follow. Here we present a compact sensor technology for rapid, on-site exosome screening. The sensor is based on an integrated magneto-electrochemical assay: exosomes are immunomagnetically captured from patient samples and profiled through electrochemical reaction. By combining magnetic enrichment and enzymatic amplification, the approach enables (i) highly sensitive, cell-specific exosome detection and (ii) sensor miniaturization and scale-up for high-throughput measurements. As a proof-of-concept, we implemented a portable, eight-channel device and applied it to screen extracellular vesicles in plasma samples from ovarian cancer patients. The sensor allowed for the simultaneous profiling of multiple protein markers within an hour, outperforming conventional methods in assay sensitivity and speed.
Collapse
Affiliation(s)
- Sangmoo Jeong
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
| | - Jongmin Park
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
| | - Divya Pathania
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
| | - Cesar M. Castro
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
- Massachusetts General Hospital Cancer Center, Boston, MA 02114
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
- Massachusetts General Hospital Cancer Center, Boston, MA 02114
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
- Corresponding author: H. Lee, PhD, Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114, 617-726-8226,
| |
Collapse
|
2110
|
Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, Dingli F, Loew D, Tkach M, Théry C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A 2016; 113:E968-77. [PMID: 26858453 PMCID: PMC4776515 DOI: 10.1073/pnas.1521230113] [Citation(s) in RCA: 2530] [Impact Index Per Article: 281.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) have become the focus of rising interest because of their numerous functions in physiology and pathology. Cells release heterogeneous vesicles of different sizes and intracellular origins, including small EVs formed inside endosomal compartments (i.e., exosomes) and EVs of various sizes budding from the plasma membrane. Specific markers for the analysis and isolation of different EV populations are missing, imposing important limitations to understanding EV functions. Here, EVs from human dendritic cells were first separated by their sedimentation speed, and then either by their behavior upon upward floatation into iodixanol gradients or by immuno-isolation. Extensive quantitative proteomic analysis allowing comparison of the isolated populations showed that several classically used exosome markers, like major histocompatibility complex, flotillin, and heat-shock 70-kDa proteins, are similarly present in all EVs. We identified proteins specifically enriched in small EVs, and define a set of five protein categories displaying different relative abundance in distinct EV populations. We demonstrate the presence of exosomal and nonexosomal subpopulations within small EVs, and propose their differential separation by immuno-isolation using either CD63, CD81, or CD9. Our work thus provides guidelines to define subtypes of EVs for future functional studies.
Collapse
Affiliation(s)
- Joanna Kowal
- Institut Curie, PSL Research University, INSERM U932, Department "Immunité et Cancer", 75248 Paris, France
| | - Guillaume Arras
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de masse Protéomique, 75248 Paris, France
| | - Marina Colombo
- Institut Curie, PSL Research University, INSERM U932, Department "Immunité et Cancer", 75248 Paris, France
| | - Mabel Jouve
- Institut Curie, PSL Research University, INSERM U932, Department "Immunité et Cancer", 75248 Paris, France
| | - Jakob Paul Morath
- Institut Curie, PSL Research University, INSERM U932, Department "Immunité et Cancer", 75248 Paris, France
| | - Bjarke Primdal-Bengtson
- Institut Curie, PSL Research University, INSERM U932, Department "Immunité et Cancer", 75248 Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de masse Protéomique, 75248 Paris, France
| | - Damarys Loew
- Institut Curie, PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de masse Protéomique, 75248 Paris, France
| | - Mercedes Tkach
- Institut Curie, PSL Research University, INSERM U932, Department "Immunité et Cancer", 75248 Paris, France
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERM U932, Department "Immunité et Cancer", 75248 Paris, France;
| |
Collapse
|
2111
|
Lu L, Risch HA. Exosomes: potential for early detection in pancreatic cancer. Future Oncol 2016; 12:1081-90. [PMID: 26860951 DOI: 10.2217/fon-2015-0005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Progress in the treatment of patients with pancreatic cancer at earlier stages has motivated research in identifying novel noninvasive or minimally invasive biomarkers for early detection. Exosomes, which contain bioactive molecules (such as proteins, RNAs and lipids), are membrane-structured nanovesicles that are secreted from living cells and are found in human body fluids. As functional mediators, exosomes play key roles in cell-cell communications, regulating diverse biological processes. Here we aim to examine recent findings in the potential diagnostic value of serum exosomes in pancreatic cancer.
Collapse
Affiliation(s)
- Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale School of Medicine, Yale Cancer Center, New Haven, CT 06520-8034, USA
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale School of Medicine, Yale Cancer Center, New Haven, CT 06520-8034, USA
| |
Collapse
|
2112
|
Zhao Z, Yang Y, Zeng Y, He M. A microfluidic ExoSearch chip for multiplexed exosome detection towards blood-based ovarian cancer diagnosis. LAB ON A CHIP 2016; 16:489-96. [PMID: 26645590 PMCID: PMC4729647 DOI: 10.1039/c5lc01117e] [Citation(s) in RCA: 491] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Tumor-derived circulating exosomes, enriched with a group of tumor antigens, have been recognized as a promising biomarker source for cancer diagnosis via a less invasive procedure. Quantitatively pinpointing exosome tumor markers is appealing, yet challenging. In this study, we developed a simple microfluidic approach (ExoSearch) which provides enriched preparation of blood plasma exosomes for in situ, multiplexed detection using immunomagnetic beads. The ExoSearch chip offers a robust, continuous-flow design for quantitative isolation and release of blood plasma exosomes in a wide range of preparation volumes (10 μL to 10 mL). We employed the ExoSearch chip for blood-based diagnosis of ovarian cancer by multiplexed measurement of three exosomal tumor markers (CA-125, EpCAM, CD24) using a training set of ovarian cancer patient plasma, which showed significant diagnostic power (a.u.c. = 1.0, p = 0.001) and was comparable with the standard Bradford assay. This work provides an essentially needed platform for utilization of exosomes in clinical cancer diagnosis, as well as fundamental exosome research.
Collapse
Affiliation(s)
- Zheng Zhao
- Department of Biological and Agricultural Engineering, Kansas State University, Manhattan, KS 66506, USA. and Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA.
| | - Yang Yang
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA.
| | - Yong Zeng
- Department of Chemistry, University of Kansas, Lawrence, KS 66045, USA. and University of Kansas Cancer Center, Kansas City, KS 66160, USA
| | - Mei He
- Department of Biological and Agricultural Engineering, Kansas State University, Manhattan, KS 66506, USA. and Terry C. Johnson Cancer Research Center, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
2113
|
Abstract
The majority of therapeutics target membrane proteins, accessible on the surface of cells, to alter cellular signaling. Cells use membrane proteins to transduce signals into cells, transport ions and molecules, bind cells to a surface or substrate, and catalyze reactions. Newly devised technologies allow us to drug conventionally "undruggable" regions of membrane proteins, enabling modulation of protein-protein, protein-lipid, and protein-nucleic acid interactions. In this review, we survey the state of the art of high-throughput screening and rational design in drug discovery, and we evaluate the advances in biological understanding and technological capacity that will drive pharmacotherapy forward against unorthodox membrane protein targets.
