101
|
Chutipongtanate S, Greis KD. Multiplex Biomarker Screening Assay for Urinary Extracellular Vesicles Study: A Targeted Label-Free Proteomic Approach. Sci Rep 2018; 8:15039. [PMID: 30301925 PMCID: PMC6177456 DOI: 10.1038/s41598-018-33280-7] [Citation(s) in RCA: 32] [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: 03/29/2018] [Accepted: 09/26/2018] [Indexed: 12/18/2022] Open
Abstract
The recent advance in targeted label-free proteomics, SWATH-MS, can provide consistent protein detection and reproducible protein quantitation, which is a considerable advantage for biomarker study of urinary extracellular vesicles. We developed a SWATH-MS workflow with a curated spectral library of 1,145 targets. Application of the workflow across nine replicates of three sample types (exosome-like vesicles (ELVs), microvesicles (MVs) and urine proteins (UPs)) resulted in the quantitation of 888 proteins at FDR <1%. The median-coefficient of variation of the 888 proteins in the ELV sample was 7.7%, indicating excellent reproducibility. Data analysis showed common exosome markers, (i.e. CD9, CD63, ALIX, TSG101 and HSP70) were enriched in urinary ELVs as compared to MVs and UPs. The use of a multiplex biomarker screening assay focused on ELVs was investigated, and perspectives in future applications are discussed.
Collapse
Affiliation(s)
- Somchai Chutipongtanate
- UC Proteomics Laboratory, Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States.
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | - Kenneth D Greis
- UC Proteomics Laboratory, Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States.
| |
Collapse
|
102
|
Liu YR, Ortiz-Bonilla CJ, Lee YF. Extracellular Vesicles in Bladder Cancer: Biomarkers and Beyond. Int J Mol Sci 2018; 19:E2822. [PMID: 30231589 PMCID: PMC6165150 DOI: 10.3390/ijms19092822] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 12/14/2022] Open
Abstract
Tumor-derived extracellular vesicles (TEVs) are membrane-bound, nanosized vesicles released by cancer cells and taken up by cells in the tumor microenvironment to modulate the molecular makeup and behavior of recipient cells. In this report, we summarize the pivotal roles of TEVs involved in bladder cancer (BC) development, progression and treatment resistance through transferring their bioactive cargos, including proteins and nucleic acids. We also report on the molecular profiling of TEV cargos derived from urine and blood of BC patients as non-invasive disease biomarkers. The current hurdles in EV research and plausible solutions are discussed.
Collapse
Affiliation(s)
- Yu-Ru Liu
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Carlos J Ortiz-Bonilla
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA.
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Yi-Fen Lee
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA.
- Department of Pathology and Lab Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
103
|
Liu Z, Cauvi DM, Bernardino EMA, Lara B, Lizardo RE, Hawisher D, Bickler S, De Maio A. Isolation and characterization of human urine extracellular vesicles. Cell Stress Chaperones 2018; 23:943-953. [PMID: 29796787 PMCID: PMC6111092 DOI: 10.1007/s12192-018-0902-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (ECV) reflect physiological or pathological conditions, emerging as potential biomarkers for disease. They can be obtained from a variety of body fluids, particularly urine that is an ideal source because it can be obtained in great quantities, recurrently and with minimal intervention. However, the characterization of urine ECV is challenging because the preparation is usually contaminated with soluble proteins, such as uromodulin (UMOD) or Tamm-Horsfall glycoprotein that forms large extracellular filaments co-sedimenting with ECV. We developed a method to obtain human urine ECV free of UMOD by the addition of ZnSO4 prior to vesicle isolation by differential centrifugation. Treatment with ZnSO4 did not affect the size and concentration of the vesicle preparation and preserved the storage of the samples at low temperatures. We did not observe a variation in the number of vesicles isolated during different times of the day or different days between different donors. The glycoprotein pattern of urine ECV was characterized by binding to concanavalin A (Con A) and mass spectroscopy. Several markers were found, including dipeptidyl peptidase IV (CD26), vacuolar protein sorting factor 4A (VPS4A) and dipeptidase 1 (DPEP1), and galectin 3 binding protein (G3-BP). The levels of VPS4A and DPEP1 were similar in ECV preparations obtained from several donors of both sexes. Con A binding pattern and monosaccharide composition were also comparable between subjects. In summary, our method for the isolation of highly pure ECV derived from human urine is likely to help in the use of these vesicles as potential biomarkers.
Collapse
Affiliation(s)
- Zhanguo Liu
- Department of Surgery, School of Medicine University of California San Diego, 9500 Gilman Drive, #0739, La Jolla, CA, 92093-0739, USA
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - David M Cauvi
- Department of Surgery, School of Medicine University of California San Diego, 9500 Gilman Drive, #0739, La Jolla, CA, 92093-0739, USA
| | - Erika M A Bernardino
- Department of Surgery, School of Medicine University of California San Diego, 9500 Gilman Drive, #0739, La Jolla, CA, 92093-0739, USA
| | - Bernardo Lara
- Department of Surgery, School of Medicine University of California San Diego, 9500 Gilman Drive, #0739, La Jolla, CA, 92093-0739, USA
| | | | - Dennis Hawisher
- Department of Surgery, School of Medicine University of California San Diego, 9500 Gilman Drive, #0739, La Jolla, CA, 92093-0739, USA
| | - Stephen Bickler
- Department of Surgery, School of Medicine University of California San Diego, 9500 Gilman Drive, #0739, La Jolla, CA, 92093-0739, USA
| | - Antonio De Maio
- Department of Surgery, School of Medicine University of California San Diego, 9500 Gilman Drive, #0739, La Jolla, CA, 92093-0739, USA.
- Department of Neurosciences, School of Medicine University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
104
|
Lee J, Kwon MH, Kim JA, Rhee WJ. Detection of exosome miRNAs using molecular beacons for diagnosing prostate cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S52-S63. [PMID: 30033809 DOI: 10.1080/21691401.2018.1489263] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Prostate cancer is the fifth leading cause of cancer-related deaths among males worldwide. However, the biomarker for diagnosing prostate cancer that is used currently has limitations that must be overcome. Recently, several studies have demonstrated that the cancer liquid biopsy can be implemented by using exosome miRNAs. However, the current methods for the detection of exosome miRNAs are time-consuming, expensive, and laborious. Thus, we investigated a novel method for diagnosing prostate cancer that involves the use of molecular beacons for the in situ detection of miRNAs in exosomes from prostate cancer cells. We chose miRNA-375 and miRNA-574-3p as the target miRNAs for prostate cancer, and these markers in exosomes produced by prostate cancer cells including DU145 and PC-3 were successfully detected using molecular beacons. High fluorescent signals were obtained from MB and miRNA hybridization in exosomes in a concentration-dependent manner. In addition, exosome miRNAs can be detected even in the presence of human urine, so this method can be applied directly using human urine to perform liquid biopsies for prostate cancer. Overall, the in situ detection of exosome miRNAs using molecular beacons can be developed as a simple, cost effective, and non-invasive liquid biopsy for diagnosing prostate cancer.
Collapse
Affiliation(s)
- Jinhee Lee
- a Division of Bioengineering , Incheon National University , Incheon , Republic of Korea
| | - Min Hee Kwon
- a Division of Bioengineering , Incheon National University , Incheon , Republic of Korea
| | - Jeong Ah Kim
- b Biomedical Omics Group , Korea Basic Science Institute , Cheongju , Republic of Korea
| | - Won Jong Rhee
- a Division of Bioengineering , Incheon National University , Incheon , Republic of Korea
| |
Collapse
|
105
|
Srivastava A, Moxley K, Ruskin R, Dhanasekaran DN, Zhao YD, Ramesh R. A Non-invasive Liquid Biopsy Screening of Urine-Derived Exosomes for miRNAs as Biomarkers in Endometrial Cancer Patients. AAPS J 2018. [PMID: 29987691 DOI: 10.1208/s12248-018-0220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Exosomes have great potential to serve as a source of diagnostic and prognostic biomarkers for endometrial cancer (EC). Urine-derived exosomes from patients with EC and patients with symptoms of EC, but without established EC, were used to evaluate a unique miRNA expression profile. Of the 84 miRNA studied, 57 were amplified in qPCR, suggesting the differential packaging of miRNA in exosomes. Further, hsa-miR-200c-3p was identified to be enriched the most. Various bioinformatics and in silico tools were used to evaluate the biological significance of hsa-miR-200c-3p in EC. We conclude that differential miRNA in exosomes can be utilized for discovery of biomarker signatures and EC diagnosis; hsa-miR-200c-3p is one such candidate. Urine-derived exosomes pave the way for the development of non-invasive biomarkers.
Collapse
Affiliation(s)
- Akhil Srivastava
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Katherine Moxley
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Department of Gynecology Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Rachel Ruskin
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Department of Gynecology Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Danny Natarajan Dhanasekaran
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Yan Daniel Zhao
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. .,Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. .,Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. .,Department of Pathology, Stanton L. Young Biomedical Research Center, Suite 1403, 975 N.E., 10th Street, Oklahoma City, Oklahoma, 73104, USA.
| |
Collapse
|
106
|
Srivastava A, Moxley K, Ruskin R, Dhanasekaran DN, Zhao YD, Ramesh R. A Non-invasive Liquid Biopsy Screening of Urine-Derived Exosomes for miRNAs as Biomarkers in Endometrial Cancer Patients. AAPS JOURNAL 2018; 20:82. [PMID: 29987691 DOI: 10.1208/s12248-018-0220-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Accepted: 03/16/2018] [Indexed: 12/18/2022]
Abstract
Exosomes have great potential to serve as a source of diagnostic and prognostic biomarkers for endometrial cancer (EC). Urine-derived exosomes from patients with EC and patients with symptoms of EC, but without established EC, were used to evaluate a unique miRNA expression profile. Of the 84 miRNA studied, 57 were amplified in qPCR, suggesting the differential packaging of miRNA in exosomes. Further, hsa-miR-200c-3p was identified to be enriched the most. Various bioinformatics and in silico tools were used to evaluate the biological significance of hsa-miR-200c-3p in EC. We conclude that differential miRNA in exosomes can be utilized for discovery of biomarker signatures and EC diagnosis; hsa-miR-200c-3p is one such candidate. Urine-derived exosomes pave the way for the development of non-invasive biomarkers.
Collapse
Affiliation(s)
- Akhil Srivastava
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Katherine Moxley
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Department of Gynecology Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Rachel Ruskin
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Department of Gynecology Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Danny Natarajan Dhanasekaran
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Yan Daniel Zhao
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.,Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. .,Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. .,Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA. .,Department of Pathology, Stanton L. Young Biomedical Research Center, Suite 1403, 975 N.E., 10th Street, Oklahoma City, Oklahoma, 73104, USA.
| |
Collapse
|
107
|
Sanz-Rubio D, Martin-Burriel I, Gil A, Cubero P, Forner M, Khalyfa A, Marin JM. Stability of Circulating Exosomal miRNAs in Healthy Subjects. Sci Rep 2018; 8:10306. [PMID: 29985466 PMCID: PMC6037782 DOI: 10.1038/s41598-018-28748-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 05/18/2018] [Indexed: 12/11/2022] Open
Abstract
Exosomes are nano-vesicles present in the circulation that are involved in cell-to-cell communication and regulation of different biological processes. MicroRNAs (miRNAs) are part of their cargo and are potential biomarkers. Methods of exosome isolation and the inter-individual and intra-individual variations in circulating miRNA exosomal cargo have been poorly investigated. This study aims for comparing two exosome isolation methods and to assess the stability of eleven plasma exosomal miRNAs over time. In addition to evaluate miRNA variability of both kits, the effect of freezing plasma before exosome isolation or freezing isolated exosomes on miRNA stability was also evaluated. MiRNA levels were tested in 7 healthy subjects who underwent four different blood extractions obtained in 4 consecutive weeks. One of the isolation kits displayed generally better amplification signals, and miRNAs from exosomes isolated after freezing the plasma had the highest levels. Intra-subject and inter-subject coefficients of variance were lower for the same isolation kit after freezing plasma. Finally, miRNAs that showed an acceptable expression level were stable across the consecutive extractions. This study shows for the first time the stability over time of miRNAs isolated from circulating plasma exosomes, establishing a key step in the use of exosomal miRNAs as biomarkers.
Collapse
Affiliation(s)
- David Sanz-Rubio
- Translational Respiratory Research Unit, IISAragon & CIBERES, Hospital Universitario Miguel Servet, Zaragoza, Spain.
| | | | - Ana Gil
- Translational Respiratory Research Unit, IISAragon & CIBERES, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Pablo Cubero
- Translational Respiratory Research Unit, IISAragon & CIBERES, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Marta Forner
- Translational Respiratory Research Unit, IISAragon & CIBERES, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Abdelnaby Khalyfa
- Section of Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Jose M Marin
- Translational Respiratory Research Unit, IISAragon & CIBERES, Hospital Universitario Miguel Servet, Zaragoza, Spain.,Department of Medicine, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
108
|
Ku A, Lim HC, Evander M, Lilja H, Laurell T, Scheding S, Ceder Y. Acoustic Enrichment of Extracellular Vesicles from Biological Fluids. Anal Chem 2018; 90:8011-8019. [PMID: 29806448 PMCID: PMC7556308 DOI: 10.1021/acs.analchem.8b00914] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) have emerged as a rich source of biomarkers providing diagnostic and prognostic information in diseases such as cancer. Large-scale investigations into the contents of EVs in clinical cohorts are warranted, but a major obstacle is the lack of a rapid, reproducible, efficient, and low-cost methodology to enrich EVs. Here, we demonstrate the applicability of an automated acoustic-based technique to enrich EVs, termed acoustic trapping. Using this technology, we have successfully enriched EVs from cell culture conditioned media and urine and blood plasma from healthy volunteers. The acoustically trapped samples contained EVs ranging from exosomes to microvesicles in size and contained detectable levels of intravesicular microRNAs. Importantly, this method showed high reproducibility and yielded sufficient quantities of vesicles for downstream analysis. The enrichment could be obtained from a sample volume of 300 μL or less, an equivalent to 30 min of enrichment time, depending on the sensitivity of downstream analysis. Taken together, acoustic trapping provides a rapid, automated, low-volume compatible, and robust method to enrich EVs from biofluids. Thus, it may serve as a novel tool for EV enrichment from large number of samples in a clinical setting with minimum sample preparation.
Collapse
Affiliation(s)
- Anson Ku
- Department of Translational Medicine, Lund University, SE-202 13 Malmö, Sweden
| | - Hooi Ching Lim
- Division of Molecular Hematology and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
| | - Mikael Evander
- Department of Biomedical Engineering, Lund University, SE-221 84 Lund, Sweden
| | - Hans Lilja
- Department of Translational Medicine, Lund University, SE-202 13 Malmö, Sweden
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, U.K., OX3 9DU
| | - Thomas Laurell
- Department of Biomedical Engineering, Lund University, SE-221 84 Lund, Sweden
| | - Stefan Scheding
- Division of Molecular Hematology and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
- Department of Hematology, Skåne University Hospital, SE-221-85, Lund, Sweden
| | - Yvonne Ceder
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, SE-223 81 Lund, Sweden
| |
Collapse
|
109
|
Pre-Analytical Handling Conditions and Small RNA Recovery from Urine for miRNA Profiling. J Mol Diagn 2018; 20:565-571. [PMID: 29936254 DOI: 10.1016/j.jmoldx.2018.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/19/2018] [Accepted: 04/30/2018] [Indexed: 12/24/2022] Open
Abstract
There are currently no standardized protocols for pre-analytical handling of urine to best preserve small RNA for miRNA profiling studies. miRNA is an attractive candidate as a potential biomarker because of the high level of stability in body fluids and its ability to be quantified on multiple high-throughput platforms. We present a comparison of small RNA recovery and stability in urine under alternate pre-analytical handling conditions and extend recommendations on what conditions optimize yield of miRNA from cell-free urine and urine extracellular vesicles (EVs). Using an affinity slurry for isolation of small RNA from urine, we found that urine samples held at room temperature (20°C) for up to 8 hours before processing yield the highest amounts of intact small RNAs from EVs. Some miRNA is lost from urine samples when held 2°C to 4°C and/or frozen before EV isolation, likely because of EV entrapment in uromodulin precipitates. However, we found that a simple 5-minute incubation of urine containing cold-induced precipitate at 37°C resolubilizes much of this precipitate and results in an increased recovery of EVs and miRNAs. Finally, small RNA integrity can be compromised when whole urine is held at 37°C for as little as 4 hours and is not conducive to efficient miRNA profiling.