Collapse
Affiliation(s)
- Hang Yin
- Department of Chemistry and Biochemistry.,BioFrontiers Institute, and.,Center of Basic Molecular Science, Department of Chemistry, Tsinghua University, Beijing 100082, China
| | - Aaron D Flynn
- BioFrontiers Institute, and.,Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Colorado 80309; ,
| |
Collapse
|
2114
|
Shang Y, Zeng Y, Zeng Y. Integrated Microfluidic Lectin Barcode Platform for High-Performance Focused Glycomic Profiling. Sci Rep 2016; 6:20297. [PMID: 26831207 PMCID: PMC4735825 DOI: 10.1038/srep20297] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/30/2015] [Indexed: 11/29/2022] Open
Abstract
Protein glycosylation is one of the key processes that play essential roles in biological functions and dysfunctions. However, progress in glycomics has considerably lagged behind genomics and proteomics, due in part to the enormous challenges in analysis of glycans. Here we present a new integrated and automated microfluidic lectin barcode platform to substantially improve the performance of lectin array for focused glycomic profiling. The chip design and flow control were optimized to promote the lectin-glycan binding kinetics and speed of lectin microarray. Moreover, we established an on-chip lectin assay which employs a very simple blocking method to effectively suppress the undesired background due to lectin binding of antibodies. Using this technology, we demonstrated focused differential profiling of tissue-specific glycosylation changes of a biomarker, CA125 protein purified from ovarian cancer cell line and different tissues from ovarian cancer patients in a fast, reproducible, and high-throughput fashion. Highly sensitive CA125 detection was also demonstrated with a detection limit much lower than the clinical cutoff value for cancer diagnosis. This microfluidic platform holds the potential to integrate with sample preparation functions to construct a fully integrated "sample-to-answer" microsystem for focused differential glycomic analysis. Thus, our technology should present a powerful tool in support of rapid advance in glycobiology and glyco-biomarker development.
Collapse
Affiliation(s)
- Yuqin Shang
- Department of Chemistry, University of Kansas, Lawrence, KS 66045
| | - Yun Zeng
- College of Water Resource and Hydropower, Sichuan Agricultural University, Ya’an, Sichuan 625014, P.R. China
| | - Yong Zeng
- Department of Chemistry, University of Kansas, Lawrence, KS 66045
- University of Kansas Cancer Center, Kansas City, KS 66160
| |
Collapse
|
2115
|
Extracellular matrix component signaling in cancer. Adv Drug Deliv Rev 2016; 97:28-40. [PMID: 26519775 DOI: 10.1016/j.addr.2015.10.013] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
Abstract
Cell responses to the extracellular matrix depend on specific signaling events. These are important from early development, through differentiation and tissue homeostasis, immune surveillance, and disease pathogenesis. Signaling not only regulates cell adhesion cytoskeletal organization and motility but also provides survival and proliferation cues. The major classes of cell surface receptors for matrix macromolecules are the integrins, discoidin domain receptors, and transmembrane proteoglycans such as syndecans and CD44. Cells respond not only to specific ligands, such as collagen, fibronectin, or basement membrane glycoproteins, but also in terms of matrix rigidity. This can regulate the release and subsequent biological activity of matrix-bound growth factors, for example, transforming growth factor-β. In the environment of tumors, there may be changes in cell populations and their receptor profiles as well as matrix constitution and protein cross-linking. Here we summarize roles of the three major matrix receptor types, with emphasis on how they function in tumor progression.
Collapse
|
2116
|
Herreros-Villanueva M, Bujanda L. Glypican-1 in exosomes as biomarker for early detection of pancreatic cancer. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:64. [PMID: 27004211 PMCID: PMC4779771 DOI: 10.3978/j.issn.2305-5839.2015.10.39] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 10/21/2015] [Indexed: 12/19/2022]
Abstract
On June 24, 2015 Nature published an article entitle "Glypican-1 identifies cancer exosomes and detects early pancreatic cancer'', which demonstrates that exosomes positives for the proteoglycan glypican-1 (GPC1) are expressed in serum of patients with pancreatic cancer since very early stages but not in benign pancreatic disease. Additionally, these GPC1(+) circulating exosomes correlate with tumor burden and could be used as prognostic biomarker in pre and post-surgical patients. The study is pioneer since GPC1 biomarker in exosomes offers better sensitivity and specificity than any other under evaluation or used in clinical practice. However, methodology for exosomes isolation still remains at investigational phase. Further studies are need to translate this technology to a practicable clinical method. Further research is also required to validate this biomarker in larger prospective cohort including more cases of premalignant lesions and then replicated results could possible guide changes in clinical practice.
Collapse
Affiliation(s)
- Marta Herreros-Villanueva
- Department of Gastroenterology, Hospital Donostia/Biodonostia Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco UPV/EHU, San Sebastian, Spain
| | - Luis Bujanda
- Department of Gastroenterology, Hospital Donostia/Biodonostia Institute, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco UPV/EHU, San Sebastian, Spain
| |
Collapse
|
2117
|
Seufferlein T, Mayerle J. Pancreatic cancer in 2015: Precision medicine in pancreatic cancer--fact or fiction? Nat Rev Gastroenterol Hepatol 2016; 13:74-5. [PMID: 26758788 DOI: 10.1038/nrgastro.2015.215] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Late diagnosis and an inability to personalize treatment are major problems preventing reductions in pancreatic cancer mortality. In 2015, the identification of a highly discriminatory exosomal biomarker, culture systems that recapitulate human disease and new methods of analysing large data sets to identify prognostic markers have improved the future outlook for patients with this cancer.
Collapse
Affiliation(s)
- Thomas Seufferlein
- Ulm University Medical Center, Department of Internal Medicine I, Albert Einstein Allee 23, D-89081 Ulm, Germany
| | - Julia Mayerle
- Department of Medicine A, University Medicine, Ernst-Moritz-Arndt-University, Greifswald, Ferdinand-Sauerbruchstrasse, 17475 Greifswald, Germany
| |
Collapse
|
2118
|
Affiliation(s)
- Klaus Pantel
- Institute of Tumor Biology, University Cancer Center Hamburg, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), Department of Cellular and Tissular Biopathology of Tumors, University Medical Centre, Montpellier, France; EA2415 - Help for Personalized Decision: Methodological Aspects, University Institute of Clinical Research (IURC), Montpellier University, Montpellier, France
| |
Collapse
|
2119
|
Kanada M, Bachmann MH, Contag CH. Signaling by Extracellular Vesicles Advances Cancer Hallmarks. Trends Cancer 2016; 2:84-94. [DOI: 10.1016/j.trecan.2015.12.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 12/21/2022]
|
2120
|
Guo J, Xie K, Zheng S. Molecular Biomarkers of Pancreatic Intraepithelial Neoplasia and Their Implications in Early Diagnosis and Therapeutic Intervention of Pancreatic Cancer. Int J Biol Sci 2016; 12:292-301. [PMID: 26929736 PMCID: PMC4753158 DOI: 10.7150/ijbs.14995] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Lack of early detection and effective interventions is a major reason for the poor prognosis and dismal survival rates for pancreatic cancer. Pancreatic intraepithelial neoplasia (PanIN) is the most common precursor of invasive pancreatic ductal adenocarcinoma (PDAC). Each stage in the progression from PanIN to PDAC is well characterized by multiple significant genetic alterations affecting signaling pathways. Understanding the biological behavior and molecular alterations in the progression from PanIN to PDAC is crucial to the identification of noninvasive biomarkers for early detection and diagnosis and the development of preventive and therapeutic strategies for control of pancreatic cancer progression. This review focuses on molecular biomarkers of PanIN and their important roles in early detection and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Junli Guo
- 1. Department of Pathology, Affiliated Hospital of Hainan Medical College, Hainan Cancer Hospital, Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Haikou 571199, People's Republic of China; 2. Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Keping Xie
- 2. Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shaojiang Zheng
- 1. Department of Pathology, Affiliated Hospital of Hainan Medical College, Hainan Cancer Hospital, Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Haikou 571199, People's Republic of China; 2. Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
2121
|
Amodu LI, Levy A, Kyaw C, Padmanabhan D, Osman A, Tiwari M, Nicastro J, Coppa G, Molmenti E, Rodriguez Rilo HL. Utility of Exosomes in the Diagnosis and Treatment of Pancreatic Adenocarcinoma. EUROPEAN MEDICAL JOURNAL 2016. [DOI: 10.33590/emj/10313200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Pancreatic cancer is the most common lethal cancer, with annual incidence and mortality rates being approximately equal. This dismal prognosis can be attributed to late diagnosis making the cancers unresectable. These cancers respond poorly to chemotherapy and radiation, and surgical resection remains the most effective treatment available. Diagnostic tests that are sensitive, specific, and capable of early detection are urgently needed and would significantly impact upon pancreatic cancer treatment and outcomes. Exosomes, small membrane-bound vesicles which are fairly uniform in size (approximately 30–100 nm in diameter), contain messenger RNA, microRNA (miRNA), and proteins. They are ubiquitous and stable in most body fluids and exosomal miRNAs are also resistant to degradation by RNAses and DNAses. Expression profiles of serum exosomal miRNAs display sensitivity and specificity in the detection of pancreatic adenocarcinoma. Markers of pancreatic cancer-initiating cells are also expressed on serum exosomes. Exosomes exhibit key functions in addition to their distinct structural properties: they are involved in immune system modulation via the transfer of antigenic proteins, and through protease activity they modulate the extracellular environment prior to metastasis. Exosomes are being studied as potent gene delivery tools and dendritic cell exosomes are already used as cancer vaccines. This review focusses on the current state of exosomal research, particularly in relation to their applicability as diagnostic and therapeutic tools for patients with pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Leo I. Amodu
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| | - Asaph Levy
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| | - Crystal Kyaw
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| | - Darshan Padmanabhan
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| | - Alexandra Osman
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| | - Mukesh Tiwari
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| | - Jeffrey Nicastro
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| | - Gene Coppa
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| | - Ernesto Molmenti
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| | - Horacio L. Rodriguez Rilo
- Center for Diseases of the Pancreas, Department of General Surgery, Hofstra North Shore-Long Island Jewish School of Medicine, Hempstead, New York, USA
| |
Collapse
|
2122
|
Berrondo C, Flax J, Kucherov V, Siebert A, Osinski T, Rosenberg A, Fucile C, Richheimer S, Beckham CJ. Expression of the Long Non-Coding RNA HOTAIR Correlates with Disease Progression in Bladder Cancer and Is Contained in Bladder Cancer Patient Urinary Exosomes. PLoS One 2016; 11:e0147236. [PMID: 26800519 PMCID: PMC4723257 DOI: 10.1371/journal.pone.0147236] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/30/2015] [Indexed: 12/14/2022] Open
Abstract
Exosomes are 30-150nM membrane-bound secreted vesicles that are readily isolated from biological fluids such as urine (UEs). Exosomes contain proteins, micro RNA (miRNA), messenger RNA (mRNA), and long non-coding RNA (lncRNA) from their cells of origin. Although miRNA, protein and lncRNA have been isolated from serum as potential biomarkers for benign and malignant disease, it is unknown if lncRNAs in UEs from urothelial bladder cancer (UBC) patients can serve as biomarkers. lncRNAs are > 200 nucleotide long transcripts that do not encode protein and play critical roles in tumor biology. As the number of recognized tumor-associated lncRNAs continues to increase, there is a parallel need to include lncRNAs into biomarker discovery and therapeutic target algorithms. The lncRNA HOX transcript antisense RNA (HOTAIR) has been shown to facilitate tumor initiation and progression and is associated with poor prognosis in several cancers. The importance of HOTAIR in cancer biology has sparked interest in using HOTAIR as a biomarker and potential therapeutic target. Here we show HOTAIR and several tumor-associated lncRNAs are enriched in UEs from UBC patients with high-grade muscle-invasive disease (HGMI pT2-pT4). Knockdown of HOTAIR in UBC cell lines reduces in vitro migration and invasion. Importantly, loss of HOTAIR expression in UBC cell lines alters expression of epithelial-to-mesenchyme transition (EMT) genes including SNAI1, TWIST1, ZEB1, ZO1, MMP1 LAMB3, and LAMC2. Finally, we used RNA-sequencing to identify four additional lncRNAs enriched in UBC patient UEs. These data, suggest that UE-derived lncRNA may potentially serve as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Claudia Berrondo
- University of Rochester Department of Urology, Strong Memorial Hospital Rochester, New York, United States of America
| | - Jonathan Flax
- University of Rochester Department of Urology, Strong Memorial Hospital Rochester, New York, United States of America
| | - Victor Kucherov
- University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Aisha Siebert
- University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Thomas Osinski
- University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Alex Rosenberg
- Division of Immunology and Rheumatology, University of Rochester, Strong Memorial Hospital Rochester, New York, United States of America
| | - Christopher Fucile
- Division of Immunology and Rheumatology, University of Rochester, Strong Memorial Hospital Rochester, New York, United States of America
| | - Samuel Richheimer
- University of Rochester Department of Urology, Strong Memorial Hospital Rochester, New York, United States of America
| | - Carla J. Beckham
- University of Rochester Department of Urology, Strong Memorial Hospital Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
2123
|