Collapse
|
110
|
Raimondo F, Chinello C, Stella M, Santorelli L, Magni F, Pitto M. Effects of Hematuria on the Proteomic Profile of Urinary Extracellular Vesicles: Technical Challenges. J Proteome Res 2018; 17:2572-2580. [DOI: 10.1021/acs.jproteome.7b00763] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Francesca Raimondo
- School of Medicine and Surgery, University Milan Bicocca, 20900 Monza, Italy
| | - Clizia Chinello
- School of Medicine and Surgery, University Milan Bicocca, 20900 Monza, Italy
| | - Martina Stella
- School of Medicine and Surgery, University Milan Bicocca, 20900 Monza, Italy
| | - Lucia Santorelli
- School of Medicine and Surgery, University Milan Bicocca, 20900 Monza, Italy
| | - Fulvio Magni
- School of Medicine and Surgery, University Milan Bicocca, 20900 Monza, Italy
| | - Marina Pitto
- School of Medicine and Surgery, University Milan Bicocca, 20900 Monza, Italy
| |
Collapse
|
111
|
Lucchetti D, Fattorossi A, Sgambato A. Extracellular Vesicles in Oncology: Progress and Pitfalls in the Methods of Isolation and Analysis. Biotechnol J 2018; 14:e1700716. [PMID: 29878510 DOI: 10.1002/biot.201700716] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 05/28/2018] [Indexed: 12/22/2022]
Abstract
The possibility to study solid tumors through the analysis of extracellular vesicles in biological fluids is one of the most exciting and rapidly advancing field in cancer research. The extracellular vesicles are tiny sacs released in both physiological and pathological conditions and can be used to monitor the evolution of several pathological states, including neoplastic diseases. Indeed, these vesicles carry biological informations and can affect the behavior of recipient cells by transferring proteins, DNA, RNA, and microRNA. In this review the authors analyze the methods to collect biological fluid samples (urine, plasma/serum, and cell supernatant), and to isolate and quantify extracellular vesicles highlighting advantages and drawbacks. Moreover, the authors provide an overview on the adoption and the advantages of the methods (such as digital PCR, next generation sequencing, reverse-phase protein microarrays, flow-cytometry, etc.) most frequently used to analyze the molecular content of extracellular vesicles. Despite the great scientific interest on this topic, there is still a great uncertainty about which is the best method for the collection, isolation, quantification, and molecular evaluation of these vesicles and a standardization is needed. The features of EVs make them ideal candidates for liquid biopsy-based biomarkers. However, the small size of EVs makes their analysis very difficult and requires multiple advanced technologies, being therefore a limitation.
Collapse
Affiliation(s)
- Donatella Lucchetti
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Andrea Fattorossi
- Department of Obstetrics and Gynecology, Fondazione Policlinico A. Gemelli, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Alessandro Sgambato
- Institute of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| |
Collapse
|
112
|
McNicholas K, Li JY, Michael MZ, Gleadle JM. Albuminuria is not associated with elevated urinary vesicle concentration but can confound nanoparticle tracking analysis. Nephrology (Carlton) 2018; 22:854-863. [PMID: 27496221 DOI: 10.1111/nep.12867] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/18/2016] [Accepted: 07/29/2016] [Indexed: 12/13/2022]
Abstract
AIM Extracellular vesicles, such as exosomes, are present in urine with reports of roles in intercellular signalling and diagnostic utility. However, the extent to which the concentration and characteristics of urinary vesicles are altered in albuminuric renal disease has not been well characterized. In this study, we examined the number and characteristics of extracellular vesicles in albuminuric urine. METHODS Vesicles were isolated from the urine of 32 patients with varying levels of albuminuria using ultracentrifugation and density gradient purification and were examined using nanoparticle tracking analysis, immunoblotting and transmission electron microscopy. The size profile of particles in these urine preparations was compared with albumin-containing solutions. RESULTS Overall, there were no substantial differences in the number, or characteristics, of vesicles released into proteinuric urine. Analysis of albumin-containing solutions showed particles of exosome-like size, suggesting that such particles can mimic exosomes in standard nanoparticle tracking analysis. Albumin and IgG depletion of proteinuric urine resulted in a substantial reduction in the concentration of particles detected by nanoparticle tracking analysis. CONCLUSION There was no increase in urinary vesicle concentration in patients with albuminuria. Furthermore, these results demonstrate the need for cautious interpretation of nanoparticle tracking analysis of vesicle concentration in biological fluids containing protein and for sophisticated preparative methods in vesicle purification from urine.
Collapse
Affiliation(s)
- Kym McNicholas
- Department of Renal Medicine, Flinders Medical Centre, Adelaide, South Australia, Australia.,School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Jordan Yz Li
- Department of Renal Medicine, Flinders Medical Centre, Adelaide, South Australia, Australia.,School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Michael Z Michael
- Department of Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia.,School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Jonathan M Gleadle
- Department of Renal Medicine, Flinders Medical Centre, Adelaide, South Australia, Australia.,School of Medicine, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
113
|
Gonzalez-Calero L, Martínez PJ, Martin-Lorenzo M, Baldan-Martin M, Ruiz-Hurtado G, de la Cuesta F, Calvo E, Segura J, Lopez JA, Vázquez J, Barderas MG, Ruilope LM, Vivanco F, Alvarez-Llamas G. Urinary exosomes reveal protein signatures in hypertensive patients with albuminuria. Oncotarget 2018; 8:44217-44231. [PMID: 28562335 PMCID: PMC5546475 DOI: 10.18632/oncotarget.17787] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/20/2017] [Indexed: 01/05/2023] Open
Abstract
Albuminuria is an indicator of cardiovascular risk and renal damage in hypertensive individuals. Chronic renin-angiotensin system (RAS) suppression facilitates blood pressure control and prevents development of new-onset-albuminuria. A significant number of patients, however, develop albuminuria despite chronic RAS blockade, and the physiopathological mechanisms are underexplored. Urinary exosomes reflect pathological changes taking place in the kidney. The objective of this work was to examine exosomal protein alterations in hypertensive patients with albuminuria in the presence of chronic RAS suppression, to find novel clues underlying its development. Patients were followed-up for three years and were classified as: a) patients with persistent normoalbuminuria; b) patients developing de novo albuminuria; and c) patients with maintained albuminuria. Exosomal protein alterations between groups were identified by isobaric tag quantitation (iTRAQ). Confirmation was approached by target analysis (SRM). In total, 487 proteins were identified with high confidence. Specifically, 48 proteins showed an altered pattern in response to hypertension and/or albuminuria. Out of them, 21 proteins interact together in three main functional clusters: glycosaminoglycan degradation, coagulation and complement system, and oxidative stress. The identified proteins constitute potential targets for drug development and may help to define therapeutic strategies to evade albuminuria progression in hypertensive patients chronically treated.
Collapse
Affiliation(s)
| | - Paula J Martínez
- Department of Immunology, IIS-Fundacion Jimenez Diaz, REDinREN, Madrid, Spain
| | | | | | - Gema Ruiz-Hurtado
- Hypertension Unit, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando de la Cuesta
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos SESCAM, Toledo, Spain
| | | | - Julian Segura
- Hypertension Unit, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Jesús Vázquez
- Laboratory of Cardiovascular Proteomics CNIC, Madrid, Spain
| | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos SESCAM, Toledo, Spain
| | - Luis M Ruilope
- Hypertension Unit, Instituto de Investigación Imas12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Vivanco
- Department of Immunology, IIS-Fundacion Jimenez Diaz, REDinREN, Madrid, Spain.,Department of Biochemistry and Molecular Biology I, Universidad Complutense, Madrid, Spain
| | | |
Collapse
|
114
|
Improved isolation strategies to increase the yield and purity of human urinary exosomes for biomarker discovery. Sci Rep 2018; 8:3945. [PMID: 29500443 PMCID: PMC5834546 DOI: 10.1038/s41598-018-22142-x] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/12/2018] [Indexed: 12/13/2022] Open
Abstract
Circulating miRNAs are detected in extracellular space and body fluids such as urine. Circulating RNAs can be packaged in secreted urinary extracellular vesicles (uEVs) and thus protected from degradation. Urinary exosome preparations might contain specific miRNAs, relevant as biomarkers in renal and bladder diseases. Major difficulties in application of uEVs into the clinical environment are the high variability and low reproducibility of uEV isolation methods. Here we used five different methods to isolate uEVs and compared the size distribution, morphology, yield, presence of exosomal protein markers and RNA content of uEVs. We present an optimized ultracentrifugation and size exclusion chromatography approach for highly reproducible isolation for 50-150 nm uEVs, corresponding to the exosomes, from 50 ml urine. We profiled the miRNA content of uEVs and total urine from the same samples with the NanoString platform and validated the data using qPCR. Our results indicate that 18 miRNAs, robustly detected in uEVs were always present in the total urine. However, 15 miRNAs could be detected only in the total urine preparations and might represent naked circulating miRNA species. This is a novel unbiased and reproducible strategy for uEVs isolation, content normalization and miRNA cargo analysis, suitable for biomarker discovery studies.
Collapse
|
115
|
Abstract
PURPOSE The authors' purpose is to reveal the value of osteoblast-derived exosomes in bone diseases. METHODS Microvesicles from supernatants of mouse Mc3t3 were isolated by ultracentrifugation and then the authors presented the protein profile by proteomics analysis. RESULTS The authors detected a total number of 1536 proteins by mass spectrometry and found 172 proteins overlap with bone database. The Ingenuity Pathway Analysis shows network of "Skeletal and Muscular System Development and Function, Developmental Disorder, Hereditary Disorder" and pathway about osteogenesis. EFNB1 and transforming growth factor beta receptor 3 in the network, LRP6, bone morphogenetic protein receptor type-1, and SMURF1 in the pathway seemed to be valuable in the exosome research of related bone disease. CONCLUSIONS The authors' study unveiled the content of osteoblast-derived exosome and discussed valuable protein in it which might provide novel prospective in bone diseases research.
Collapse
|
116
|
Konoshenko MY, Lekchnov EA, Vlassov AV, Laktionov PP. Isolation of Extracellular Vesicles: General Methodologies and Latest Trends. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8545347. [PMID: 29662902 PMCID: PMC5831698 DOI: 10.1155/2018/8545347] [Citation(s) in RCA: 808] [Impact Index Per Article: 115.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/28/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Extracellular vesicles (EVs) play an essential role in the communication between cells and transport of diagnostically significant molecules. A wide diversity of approaches utilizing different biochemical properties of EVs and a lack of accepted protocols make data interpretation very challenging. SCOPE OF REVIEW This review consolidates the data on the classical and state-of-the-art methods for isolation of EVs, including exosomes, highlighting the advantages and disadvantages of each method. Various characteristics of individual methods, including isolation efficiency, EV yield, properties of isolated EVs, and labor consumption are compared. MAJOR CONCLUSIONS A mixed population of vesicles is obtained in most studies of EVs for all used isolation methods. The properties of an analyzed sample should be taken into account when planning an experiment aimed at studying and using these vesicles. The problem of adequate EVs isolation methods still remains; it might not be possible to develop a universal EV isolation method but the available protocols can be used towards solving particular types of problems. GENERAL SIGNIFICANCE With the wide use of EVs for diagnosis and therapy of various diseases the evaluation of existing methods for EV isolation is one of the key problems in modern biology and medicine.
Collapse
Affiliation(s)
- Maria Yu. Konoshenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, Novosibirsk 630055, Russia
| | - Evgeniy A. Lekchnov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, Novosibirsk 630055, Russia
| | - Alexander V. Vlassov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Pavel P. Laktionov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Novosibirsk 630090, Russia
- Meshalkin Siberian Federal Biomedical Research Center, Ministry of Public Health of the Russian Federation, Novosibirsk 630055, Russia
| |
Collapse
|
117
|
Pathare G, Dhayat NA, Mohebbi N, Wagner CA, Bobulescu IA, Moe OW, Fuster DG. Changes in V-ATPase subunits of human urinary exosomes reflect the renal response to acute acid/alkali loading and the defects in distal renal tubular acidosis. Kidney Int 2018; 93:871-880. [PMID: 29310826 DOI: 10.1016/j.kint.2017.10.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/02/2017] [Accepted: 10/05/2017] [Indexed: 12/26/2022]
Abstract
In the kidney, final urinary acidification is achieved by V-ATPases expressed in type A intercalated cells. The B1 subunit of the V-ATPase is required for maximal urinary acidification, while the role of the homologous B2 subunit is less clear. Here we examined the effect of acute acid/alkali loading in humans on B1 and B2 subunit abundance in urinary exosomes in normal individuals and of acid loading in patients with distal renal tubular acidosis (dRTA). Specificities of B1 and B2 subunit antibodies were verified by yeast heterologously expressing human B1 and B2 subunits, and murine wild-type and B1-deleted kidney lysates. Acute ammonium chloride loading elicited systemic acidemia, a drop in urinary pH, and increased urinary ammonium excretion. Nadir urinary pH was achieved at four to five hours, and exosomal B1 abundance was significantly increased at two through six hours after ammonium chloride loading. After acute equimolar sodium bicarbonate loading, blood and urinary pH rose rapidly, with a concomitant reduction of exosomal B1 abundance within two hours, which remained lower throughout the test. In contrast, no change in exosomal B2 abundance was found following acid or alkali loading. In patients with inherited or acquired distal RTA, the urinary B1 subunit was extremely low or undetectable and did not respond to acid loading in urine, whereas no change in B2 subunit was found. Thus, both B1 and B2 subunits of the V-ATPase are detectable in human urinary exosomes, and acid and alkali loading or distal RTA cause changes in the B1 but not B2 subunit abundance in urinary exosomes.
Collapse
Affiliation(s)
- Ganesh Pathare
- Division of Nephrology and Hypertension, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland; National Centre of Competence in Research Transcure, University of Bern, Bern, Switzerland
| | - Nasser A Dhayat
- Division of Nephrology and Hypertension, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nilufar Mohebbi
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland; National Center for Competence in Research Kidney.CH, Zurich, Switzerland
| | - Ion A Bobulescu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel G Fuster
- Division of Nephrology and Hypertension, Bern University Hospital, University of Bern, Bern, Switzerland; Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland; National Centre of Competence in Research Transcure, University of Bern, Bern, Switzerland.