Berrondo C, Flax J, Kucherov V, Siebert A, Osinski T, Rosenberg A, Fucile C, Richheimer S, Beckham CJ. Expression of the Long Non-Coding RNA HOTAIR Correlates with Disease Progression in Bladder Cancer and Is Contained in Bladder Cancer Patient Urinary Exosomes. PLoS One 2016. [PMID: 26800519 DOI: 10.1371/journal.pone.0147236.ecollection2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Exosomes are 30-150nM membrane-bound secreted vesicles that are readily isolated from biological fluids such as urine (UEs). Exosomes contain proteins, micro RNA (miRNA), messenger RNA (mRNA), and long non-coding RNA (lncRNA) from their cells of origin. Although miRNA, protein and lncRNA have been isolated from serum as potential biomarkers for benign and malignant disease, it is unknown if lncRNAs in UEs from urothelial bladder cancer (UBC) patients can serve as biomarkers. lncRNAs are > 200 nucleotide long transcripts that do not encode protein and play critical roles in tumor biology. As the number of recognized tumor-associated lncRNAs continues to increase, there is a parallel need to include lncRNAs into biomarker discovery and therapeutic target algorithms. The lncRNA HOX transcript antisense RNA (HOTAIR) has been shown to facilitate tumor initiation and progression and is associated with poor prognosis in several cancers. The importance of HOTAIR in cancer biology has sparked interest in using HOTAIR as a biomarker and potential therapeutic target. Here we show HOTAIR and several tumor-associated lncRNAs are enriched in UEs from UBC patients with high-grade muscle-invasive disease (HGMI pT2-pT4). Knockdown of HOTAIR in UBC cell lines reduces in vitro migration and invasion. Importantly, loss of HOTAIR expression in UBC cell lines alters expression of epithelial-to-mesenchyme transition (EMT) genes including SNAI1, TWIST1, ZEB1, ZO1, MMP1 LAMB3, and LAMC2. Finally, we used RNA-sequencing to identify four additional lncRNAs enriched in UBC patient UEs. These data, suggest that UE-derived lncRNA may potentially serve as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Claudia Berrondo
- University of Rochester Department of Urology, Strong Memorial Hospital Rochester, New York, United States of America
| | - Jonathan Flax
- University of Rochester Department of Urology, Strong Memorial Hospital Rochester, New York, United States of America
| | - Victor Kucherov
- University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Aisha Siebert
- University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Thomas Osinski
- University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Alex Rosenberg
- Division of Immunology and Rheumatology, University of Rochester, Strong Memorial Hospital Rochester, New York, United States of America
| | - Christopher Fucile
- Division of Immunology and Rheumatology, University of Rochester, Strong Memorial Hospital Rochester, New York, United States of America
| | - Samuel Richheimer
- University of Rochester Department of Urology, Strong Memorial Hospital Rochester, New York, United States of America
| | - Carla J Beckham
- University of Rochester Department of Urology, Strong Memorial Hospital Rochester, New York, United States of America
| |
Collapse
|
2124
|
Hah N, Sherman MH, Yu RT, Downes M, Evans RM. Targeting Transcriptional and Epigenetic Reprogramming in Stromal Cells in Fibrosis and Cancer. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2016; 80:249-55. [PMID: 26801159 DOI: 10.1101/sqb.2015.80.027185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The basis of many human diseases arises from both genetic and epigenetic regulation. Recent advances in the understanding of the mechanisms underlying transcriptional and epigenetic regulation and their prevalence as contributors to a diverse range of human diseases have led us to focus on transcription and epigenetic changes in a variety of human disease conditions. Specifically, our recent studies in liver fibrosis and pancreatic cancer have demonstrated that the epigenetic regulation in hepatic stellate cells (HSCs) and pancreatic stellate cells (PSCs) significantly contributes to the progress in such diseases and presents great therapeutic potential. We show that the vitamin D receptor (VDR) acts as a master genomic suppressor in both HSC and PSC activation. The studies also have demonstrated that the VDR ligand reduces fibrosis and inflammation in a murine liver fibrosis and pancreatitis model. Although our current studies focus on characterizing the roles of VDR and regulatory regions within gene promoters and regulatory enhancers, we have expanded our effort to epigenetic mechanisms as major determinants of gene activation and repression in order to develop potential therapeutics to modulate stroma-associated pathologies including inflammation, fibrosis, and cancer.
Collapse
Affiliation(s)
- Nasun Hah
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Mara H Sherman
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Ruth T Yu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Michael Downes
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Ronald M Evans
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037 Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, California 92037
| |
Collapse
|
2125
|
Purushothaman A, Bandari SK, Liu J, Mobley JA, Brown EE, Sanderson RD. Fibronectin on the Surface of Myeloma Cell-derived Exosomes Mediates Exosome-Cell Interactions. J Biol Chem 2016; 291:1652-1663. [PMID: 26601950 PMCID: PMC4722448 DOI: 10.1074/jbc.m115.686295] [Citation(s) in RCA: 238] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/11/2015] [Indexed: 12/12/2022] Open
Abstract
Exosomes regulate cell behavior by binding to and delivering their cargo to target cells; however, the mechanisms mediating exosome-cell interactions are poorly understood. Heparan sulfates on target cell surfaces can act as receptors for exosome uptake, but the ligand for heparan sulfate on exosomes has not been identified. Using exosomes isolated from myeloma cell lines and from myeloma patients, we identify exosomal fibronectin as a key heparan sulfate-binding ligand and mediator of exosome-cell interactions. We discovered that heparan sulfate plays a dual role in exosome-cell interaction; heparan sulfate on exosomes captures fibronectin, and on target cells it acts as a receptor for fibronectin. Removal of heparan sulfate from the exosome surface releases fibronectin and dramatically inhibits exosome-target cell interaction. Antibody specific for the Hep-II heparin-binding domain of fibronectin blocks exosome interaction with tumor cells or with marrow stromal cells. Regarding exosome function, fibronectin-mediated binding of exosomes to myeloma cells activated p38 and pERK signaling and expression of downstream target genes DKK1 and MMP-9, two molecules that promote myeloma progression. Antibody against fibronectin inhibited the ability of myeloma-derived exosomes to stimulate endothelial cell invasion. Heparin or heparin mimetics including Roneparstat, a modified heparin in phase I trials in myeloma patients, significantly inhibited exosome-cell interactions. These studies provide the first evidence that fibronectin binding to heparan sulfate mediates exosome-cell interactions, revealing a fundamental mechanism important for exosome-mediated cross-talk within tumor microenvironments. Moreover, these results imply that therapeutic disruption of fibronectin-heparan sulfate interactions will negatively impact myeloma tumor growth and progression.
Collapse
Affiliation(s)
- Anurag Purushothaman
- From the Departments of Pathology and; University of Alabama at Birmingham Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294 and.
| | | | - Jian Liu
- the Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599
| | | | - Elizabeth E Brown
- From the Departments of Pathology and; University of Alabama at Birmingham Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294 and
| | - Ralph D Sanderson
- From the Departments of Pathology and; University of Alabama at Birmingham Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294 and.
| |
Collapse
|
2126
|
Ko J, Carpenter E, Issadore D. Detection and isolation of circulating exosomes and microvesicles for cancer monitoring and diagnostics using micro-/nano-based devices. Analyst 2016; 141:450-460. [PMID: 26378496 PMCID: PMC4881422 DOI: 10.1039/c5an01610j] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the last several years, nanoscale vesicles that originate from tumor cells and which can be found circulating in the blood (i.e. exosomes and microvesicles) have been discovered to contain a wealth of proteomic and genetic information to monitor cancer progression, metastasis, and drug efficacy. However, the use of exosomes and microvesicles as biomarkers to improve patient care has been limited by their small size (30 nm-1 μm) and the extensive sample preparation required for their isolation and measurement. In this Critical Review, we explore the emerging use of micro and nano-technology to isolate and detect exosomes and microvesicles in clinical samples and the application of this technology to the monitoring and diagnosis of cancer.
Collapse
Affiliation(s)
- Jina Ko
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erica Carpenter
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Issadore
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Electrical and Systems engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2127
|
Lodillinsky C, Podsypanina K, Chavrier P. Social networking in tumor cell communities is associated with increased aggressiveness. INTRAVITAL 2016; 5:e1112476. [PMID: 28243516 DOI: 10.1080/21659087.2015.1112476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/21/2015] [Accepted: 10/21/2015] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs) are lipid-bilayer-enclosed vesicles that contain proteins, lipids and nucleic acids. EVs produced by cells from healthy tissues circulate in the blood and body fluids, and can be taken up by unrelated cells. As they have the capacity to transfer cargo proteins, lipids and nucleic acids (mostly mRNAs and miRNAs) between different cells in the body, EVs are emerging as mediators of intercellular communication that could modulate cell behavior, tissue homeostasis and regulation of physiological functions. EV-mediated cell-cell communications are also proposed to play a role in disease, for example, cancer, where they could contribute to transfer of traits required for tumor progression and metastasis. However, direct evidence for EV-mediated mRNA transfer to individual cells and for its biological consequences in vivo has been missing until recently. Recent studies have reported elegant experiments using genetic tracing with the Cre recombinase system and intravital imaging that visualize and quantify functional transfer of mRNA mediated by EVs in the context of cancer and metastasis.