| |
Collapse
|
118
|
Lv LL, Feng Y, Wen Y, Wu WJ, Ni HF, Li ZL, Zhou LT, Wang B, Zhang JD, Crowley SD, Liu BC. Exosomal CCL2 from Tubular Epithelial Cells Is Critical for Albumin-Induced Tubulointerstitial Inflammation. J Am Soc Nephrol 2018; 29:919-935. [PMID: 29295871 DOI: 10.1681/asn.2017050523] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 11/14/2017] [Indexed: 11/03/2022] Open
Abstract
Albuminuria is a key instigator of tubulointerstitial inflammation associated with CKD, but the mechanism through which filtered albumin propagates renal injury remains unclear. In this study, we explored the role in this process of exosome mRNA released from tubular epithelial cells (TECs). Compared with control mice, acute and chronic kidney injury models had more exosomes containing inflammatory cytokine mRNA, particularly the chemokine CCL2, in kidneys and urine. In vitro stimulation of TECs with BSA recapitulated this finding. Notably, the internalization of purified TEC exosomes by cultured macrophages increased if TECs were exposed to BSA. Macrophage internalization of exosomes from BSA-treated TECs led to an enhanced inflammatory response and macrophage migration, but CCL2 silencing in TECs prevented these effects. Using a GFP-CCL2 fusion mRNA construct, we observed direct transfer of CCL2 mRNA from TEC exosomes to macrophages. Mice subjected to tail vein injection of purified BSA-treated TEC exosomes developed tubular injury with renal inflammatory cell infiltration. However, injection of exosomes from BSA-treated CCL2-deficient TECs induced less severe kidney inflammation. Finally, in patients with IgA nephropathy, the increase of proteinuria correlated with augmented urinary excretion of exosomes with exaggerated expression of CCL2 mRNA. Moreover, the level of CCL2 mRNA in urinary exosomes correlated closely with levels of renal interstitial macrophage infiltration in these patients. Our studies demonstrate that the increasing release of exosomes that transfer CCL2 mRNA from TECs to macrophages constitutes a critical mechanism of albumin-induced tubulointerstitial inflammation.
Collapse
Affiliation(s)
- Lin-Li Lv
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Ye Feng
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Yi Wen
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Wei-Jun Wu
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Hai-Feng Ni
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Zuo-Lin Li
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Le-Ting Zhou
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Bin Wang
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Jian-Dong Zhang
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham Veterans Affairs Medical Centers, Durham, North Carolina
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| |
Collapse
|
119
|
Isolation and characterization of urinary extracellular vesicles: implications for biomarker discovery. Nat Rev Nephrol 2017. [PMID: 29081510 DOI: 10.1038/nrneph.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Urine is a valuable diagnostic medium and, with the discovery of urinary extracellular vesicles, is viewed as a dynamic bioactive fluid. Extracellular vesicles are lipid-enclosed structures that can be classified into three categories: exosomes, microvesicles (or ectosomes) and apoptotic bodies. This classification is based on the mechanisms by which membrane vesicles are formed: fusion of multivesicular bodies with the plasma membranes (exosomes), budding of vesicles directly from the plasma membrane (microvesicles) or those shed from dying cells (apoptotic bodies). During their formation, urinary extracellular vesicles incorporate various cell-specific components (proteins, lipids and nucleic acids) that can be transferred to target cells. The rigour needed for comparative studies has fueled the search for optimal approaches for their isolation, purification, and characterization. RNA, the newest extracellular vesicle component to be discovered, has received substantial attention as an extracellular vesicle therapeutic, and compelling evidence suggests that ex vivo manipulation of microRNA composition may have uses in the treatment of kidney disorders. The results of these studies are building the case that urinary extracellular vesicles act as mediators of renal pathophysiology. As the field of extracellular vesicle studies is burgeoning, this Review focuses on primary data obtained from studies of human urine rather than on data from studies of laboratory animals or cultured immortalized cells.
Collapse
|
120
|
Hu S, Musante L, Tataruch D, Xu X, Kretz O, Henry M, Meleady P, Luo H, Zou H, Jiang Y, Holthofer H. Purification and Identification of Membrane Proteins from Urinary Extracellular Vesicles using Triton X-114 Phase Partitioning. J Proteome Res 2017; 17:86-96. [PMID: 29090927 DOI: 10.1021/acs.jproteome.7b00386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Urinary extracellular vesicles (uEVs) have become a promising source for biomarkers accurately reflecting biochemical changes in kidney and urogenital diseases. Characteristically, uEVs are rich in membrane proteins associated with several cellular functions like adhesion, transport, and signaling. Hence, membrane proteins of uEVs should represent an exciting protein class with unique biological properties. In this study, we utilized uEVs to optimize the Triton X-114 detergent partitioning protocol targeted for membrane proteins and proceeded to their subsequent characterization while eliminating effects of Tamm-Horsfall protein, the most abundant interfering protein in urine. This is the first report aiming to enrich and characterize the integral transmembrane proteins present in human urinary vesicles. First, uEVs were enriched using a "hydrostatic filtration dialysis'' appliance, and then the enriched uEVs and lysates were verified by transmission electron microscopy. After using Triton X-114 phase partitioning, we generated an insoluble pellet fraction and aqueous phase (AP) and detergent phase (DP) fractions and analyzed them with LC-MS/MS. Both in- and off-gel protein digestion methods were used to reveal an increased number of membrane proteins of uEVs. After comparing with the identified proteins without phase separation as in our earlier publication, 199 different proteins were detected in DP. Prediction of transmembrane domains (TMDs) from these protein fractions showed that DP had more TMDs than other groups. The analyses of hydrophobicity revealed that the GRAVY score of DP was much higher than those of the other fractions. Furthermore, the analysis of proteins with lipid anchor revealed that DP proteins had more lipid anchors than other fractions. Additionally, KEGG pathway analysis showed that the DP proteins detected participate in endocytosis and signaling, which is consistent with the expected biological functions of membrane proteins. Finally, results of Western blotting confirmed that the membrane protein bands are found in the DP fraction instead of AP. In conclusion, our study validates the use of Triton X-114 phase partitioning protocol on uEVs for a targeted isolation of membrane proteins and to reduce sample complexity. This method successfully facilitates detection of potential biomarkers and druggable targets in uEVs.
Collapse
Affiliation(s)
- Shuiwang Hu
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Southern Medical University , Guangzhou, China
| | | | | | - Xiaomeng Xu
- Institute of Nephrology and Urology, The Third Affiliated Hospital, Southern Medical University , Guangzhou, China
| | - Oliver Kretz
- III. Medical Clinic, University Hospital Hamburg-Eppendorf , Hamburg, Germany
| | | | | | - Haihua Luo
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Southern Medical University , Guangzhou, China
| | - Hequn Zou
- Institute of Nephrology and Urology, The Third Affiliated Hospital, Southern Medical University , Guangzhou, China
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Southern Medical University , Guangzhou, China
| | - Harry Holthofer
- Freiburg Institute for Advanced Studies (FRIAS), Albert-Ludwigs University , Freiburg, Germany
| |
Collapse
|
121
|
Isolation and characterization of urinary extracellular vesicles: implications for biomarker discovery. Nat Rev Nephrol 2017; 13:731-749. [PMID: 29081510 DOI: 10.1038/nrneph.2017.148] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Urine is a valuable diagnostic medium and, with the discovery of urinary extracellular vesicles, is viewed as a dynamic bioactive fluid. Extracellular vesicles are lipid-enclosed structures that can be classified into three categories: exosomes, microvesicles (or ectosomes) and apoptotic bodies. This classification is based on the mechanisms by which membrane vesicles are formed: fusion of multivesicular bodies with the plasma membranes (exosomes), budding of vesicles directly from the plasma membrane (microvesicles) or those shed from dying cells (apoptotic bodies). During their formation, urinary extracellular vesicles incorporate various cell-specific components (proteins, lipids and nucleic acids) that can be transferred to target cells. The rigour needed for comparative studies has fueled the search for optimal approaches for their isolation, purification, and characterization. RNA, the newest extracellular vesicle component to be discovered, has received substantial attention as an extracellular vesicle therapeutic, and compelling evidence suggests that ex vivo manipulation of microRNA composition may have uses in the treatment of kidney disorders. The results of these studies are building the case that urinary extracellular vesicles act as mediators of renal pathophysiology. As the field of extracellular vesicle studies is burgeoning, this Review focuses on primary data obtained from studies of human urine rather than on data from studies of laboratory animals or cultured immortalized cells.