Collapse
Affiliation(s)
- Catalina Lodillinsky
- Institut Curie, PSL Research University, Paris, France; CNRS UMR 144, Membrane and Cytoskeleton Dynamics Team, Paris, France
| | - Katrina Podsypanina
- Institut Curie, PSL Research University, Paris, France; CNRS UMR 144, Membrane and Cytoskeleton Dynamics Team, Paris, France
| | - Philippe Chavrier
- Institut Curie, PSL Research University, Paris, France; CNRS UMR 144, Membrane and Cytoskeleton Dynamics Team, Paris, France
| |
Collapse
|
2128
|
Pancreatic cancer: Current research and future directions. Biochim Biophys Acta Rev Cancer 2016; 1865:123-32. [PMID: 26794394 DOI: 10.1016/j.bbcan.2016.01.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/27/2015] [Accepted: 01/09/2016] [Indexed: 12/13/2022]
Abstract
Despite the survival rate advancements in different types of cancer in the last 40 years, the perspective for pancreatic cancer patients has seen no substantial changes. Indeed, the five year survival rate remains around 5%. Nevertheless, in the last decade we have witnessed an increased interest in pancreatic cancer biology and this has produced a substantial increment in our knowledge on pancreatic cancer progression. The big challenge is now to translate this knowledge in better outcomes for patients. The aim of this review is to describe the latest discoveries and advancements in pancreatic cancer research and to discuss future directions.
Collapse
|
2129
|
Pocsfalvi G, Stanly C, Fiume I, Vékey K. Chromatography and its hyphenation to mass spectrometry for extracellular vesicle analysis. J Chromatogr A 2016; 1439:26-41. [PMID: 26830636 DOI: 10.1016/j.chroma.2016.01.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs), such as exosomes, microvesicles and apoptotic bodies are released by cells, both under physiological and pathological conditions. EVs can participate in a novel type of intercellular communication and deliver cargo of nucleic acids, proteins and lipids near or to distant host cells. EV research is proceeding at a fast pace; now they start to appear as promising therapeutic targets, diagnostic tools and drug delivery systems. Isolation and analysis of EVs are prerequisites for understanding their biological roles and for their clinical exploitation. In this process chromatography and mass spectrometry (MS)-based strategies are rapidly gaining importance; and are reviewed in the present communication. Isolation and purification of EVs is mostly performed by ultracentrifugation at present. Chromatography-based strategies are gaining ground, among which affinity and size exclusion chromatography (SEC) are particularly strong contenders. Their major advantages are the relative simplicity, robustness and throughput. Affinity chromatography has the added advantage of separating EV subtypes based on molecular recognition of EV surface motifs. SEC has the advantage that isolated EVs may retain their biological activity. EVs are typically isolated in small amounts, therefore high sensitivity is required for their analysis. Study of the molecular content of EVs (all compounds beside nucleic acids) is predominantly based on liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis. The chromatographic separation is mostly performed by reverse phase, nanoscale, ultra high performance LC technique. The MS analysis relying typically on nano-electrospray ionization MS/MS provides high sensitivity, selectivity and resolution, so that thousand(s) of proteins can be detected/identified/quantified in a EV sample. Beside protein identification, quantitation and characterization of protein post-translational modifications (PTMs), like glycosylation and phosphorylation are becoming feasible and increasingly important. Along with conventional LC-MS/MS, other chromatographic approaches hyphenated to MS are gaining importance for EV characterization. Hydrophilic interaction LC is used to characterize PTMs; LC-inductively coupled plasma/MS to identify metal containing molecules; while gas chromatography-MS to analyze some lipids and metabolites.
Collapse
Affiliation(s)
- Gabriella Pocsfalvi
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy.
| | - Christopher Stanly
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy
| | - Immacolata Fiume
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy
| | - Károly Vékey
- Mass Spectrometry Proteomics Group, Institute of Organic Chemistry, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
2130
|
Patel GK, Patton MC, Singh S, Khushman M, Singh AP. Pancreatic Cancer Exosomes: Shedding Off for a Meaningful Journey. ACTA ACUST UNITED AC 2016; 6:e148. [PMID: 27030812 DOI: 10.4172/2165-7092.1000e148] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
2131
|
Zhu SK. Role of precision medicine in pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2016; 24:4752. [DOI: 10.11569/wcjd.v24.i36.4752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
2132
|
Yang H, Fu H, Xu W, Zhang X. Exosomal non-coding RNAs: a promising cancer biomarker. ACTA ACUST UNITED AC 2016; 54:1871-1879. [DOI: 10.1515/cclm-2016-0029] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/18/2016] [Indexed: 12/13/2022]
Abstract
AbstractNovel and non-invasive biomarkers are urgently needed for early detection of cancer. Exosomes are nano-sized particles released by cells and contain various bioactive molecules including proteins, DNA, mRNAs, and non-coding RNAs. Increasing evidence suggests that exosomes play critical roles in tumorigenesis, tumor growth, metastasis, and therapy resistance. Exosomes could be readily accessible in nearly all the body fluids. The altered production of exosomes and aberrant expression of exosomal contents could reflect the pathological state of the body, indicating that exosomes and exosomal contents can be utilized as novel cancer biomarkers. Herein, we review the basic properties of exosomes, the functional roles of exosomes in cancer, and the methods of detecting exosomes and exosomal contents. In particular, we highlight the clinical values of exosomal non-coding RNAs in cancer diagnosis and prognosis.
Collapse
|
2133
|
Yuan MJ, Maghsoudi T, Wang T. Exosomes Mediate the Intercellular Communication after Myocardial Infarction. Int J Med Sci 2016; 13:113-6. [PMID: 26941569 PMCID: PMC4764777 DOI: 10.7150/ijms.14112] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/10/2015] [Indexed: 01/08/2023] Open
Abstract
The mechanisms of cardiac repair after myocardial infarction (MI) are complicated and not well-understood currently. It is known that exosomes are released from most cells, recognized as new candidates with important roles in intercellular and tissue-level communication. Cells can package proteins and RNA messages into exosome and secret to recipient cells, which regulate gene expression in recipient cells. The research on exosomes in cardiovascular disease is just emerging. It is well-known that exosomes from cardiomyocyte can transfect endothelial cells, stem cells, fibroblasts and smooth muscle cells to induce cellular changes. After myocardial infarction (MI), the exosomes play important roles in local and distant microcommunication. Nowadays, exosomal microRNAs transportation has been found to deliver signals to mediate cardiac repair after MI. However, the exosomes quality and quantities are variable under different pathological conditions. Therefore, we speculate that the monitoring of the quality and quantity of exosomes may serve as diagnosis and prognosis biomarkers of MI, and the study of exosomes will provide insights for the new therapeutics to cardiac remodeling after MI.