Collapse
|
122
|
Sódar BW, Kovács Á, Visnovitz T, Pállinger É, Vékey K, Pocsfalvi G, Turiák L, Buzás EI. Best practice of identification and proteomic analysis of extracellular vesicles in human health and disease. Expert Rev Proteomics 2017; 14:1073-1090. [DOI: 10.1080/14789450.2017.1392244] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Barbara W. Sódar
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Árpád Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Tamás Visnovitz
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Éva Pállinger
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Károly Vékey
- MS Proteomics Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest Hungary
| | - Gabriella Pocsfalvi
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy
| | - Lilla Turiák
- MS Proteomics Research Group, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest Hungary
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
- MTA-SE Immunoproteogenomic Extracellular Vesicle Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
123
|
Kang YT, Kim YJ, Bu J, Cho YH, Han SW, Moon BI. High-purity capture and release of circulating exosomes using an exosome-specific dual-patterned immunofiltration (ExoDIF) device. NANOSCALE 2017; 9:13495-13505. [PMID: 28862274 DOI: 10.1039/c7nr04557c] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
We present a microfluidic device for the capture and release of circulating exosomes from human blood. The exosome-specific dual-patterned immunofiltration (ExoDIF) device is composed of two distinct immuno-patterned layers, and is capable of enhancing the chance of binding between the antibody and exosomes by generating mechanical whirling, thus achieving high-throughput exosome isolation with high specificity. Moreover, follow-up recovery after the immuno-affinity based isolation, via cleavage of a linker, enables further downstream analysis. We verified the performance of the present device using MCF-7 secreted exosomes and found that both the concentration and proportion of exosome-sized vesicles were higher than in the samples obtained from the conventional exosome isolation kit. We then isolated exosomes from the human blood samples with our device to compare the exosome level between cancer patients and healthy donors. Cancer patients show a significantly higher exosome level with higher selectivity when validating the exosome-sized vesicles using both electron microscopy and nanoparticle tracking analysis. The captured exosomes from cancer patients also express abundant cancer-associated antigens, the epithelial cell adhesion molecule (EpCAM) on their surface. Our simple and rapid exosome recovery technique has huge potential to elucidate the function of exosomes in cancer patients and can thus be applied for various exosome-based cancer research studies.
Collapse
Affiliation(s)
- Yoon-Tae Kang
- Cell Bench Research Center, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
124
|
Puhka M, Takatalo M, Nordberg ME, Valkonen S, Nandania J, Aatonen M, Yliperttula M, Laitinen S, Velagapudi V, Mirtti T, Kallioniemi O, Rannikko A, Siljander PRM, af Hällström TM. Metabolomic Profiling of Extracellular Vesicles and Alternative Normalization Methods Reveal Enriched Metabolites and Strategies to Study Prostate Cancer-Related Changes. Am J Cancer Res 2017; 7:3824-3841. [PMID: 29109780 PMCID: PMC5667407 DOI: 10.7150/thno.19890] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
Body fluids are a rich source of extracellular vesicles (EVs), which carry cargo derived from the secreting cells. So far, biomarkers for pathological conditions have been mainly searched from their protein, (mi)RNA, DNA and lipid cargo. Here, we explored the small molecule metabolites from urinary and platelet EVs relative to their matched source samples. As a proof-of-concept study of intra-EV metabolites, we compared alternative normalization methods to profile urinary EVs from prostate cancer patients before and after prostatectomy and from healthy controls. Methods: We employed targeted ultra-performance liquid chromatography-tandem mass spectrometry to profile over 100 metabolites in the isolated EVs, original urine samples and platelets. We determined the enrichment of the metabolites in the EVs and analyzed their subcellular origin, pathways and relevant enzymes or transporters through data base searches. EV- and urine-derived factors and ratios between metabolites were tested for normalization of the metabolomics data. Results: Approximately 1 x 1010 EVs were sufficient for detection of metabolite profiles from EVs. The profiles of the urinary and platelet EVs overlapped with each other and with those of the source materials, but they also contained unique metabolites. The EVs enriched a selection of cytosolic metabolites including members from the nucleotide and spermidine pathways, which linked to a number of EV-resident enzymes or transporters. Analysis of the urinary EVs from the patients indicated that the levels of glucuronate, D-ribose 5-phosphate and isobutyryl-L-carnitine were 2-26-fold lower in all pre-prostatectomy samples compared to the healthy control and post-prostatectomy samples (p < 0.05). These changes were only detected from EVs by normalization to EV-derived factors or with metabolite ratios, and not from the original urine samples. Conclusions: Our results suggest that metabolite analysis of EVs from different samples is feasible using a high-throughput platform and relatively small amount of sample material. With the knowledge about the specific enrichment of metabolites and normalization methods, EV metabolomics could be used to gain novel biomarker data not revealed by the analysis of the original EV source materials.
Collapse
|
125
|
Acute regulated expression of pendrin in human urinary exosomes. Pflugers Arch 2017; 470:427-438. [PMID: 28803436 DOI: 10.1007/s00424-017-2049-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/23/2017] [Accepted: 07/26/2017] [Indexed: 10/19/2022]
Abstract
It is well known that pendrin, an apical Cl-/HCO3-exchanger in type B intercalated cells, is modulated by chronic acid-base disturbances and electrolyte intake. To study this adaptation further at the acute level, we analyzed urinary exosomes from individuals subjected to oral acute acid, alkali, and NaCl loading. Acute oral NH4Cl loading (n = 8) elicited systemic acidemia with a drop in urinary pH and an increase in urinary NH4 excretion. Nadir urinary pH was achieved 5 h after NH4Cl loading. Exosomal pendrin abundance was dramatically decreased at 3 h after acid loading. In contrast, after acute equimolar oral NaHCO3 loading (n = 8), urinary and venous blood pH rose rapidly with a significant attenuation of urinary NH4 excretion. Alkali loading caused rapid upregulation of exosomal pendrin abundance at 1 h and normalized within 3 h of treatment. Equimolar NaCl loading (n = 6) did not alter urinary or venous blood pH or urinary NH4 excretion. However, pendrin abundance in urinary exosomes was significantly reduced at 2 h of NaCl ingestion with lowest levels observed at 4 h after treatment. In patients with inherited distal renal tubular acidosis (dRTA), pendrin abundance in urinary exosomes was greatly reduced and did not change upon oral NH4Cl loading. In summary, pendrin can be detected and quantified in human urinary exosomes by immunoblotting. Acid, alkali, and NaCl loadings cause acute changes in pendrin abundance in urinary exosomes within a few hours. Our data suggest that exosomal pendrin is a promising urinary biomarker for acute acid-base and volume status changes in humans.
Collapse
|
126
|
A method for extracting and characterizing RNA from urine: For downstream PCR and RNAseq analysis. Anal Biochem 2017; 536:8-15. [PMID: 28803886 DOI: 10.1016/j.ab.2017.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 08/04/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022]
Abstract
Readily accessible samples such as urine or blood are seemingly ideal for differentiating and stratifying patients; however, it has proven a daunting task to identify reliable biomarkers in such samples. Noncoding RNA holds great promise as a source of biomarkers distinguishing physiologic wellbeing or illness. Current methods to isolate and characterize RNA molecules in urine are limited. In this proof of concept study, we present a method to extract and identify small noncoding RNAs in urine. Initially, quantitative reverse transcription PCR was applied to confirm the presence of microRNAs in total RNA extracted from urine. Once the presence of micro RNA in urine was confirmed, we developed a method to scale up RNA extraction to provide adequate amounts of RNA for next generation sequence analysis. The method described in this study is applicable to detecting a broad range of small noncoding RNAs in urine; thus, they have wide applicability for health and disease analyses.