Collapse
Affiliation(s)
- Ming-Jie Yuan
- 1. Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China
| | | | - Tao Wang
- 2. Cardiovascular Research Center, University of Virginia, USA
| |
Collapse
|
2134
|
Abstract
Extracellular vesicles (ECV) are membrane compartments shed from all types of cells in various physiological and pathological states. In recent years, ECV have gained an increasing interest from the scientific community for their role as an intercellular communicator that plays important roles in modifying the tumor microenvironment. Multiple techniques have been established to collect ECV from conditioned media of cell culture or physiological fluids. The gold standard methodology is differential centrifugation. Although alternative techniques exist to collect ECV, these techniques have not proven suitable as a substitution for the ultracentrifugation procedure.
Collapse
Affiliation(s)
- Khalid Al-Nedawi
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, ON, Canada. .,Hamilton Centre for Kidney Research (HCKR), St. Joseph's Hospital, Hamilton, ON, Canada.
| | - Jolene Read
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, ON, Canada.,Hamilton Centre for Kidney Research (HCKR), St. Joseph's Hospital, Hamilton, ON, Canada
| |
Collapse
|
2135
|
Carrière J, Barnich N, Nguyen HTT. Exosomes: From Functions in Host-Pathogen Interactions and Immunity to Diagnostic and Therapeutic Opportunities. Rev Physiol Biochem Pharmacol 2016; 172:39-75. [PMID: 27600934 DOI: 10.1007/112_2016_7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since their first description in the 1980s, exosomes, small endosomal-derived extracellular vesicles, have been involved in innate and adaptive immunity through modulating immune responses and mediating antigen presentation. Increasing evidence has reported the role of exosomes in host-pathogen interactions and particularly in the activation of antimicrobial immune responses. The growing interest concerning exosomes in infectious diseases, their accessibility in various body fluids, and their capacity to convey a rich content (e.g., proteins, lipids, and nucleic acids) to distant recipient cells led the scientific community to consider the use of exosomes as potential new diagnostic and therapeutic tools. In this review, we summarize current understandings of exosome biogenesis and their composition and highlight the function of exosomes as immunomodulators in pathological states such as in infectious disorders. The potential of using exosomes as diagnostic and therapeutic tools is also discussed.
Collapse
Affiliation(s)
- Jessica Carrière
- University of Clermont Auvergne, M2iSH, UMR 1071 INSERM/University of Auvergne, Clermont-Ferrand, 63001, France.,INRA USC 2018, Clermont-Ferrand, 63001, France
| | - Nicolas Barnich
- University of Clermont Auvergne, M2iSH, UMR 1071 INSERM/University of Auvergne, Clermont-Ferrand, 63001, France.,INRA USC 2018, Clermont-Ferrand, 63001, France
| | - Hang Thi Thu Nguyen
- University of Clermont Auvergne, M2iSH, UMR 1071 INSERM/University of Auvergne, Clermont-Ferrand, 63001, France. .,INRA USC 2018, Clermont-Ferrand, 63001, France.
| |
Collapse
|
2136
|
van der Pol E, Böing AN, Gool EL, Nieuwland R. Recent developments in the nomenclature, presence, isolation, detection and clinical impact of extracellular vesicles. J Thromb Haemost 2016; 14:48-56. [PMID: 26564379 DOI: 10.1111/jth.13190] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Indexed: 02/06/2023]
Abstract
The research field of extracellular vesicles (EVs), such as microparticles and exosomes, is growing exponentially. The goal of this review is to provide an overview of recent developments relevant to the readers of the Journal of Thrombosis and Haemostasis. We will discuss nomenclature, the presence of EVs in fluids, methods of isolation and detection, and emerging clinical implications. Although research on EVs has been performed within the ISTH for over a decade, most of the recent research on EVs has been brought together by the International Society on Extracellular Vesicles (ISEV). To achieve an overview of recent developments, the information provided in this review comes not only from publications, but also from latest meetings of the ISEV (April 2015, Washington, DC, USA), the International Society on Advancement of Cytometry (June 2015, Glasgow, UK), and the ISTH (June 2015, Toronto, Canada).
Collapse
Affiliation(s)
- E van der Pol
- Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - A N Böing
- Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - E L Gool
- Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - R Nieuwland
- Laboratory of Experimental Clinical Chemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
2137
|
Babic A, Wolpin BM. Circulating Exosomes in Pancreatic Cancer: Will They Succeed on the Long, Littered Road to Early Detection Marker? Clin Chem 2015; 62:307-9. [PMID: 26721295 DOI: 10.1373/clinchem.2015.246538] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 10/28/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, Boston, MA.
| |
Collapse
|
2138
|
Alegre E, Zubiri L, Perez-Gracia JL, González-Cao M, Soria L, Martín-Algarra S, González A. Circulating melanoma exosomes as diagnostic and prognosis biomarkers. Clin Chim Acta 2015; 454:28-32. [PMID: 26724367 DOI: 10.1016/j.cca.2015.12.031] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/11/2015] [Accepted: 12/21/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Malignant melanoma is an aggressive cancer with an increasing incidence. Exosomes are actively secreted microvesicles, whose characteristics reflect those of the cell they are originated in. The aim of this study was to identify and evaluate the presence of the melanoma biomarkers MIA, S100B and tyrosinase-related protein 2 (TYRP2) in exosomes and their potential clinical utility. METHODS Serum samples were obtained from stage IV melanoma patients, melanoma-free patients and healthy controls. Exosomes were precipitated and TYRP2, MIA and S100B concentrations were quantified in serum, exosomes, and exosome-free serum. RESULTS Both MIA and S100B were detected in exosomes and correlated significantly with serum concentrations (S100B: r=0.968; MIA: r=0.799; p<0.001). MIA and S100B concentrations in exosomes were significantly higher in melanoma patients than in healthy controls and disease-free patients. However, TYRP2 concentrations in exosomes did not differ between these three groups. ROC curves analysis rendered AUCs for MIA of 0.883 (p<0.01) and of 0.840 for S100B (p<0.01). Patients with exosome MIA concentration higher than 2.5 μg/L showed shorter median survival related to those with lower level (4 versus 11 months; p<0.05). CONCLUSIONS MIA and S100B can be detected in exosomes from melanoma patients and their quantification presents diagnostic and prognostic utility.
Collapse
Affiliation(s)
| | - Leyre Zubiri
- Department of Medical Oncology, University Clinic of Navarra, Spain
| | | | - María González-Cao
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Lourdes Soria
- Department of Medical Oncology, University Clinic of Navarra, Spain
| | | | - Alvaro González
- Laboratory of Biochemistry, University Clinic of Navarra, Spain.
| |
Collapse
|
2139
|
Mora EM, Álvarez-Cubela S, Oltra E. Biobanking of Exosomes in the Era of Precision Medicine: Are We There Yet? Int J Mol Sci 2015; 17:13. [PMID: 26712742 PMCID: PMC4730260 DOI: 10.3390/ijms17010013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 12/14/2015] [Accepted: 12/17/2015] [Indexed: 12/21/2022] Open
Abstract
The emerge of personalized medicine demands high-quality human biospecimens with appropriate clinical annotation, especially in complex diseases such as cancer, neurodegenerative, cardiovascular, and metabolic alterations in which specimen heterogeneity and individual responses often complicate the development of precision therapeutic programs. In the growing field of extracellular vesicles (EVs) research, exosomes (EXOs)--a particular type of EVs--have been proposed as an advantageous diagnostic tool, as effective delivery vehicles and as therapeutic targets. However, the lack of consensus on isolation methods and rigorous criteria to characterize them puts the term EXO into question at the time that might explain some of the controversial results found in the literature. A lack of response in the biobank network to warrant standard optimized procedures for the isolation, characterization, and storage of EXOs will undoubtedly lead to a waste of resources and failure. This review is aimed at highlighting the increasing importance of EXOs for the clinic, especially in the cancer field, and at summarizing the initiatives taken to improve current isolation procedures, classification criteria, and storage conditions of EXOs as an effort to identify technological demands that biobank platforms face for the incorporation of EXOs and other extracellular vesicle fractions as valuable biospecimens for research.