Collapse
|
127
|
Urinary glycated uromodulin in diabetic kidney disease. Clin Sci (Lond) 2017; 131:1815-1829. [DOI: 10.1042/cs20160978] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/22/2017] [Accepted: 06/07/2017] [Indexed: 12/22/2022]
Abstract
Advanced glycation end-products (AGEs) form during oxidative stress, which is increased in diabetes mellitus (DM). Uromodulin is a protein with a renal protective effect, and may be subject to glycation. The implications of uromodulin glycation and AGEs in the urine are not understood. Here, immunoprecipitation and liquid chromatography–mass spectrometry identified glycated uromodulin (glcUMOD) in the urine of 62.5% of patients with diabetic kidney disease (DKD), 20.0% of patients with non-diabetic chronic kidney disease (CKD), and no DM patients with normal renal function or healthy control participants; a finding replicated in a larger cohort of 84 patients with CKD in a case–control study (35 with DM, 49 without). Uromodulin forms high molecular weight polymers that associate with microvesicles and exosomes. Differential centrifugation identified uromodulin in the supernatant, microvesicles, and exosomes of the urine of healthy participants, but only in the supernatant of samples from patients with DKD, suggesting that glycation influences uromodulin function. Finally, the diagnostic and prognostic utility of measuring urinary glcUMOD concentration was examined. Urinary glcUMOD concentration was substantially higher in DKD patients than non-diabetic CKD patients. Urinary glcUMOD concentration predicted DKD status, particularly in patients with CKD stages 1–3a aged <65 years and with urine glcUMOD concentration ≥9,000 arbitrary units (AU). Urinary uromodulin is apparently glycated in DKD and forms AGEs, and glcUMOD may serve as a biomarker for DKD.
Collapse
|
128
|
Hunter RW, Moorhouse R, Farrah TE, MacIntyre IM, Asai T, Gallacher PJ, Kerr D, Melville V, Czopek A, Morrison EE, Ivy JR, Dear JW, Bailey MA, Goddard J, Webb DJ, Dhaun N. First-in-Man Demonstration of Direct Endothelin-Mediated Natriuresis and Diuresis. Hypertension 2017; 70:192-200. [PMID: 28507171 PMCID: PMC5739104 DOI: 10.1161/hypertensionaha.116.08832] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/19/2016] [Accepted: 04/19/2017] [Indexed: 01/23/2023]
Abstract
Endothelin (ET) receptor antagonists are potentially novel therapeutic agents in chronic kidney disease and resistant hypertension, but their use is complicated by sodium and water retention. In animal studies, this side effect arises from ETB receptor blockade in the renal tubule. Previous attempts to determine whether this mechanism operates in humans have been confounded by the hemodynamic consequences of ET receptor stimulation/blockade. We aimed to determine the effects of ET signaling on salt transport in the human nephron by administering subpressor doses of the ET-1 precursor, big ET-1. We conducted a 2-phase randomized, double-blind, placebo-controlled crossover study in 10 healthy volunteers. After sodium restriction, subjects received either intravenous placebo or big ET-1, in escalating dose (≤300 pmol/min). This increased plasma concentration and urinary excretion of ET-1. Big ET-1 reduced heart rate (≈8 beats/min) but did not otherwise affect systemic hemodynamics or glomerular filtration rate. Big ET-1 increased the fractional excretion of sodium (from 0.5 to 1.0%). It also increased free water clearance and tended to increase the abundance of the sodium-potassium-chloride cotransporter (NKCC2) in urinary extracellular vesicles. Our protocol induced modest increases in circulating and urinary ET-1. Sodium and water excretion increased in the absence of significant hemodynamic perturbation, supporting a direct action of ET-1 on the renal tubule. Our data also suggest that sodium reabsorption is stimulated by ET-1 in the thick ascending limb and suppressed in the distal renal tubule. Fluid retention associated with ET receptor antagonist therapy may be circumvented by coprescribing potassium-sparing diuretics.
Collapse
Affiliation(s)
- Robert W Hunter
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Rebecca Moorhouse
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Tariq E Farrah
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Iain M MacIntyre
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Takae Asai
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Peter J Gallacher
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Debbie Kerr
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Vanessa Melville
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Alicja Czopek
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Emma E Morrison
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Jess R Ivy
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - James W Dear
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Matthew A Bailey
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Jane Goddard
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - David J Webb
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Neeraj Dhaun
- From the British Heart Foundation Centre of Research Excellence and The Queen's Medical Research Institute, University of Edinburgh, United Kingdom.
| |
Collapse
|
129
|
Park J, Hwang M, Choi B, Jeong H, Jung JH, Kim HK, Hong S, Park JH, Choi Y. Exosome Classification by Pattern Analysis of Surface-Enhanced Raman Spectroscopy Data for Lung Cancer Diagnosis. Anal Chem 2017; 89:6695-6701. [PMID: 28541032 DOI: 10.1021/acs.analchem.7b00911] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Owing to the role of exosome as a cargo for intercellular communication, especially in cancer metastasis, the evidence has been consistently accumulated that exosomes can be used as a noninvasive indicator of cancer. Consequently, several studies applying exosome have been proposed for cancer diagnostic methods such as ELISA assay. However, it has been still challenging to get reliable results due to the requirement of a labeling process and high concentration of exosome. Here, we demonstrate a label-free and highly sensitive classification method of exosome by combining surface-enhanced Raman scattering (SERS) and statistical pattern analysis. Unlike the conventional method to read different peak positions and amplitudes of a spectrum, whole SERS spectra of exosomes were analyzed by principal component analysis (PCA). By employing this pattern analysis, lung cancer cell derived exosomes were clearly distinguished from normal cell derived exosomes by 95.3% sensitivity and 97.3% specificity. Moreover, by analyzing the PCA result, we could suggest that this difference was induced by 11 different points in SERS signals from lung cancer cell derived exosomes. This result paved the way for new real-time diagnosis and classification of lung cancer by using exosome as a cancer marker.
Collapse
Affiliation(s)
- Jaena Park
- Department of Bio-convergence Engineering, Korea University , Seoul 02841, South Korea
| | - Miyeon Hwang
- School of Bio-medical Engineering, Korea University , Seoul 02841, South Korea
| | - ByeongHyeon Choi
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University Guro Hospital , Seoul 08308, South Korea
| | - Hyesun Jeong
- School of Biosystem and Biomedical Science, Korea University , Seoul, 02841, South Korea
| | - Jik-Han Jung
- Department of Bio and Brain Bioengineering, Korea Advanced Institute of Science and Technology (KAIST) , Daejeon 34141, South Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Korea University Guro Hospital , Seoul 08308, South Korea
| | - Sunghoi Hong
- School of Biosystem and Biomedical Science, Korea University , Seoul, 02841, South Korea
| | - Ji-Ho Park
- Department of Bio and Brain Bioengineering, Korea Advanced Institute of Science and Technology (KAIST) , Daejeon 34141, South Korea
| | - Yeonho Choi
- Department of Bio-convergence Engineering, Korea University , Seoul 02841, South Korea.,School of Bio-medical Engineering, Korea University , Seoul 02841, South Korea
| |
Collapse
|
130
|
Zou G, Benktander JD, Gizaw ST, Gaunitz S, Novotny MV. Comprehensive Analytical Approach toward Glycomic Characterization and Profiling in Urinary Exosomes. Anal Chem 2017; 89:5364-5372. [PMID: 28402650 DOI: 10.1021/acs.analchem.7b00062] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exosomes are extracellular nanosized vesicles with lipid bilayers encapsulating nucleic acids and proteins, both with and without glycosylation. While exosomal nucleic acids and proteins have previously been explored to identify cancer biomarkers with some promising results, little information has been available concerning their glycoconjugate content. Exosomes were isolated from normal urine samples through multistep differential centrifugation. The isolated exosomes have an average size of 146 nm and a spherical shape, as determined by dynamic light scattering and transmission electron microscopy, respectively. N-Glycans were enzymatically released from the isolated vesicles. After being reduced and permethylated, N-glycans were measured by MALDI mass spectrometry. Paucimannosidic, high-mannose, and complex type glycans were identified and their relative abundances were determined. Some detailed structures of these glycans were revealed through liquid chromatography/tandem mass spectrometry (LC/MS-MS). The reduced N-glycans, without being permethylated, were also separated and analyzed by LC/MS-MS, and their structures were further detailed through isomeric separation on porous graphitized carbon (PGC) packed in long capillaries. Using microfractionation before LC/MS-MS, minor multiantennary N-glycans were preconcentrated as based on hydrophobicity or charge. Preconcentration of the reduced and permethylated glycans on a C18 cartridge revealed numerous large glycans, whereas fractionation of the reduced N-glycans by ion-exchange cartridges facilitated detection of sulfated glycans. After removing N-glycans from the original sample aliquot, O-glycans were chemically released from urinary exosomes and profiled, revealing some unusual structures.