Collapse
Affiliation(s)
- Edna M Mora
- Department of Surgery, School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan 00936, Puerto Rico.
- University of Puerto Rico Comprehensive Cancer Center, San Juan 00936, Puerto Rico.
| | | | - Elisa Oltra
- Facultad de Medicina, Universidad Católica de Valencia "San Vicente Mártir", Valencia 46001, Spain.
- Instituto Valenciano de Patología (IVP) de la Universidad Católica de Valencia "San Vicente Mártir", Centro de Investigación Príncipe Felipe (CIPF), Valencia 46012, Spain.
| |
Collapse
|
2140
|
Linares R, Tan S, Gounou C, Arraud N, Brisson AR. High-speed centrifugation induces aggregation of extracellular vesicles. J Extracell Vesicles 2015; 4:29509. [PMID: 26700615 PMCID: PMC4689953 DOI: 10.3402/jev.v4.29509] [Citation(s) in RCA: 393] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/02/2015] [Accepted: 12/07/2015] [Indexed: 01/05/2023] Open
Abstract
Plasma and other body fluids contain cell-derived extracellular vesicles (EVs), which participate in physiopathological processes and have potential biomedical applications. In order to isolate, concentrate and purify EVs, high-speed centrifugation is often used. We show here, using electron microscopy, receptor-specific gold labelling and flow cytometry, that high-speed centrifugation induces the formation of EV aggregates composed of a mixture of EVs of various phenotypes and morphologies. The presence of aggregates made of EVs of different phenotypes may lead to erroneous interpretation concerning the existence of EVs harbouring surface antigens from different cell origins.
Collapse
Affiliation(s)
- Romain Linares
- Molecular Imaging and NanoBioTechnology, University of Bordeaux, Pessac, France
| | - Sisareuth Tan
- Molecular Imaging and NanoBioTechnology, University of Bordeaux, Pessac, France
| | - Céline Gounou
- Molecular Imaging and NanoBioTechnology, University of Bordeaux, Pessac, France
| | - Nicolas Arraud
- Molecular Imaging and NanoBioTechnology, University of Bordeaux, Pessac, France
| | - Alain R Brisson
- Molecular Imaging and NanoBioTechnology, University of Bordeaux, Pessac, France.,Institut Universitaire de France, Paris, France;
| |
Collapse
|
2141
|
Tang H, Partyka K, Hsueh P, Sinha JY, Kletter D, Zeh H, Huang Y, Brand RE, Haab BB. Glycans related to the CA19-9 antigen are elevated in distinct subsets of pancreatic cancers and improve diagnostic accuracy over CA19-9. Cell Mol Gastroenterol Hepatol 2015; 2:201-221.e15. [PMID: 26998508 PMCID: PMC4792034 DOI: 10.1016/j.jcmgh.2015.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS The CA19-9 antigen is the current best biomarker for pancreatic cancer, but it is not elevated in about 25% of pancreatic cancer patients at a cutoff that gives a 25% false-positive rate. We hypothesized that antigens related to the CA19-9 antigen, which is a glycan called sialyl-Lewis A (sLeA), are elevated in distinct subsets of pancreatic cancers. METHODS We profiled the levels of multiple glycans and mucin glycoforms in plasma from 200 subjects with either pancreatic cancer or benign pancreatic disease, and we validated selected findings in additional cohorts of 116 and 100 subjects, the latter run blinded and including cancers that exclusively were early-stage. RESULTS We found significant elevations in two glycans: an isomer of sLeA called sialyl-Lewis X, present both in sulfated and non-sulfated forms; and the sialylated form of a marker for pluripotent stem cells, type 1 N-acetyl-lactosamine. The glycans performed as well as sLeA as individual markers and were elevated in distinct groups of patients, resulting in a 3-marker panel that significantly improved upon any individual biomarker. The panel gave 85% sensitivity and 90% specificity in the combined discovery and validation cohorts, relative to 54% sensitivity and 86% specificity for sLeA; and it gave 80% sensitivity and 84% specificity in the independent test cohort, as opposed to 66% sensitivity and 72% specificity for sLeA. CONCLUSIONS Glycans related to sLeA are elevated in distinct subsets of pancreatic cancers and yield improved diagnostic accuracy over CA19-9.
Collapse
Affiliation(s)
- Huiyuan Tang
- Van Andel Research Institute, Center for Cancer and Cell Biology, Grand Rapids, Michigan
| | - Katie Partyka
- Van Andel Research Institute, Center for Cancer and Cell Biology, Grand Rapids, Michigan
| | - Peter Hsueh
- Van Andel Research Institute, Center for Cancer and Cell Biology, Grand Rapids, Michigan
| | - Jessica Y. Sinha
- Van Andel Research Institute, Center for Cancer and Cell Biology, Grand Rapids, Michigan
| | | | - Herbert Zeh
- University of Pittsburgh School of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Pittsburgh, Pennsylvania
| | - Ying Huang
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Disease Division, Seattle, Washington
| | - Randall E. Brand
- University of Pittsburgh School of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Pittsburgh, Pennsylvania
| | - Brian B. Haab
- Van Andel Research Institute, Center for Cancer and Cell Biology, Grand Rapids, Michigan,Correspondence Address correspondence to: Brian B. Haab, PhD, Van Andel Research Institute, 333 Bostwick NE, Grand Rapids, Michigan 49503. fax: (616) 234–5269.Van Andel Research Institute333 Bostwick NEGrand RapidsMichigan 49503
| |
Collapse
|
2142
|
Zhou L, Li Q, Wang J, Huang C, Nice EC. Oncoproteomics: Trials and tribulations. Proteomics Clin Appl 2015; 10:516-31. [PMID: 26518147 DOI: 10.1002/prca.201500081] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 09/19/2015] [Accepted: 10/27/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University, and Collaborative Innovation Center for Biotherapy; Chengdu P. R. China
- Department of Neurology; The Affiliated Hospital of Hainan Medical College; Haikou Hainan P. R. China
| | - Qifu Li
- Department of Neurology; The Affiliated Hospital of Hainan Medical College; Haikou Hainan P. R. China
| | - Jiandong Wang
- Department of Biomedical; Chengdu Medical College; Chengdu Sichuan Province P. R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University, and Collaborative Innovation Center for Biotherapy; Chengdu P. R. China
| | - Edouard C. Nice
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University, and Collaborative Innovation Center for Biotherapy; Chengdu P. R. China
- Department of Biochemistry and Molecular Biology; Monash University; Clayton Australia
| |
Collapse
|
2143
|
Alberter B, Klein CA, Polzer B. Single-cell analysis of CTCs with diagnostic precision: opportunities and challenges for personalized medicine. Expert Rev Mol Diagn 2015; 16:25-38. [PMID: 26567956 DOI: 10.1586/14737159.2016.1121099] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The generation of variant cancer cells is the major cause of acquired resistance against systemic therapies and consequently, of our inability to cure advanced cancer patients. Circulating tumor cells are gaining increasing clinical attention because they may enable the monitoring cancer progression and adjustment of treatment. In recent years multiple technologies for enrichment, isolation as well as molecular and functional analysis of circulating tumor cells have been developed. Implementation of these technologies in standardized and automated workflows in clinical diagnostics could provide valuable information for real-time monitoring of cancer and eventually new therapeutic strategies for the benefit of patients.
Collapse
Affiliation(s)
- Barbara Alberter
- a Project Group "Personalized Tumor Therapy" , Fraunhofer Institute for Toxicology and Experimental Medicine , Regensburg , Germany
| | - Christoph A Klein
- a Project Group "Personalized Tumor Therapy" , Fraunhofer Institute for Toxicology and Experimental Medicine , Regensburg , Germany.,b Experimental Medicine and Therapy Research , University of Regensburg , Regensburg , Germany
| | - Bernhard Polzer
- a Project Group "Personalized Tumor Therapy" , Fraunhofer Institute for Toxicology and Experimental Medicine , Regensburg , Germany
| |
Collapse
|
2144
|
Sundaresan TK, Haber DA. Fantastic voyage: the future of cancer diagnostics. Lancet Oncol 2015; 16:1596-8. [DOI: 10.1016/s1470-2045(15)00469-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 10/29/2015] [Indexed: 12/11/2022]
|
2145
|
Exosomes as novel biomarkers in anticancer therapy. MEMO-MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2015. [DOI: 10.1007/s12254-015-0245-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
2146
|
Roy BT, Roy RB. Metastatic pancreatic cancer: abdominal pain in a 22-year-old woman. BMJ Case Rep 2015; 2015:bcr-2015-211361. [PMID: 26611479 DOI: 10.1136/bcr-2015-211361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer remains fourth among cancer-related deaths. Its diagnosis is often missed, owing to its vague abdominal symptoms, leading to a diagnosis of stage IV pancreatic cancer. FOLFIRINOX chemotherapy was administered to the patient in this report, resulting in significant improvement based on tests of biological markers as well as radiographic resolution of the metastatic disease. A keen awareness of this condition along with appropriate diagnostic tests can allow for an earlier diagnosis and may impact its high mortality.
Collapse
Affiliation(s)
- Bakul T Roy
- Department of Internal Medicine, Roy Medical Associates, Inc, San Ramon, California, USA
| | - Roshan B Roy
- Roy Medical Associates, Inc, San Ramon, California, USA
| |
Collapse
|
2147
|
Joshi GK, Deitz-McElyea S, Liyanage T, Lawrence K, Mali S, Sardar R, Korc M. Label-Free Nanoplasmonic-Based Short Noncoding RNA Sensing at Attomolar Concentrations Allows for Quantitative and Highly Specific Assay of MicroRNA-10b in Biological Fluids and Circulating Exosomes. ACS NANO 2015; 9:11075-89. [PMID: 26444644 PMCID: PMC4660391 DOI: 10.1021/acsnano.5b04527] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/29/2015] [Indexed: 05/20/2023]
Abstract
MicroRNAs are short noncoding RNAs consisting of 18-25 nucleotides that target specific mRNA moieties for translational repression or degradation, thereby modulating numerous biological processes. Although microRNAs have the ability to behave like oncogenes or tumor suppressors in a cell-autonomous manner, their exact roles following release into the circulation are only now being unraveled and it is important to establish sensitive assays to measure their levels in different compartments in the circulation. Here, an ultrasensitive localized surface plasmon resonance (LSPR)-based microRNA sensor with single nucleotide specificity was developed using chemically synthesized gold nanoprisms attached onto a solid substrate with unprecedented long-term stability and reversibility. The sensor was used to specifically detect microRNA-10b at the attomolar (10(-18) M) concentration in pancreatic cancer cell lines, derived tissue culture media, human plasma, and media and plasma exosomes. In addition, for the first time, our label-free and nondestructive sensing technique was used to quantify microRNA-10b in highly purified exosomes isolated from patients with pancreatic cancer or chronic pancreatitis, and from normal controls. We show that microRNA-10b levels were significantly higher in plasma-derived exosomes from pancreatic ductal adenocarcinoma patients when compared with patients with chronic pancreatitis or normal controls. Our findings suggest that this unique technique can be used to design novel diagnostic strategies for pancreatic and other cancers based on the direct quantitative measurement of plasma and exosome microRNAs, and can be readily extended to other diseases with identifiable microRNA signatures.
Collapse
Affiliation(s)
- Gayatri K. Joshi
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis, 402 North Blackford Street, LD 326, Indianapolis, Indiana 46202, United States
| | - Samantha Deitz-McElyea
- Departments of Medicine, and Biochemistry and Molecular Biology, the Indiana University Simon Cancer Center, and the Pancreatic Cancer Signature Center, Indiana University School of Medicine, 980 West Walnut Street, R3-C528, Indianapolis, Indiana 46202, United States
| | - Thakshila Liyanage
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis, 402 North Blackford Street, LD 326, Indianapolis, Indiana 46202, United States
| | - Katie Lawrence
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis, 402 North Blackford Street, LD 326, Indianapolis, Indiana 46202, United States
| | - Sonali Mali
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis, 402 North Blackford Street, LD 326, Indianapolis, Indiana 46202, United States
| | - Rajesh Sardar
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis, 402 North Blackford Street, LD 326, Indianapolis, Indiana 46202, United States
- Integrated Nanosystems Development Institute, Indiana University-Purdue University Indianapolis, 402 North Blackford Street, Indianapolis, Indiana 46202, United States
- Address correspondence to ,
| | - Murray Korc
- Departments of Medicine, and Biochemistry and Molecular Biology, the Indiana University Simon Cancer Center, and the Pancreatic Cancer Signature Center, Indiana University School of Medicine, 980 West Walnut Street, R3-C528, Indianapolis, Indiana 46202, United States
- Address correspondence to ,
| |
Collapse
|
2148
|
Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, Wu CC, LeBleu VS, Kalluri R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015; 527:525-530. [PMID: 26560028 PMCID: PMC4849281 DOI: 10.1038/nature16064] [Citation(s) in RCA: 1629] [Impact Index Per Article: 162.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/08/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Xiaofeng Zheng
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Julienne L Carstens
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiha Kim
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Matthew Scheible
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Judith Kaye
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hikaru Sugimoto
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chia-Chin Wu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Valerie S LeBleu
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.,Department of Bioengineering, Rice University, Houston, Texas
| |
Collapse
|
2149
|
Abstract
Real-time monitoring of changes in cells or cell products released from malignant lesions into the blood has opened new diagnostic avenues ("liquid biopsy"). In this issue of Cancer Cell, Best and colleagues describe that tumor-associated blood platelets provide specific information on the location and molecular composition of the primary tumor.
Collapse
Affiliation(s)
- Simon A Joosse
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
2150
|
Brown SA. Building SuperModels: emerging patient avatars for use in precision and systems medicine. Front Physiol 2015; 6:318. [PMID: 26594179 PMCID: PMC4635220 DOI: 10.3389/fphys.2015.00318] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/20/2015] [Indexed: 12/19/2022] Open
Affiliation(s)
- Sherry-Ann Brown
- Division of Cardiovascular Diseases, Department of Medicine, Mayo ClinicRochester, MN, USA
| |
Collapse
|