Collapse
Affiliation(s)
- Guozhang Zou
- Department of Chemistry, Indiana University , Bloomington, Indiana 47405, United States
| | - John D Benktander
- Department of Chemistry, Indiana University , Bloomington, Indiana 47405, United States
| | - Solomon T Gizaw
- Department of Chemistry, Indiana University , Bloomington, Indiana 47405, United States
| | - Stefan Gaunitz
- Department of Chemistry, Indiana University , Bloomington, Indiana 47405, United States
| | - Milos V Novotny
- Department of Chemistry, Indiana University , Bloomington, Indiana 47405, United States
| |
Collapse
|
131
|
Liang LG, Kong MQ, Zhou S, Sheng YF, Wang P, Yu T, Inci F, Kuo WP, Li LJ, Demirci U, Wang S. An integrated double-filtration microfluidic device for isolation, enrichment and quantification of urinary extracellular vesicles for detection of bladder cancer. Sci Rep 2017; 7:46224. [PMID: 28436447 PMCID: PMC5402302 DOI: 10.1038/srep46224] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/13/2017] [Indexed: 01/20/2023] Open
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles, are present in a variety of bodily fluids, and the concentration of these sub-cellular vesicles and their associated biomarkers (proteins, nucleic acids, and lipids) can be used to aid clinical diagnosis. Although ultracentrifugation is commonly used for isolation of EVs, it is highly time-consuming, labor-intensive and instrument-dependent for both research laboratories and clinical settings. Here, we developed an integrated double-filtration microfluidic device that isolated and enriched EVs with a size range of 30–200 nm from urine, and subsequently quantified the EVs via a microchip ELISA. Our results showed that the concentration of urinary EVs was significantly elevated in bladder cancer patients (n = 16) compared to healthy controls (n = 8). Receiver operating characteristic (ROC) analysis demonstrated that this integrated EV double-filtration device had a sensitivity of 81.3% at a specificity of 90% (16 bladder cancer patients and 8 healthy controls). Thus, this integrated device has great potential to be used in conjunction with urine cytology and cystoscopy to improve clinical diagnosis of bladder cancer in clinics and at point-of-care (POC) settings.
Collapse
Affiliation(s)
- Li-Guo Liang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China.,Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Meng-Qi Kong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China.,Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Sherry Zhou
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University, School of Medicine, Palo Alto, CA 94304 USA
| | - Ye-Feng Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China.,Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Ping Wang
- Department of Urology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
| | - Tao Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China.,Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China
| | - Fatih Inci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University, School of Medicine, Palo Alto, CA 94304 USA
| | - Winston Patrick Kuo
- Harvard Catalyst-Laboratory for Innovative Translational Technologies, Harvard Medical School, Boston, MA 02115, USA.,CloudHealth Genomics, Ltd, Shanghai, 201499, China
| | - Lan-Juan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University, School of Medicine, Palo Alto, CA 94304 USA.,Department of Electrical Engineering (By courtesy), Stanford University, Stanford, CA 94305, USA
| | - ShuQi Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang Province, 310003, China.,Institute for Translational Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310029, China.,Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University, School of Medicine, Palo Alto, CA 94304 USA
| |
Collapse
|
132
|
Barreiro K, Holthofer H. Urinary extracellular vesicles. A promising shortcut to novel biomarker discoveries. Cell Tissue Res 2017; 369:217-227. [PMID: 28429073 PMCID: PMC5487850 DOI: 10.1007/s00441-017-2621-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/30/2017] [Indexed: 12/13/2022]
Abstract
Proteomic and genomic techniques have reached full maturity and are providing unforeseen details for the comprehensive understanding of disease pathologies at a fraction of previous costs. However, for kidney diseases, many gaps in such information remain to inhibit major advances in the prevention, treatment and diagnostics of these devastating diseases, which have enormous global impact. The discovery of ubiquitous extracellular vesicles (EV) in all bodily fluids is rapidly increasing the fundamental knowledge of disease mechanisms and the ways in which cells communicate with distant locations in processes of cancer spread, immunological regulation, barrier functions and general modulation of cellular activity. In this review, we describe some of the most prominent research streams and findings utilizing urinary extracellular vesicles as highly versatile and dynamic tools with their extraordinary protein and small regulatory RNA species. While being a highly promising approach, the relatively young field of EV research suffers from a lack of adherence to strict standardization and carefully scrutinized methods for obtaining fully reproducible results. With the appropriate guidelines and standardization achieved, urine is foreseen as forming a unique, robust and easy route for determining accurate and personalized disease signatures and as providing highly useful early biomarkers of the disease pathology of the kidney and beyond.
Collapse
Affiliation(s)
- Karina Barreiro
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Harry Holthofer
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland. .,Freiburg Institute for Advanced Studies, Albert-Ludwigs University Freiburg, Freiburg, Germany.
| |
Collapse
|
133
|
Urine exosomes from healthy and hypertensive pregnancies display elevated level of α-subunit and cleaved α- and γ-subunits of the epithelial sodium channel—ENaC. Pflugers Arch 2017; 469:1107-1119. [DOI: 10.1007/s00424-017-1977-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 10/19/2022]
|
134
|
Koritzinsky EH, Street JM, Star RA, Yuen PST. Quantification of Exosomes. J Cell Physiol 2017; 232:1587-1590. [PMID: 27018079 DOI: 10.1002/jcp.25387] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 03/23/2016] [Indexed: 12/11/2022]
Abstract
Exosomes are released by cells as self-contained vesicles with an intact lipid bilayer that encapsulates a small portion of the parent cell. Exosomes have been studied widely as information-rich sources of potential biomarkers that can reveal cellular physiology. We suggest that quantification is essential to understand basic biological relationships between exosomes and their parent cells and hence the underlying interpretation of exosome signals. The number of methods for quantifying exosomes has expanded as interest in exosomes has increased. However, a consensus on proper quantification has not developed, making each study difficult to compare to another. Overcoming this ad hoc approach will require widely available standards that have been adequately characterized, and multiple comparative studies across platforms. We outline the current status of these technical approaches and our view of how they can become more coherent. J. Cell. Physiol. 232: 1587-1590, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Jonathan M Street
- Renal Diagnostics and Therapeutics Unit, NIDDK, NIH, Bethesda, Maryland
| | - Robert A Star
- Renal Diagnostics and Therapeutics Unit, NIDDK, NIH, Bethesda, Maryland
| | - Peter S T Yuen
- Renal Diagnostics and Therapeutics Unit, NIDDK, NIH, Bethesda, Maryland
| |
Collapse
|
135
|
Abstract
Paper-based devices chemically functionalized with capturing molecules enable the isolation and characterization of extracellular vesicles (EVs) from samples of limited amount. Here, we describe the isolation of EV subpopulations from human serum samples. The morphology, content, and amount of captured EVs can be assessed using scanning electron microscopy (SEM ), transcriptome analysis, and paper-based enzyme-linked immunosorbent assays (pELISA), respectively. A colorimetric readout can be detected from 10 μL serum within 10 min.
Collapse
Affiliation(s)
- Yi-Hsing Hsiao
- Institute of Nanoengineering and Microsystems, National Tsing Hua University, No.101, Section 2, Guangfu Rd., East Dist, Hsinchu, 30013, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Chihchen Chen
- Institute of Nanoengineering and Microsystems, National Tsing Hua University, No.101, Section 2, Guangfu Rd., East Dist, Hsinchu, 30013, Taiwan.
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
136
|
Street J, Koritzinsky E, Glispie D, Star R, Yuen P. Urine Exosomes: An Emerging Trove of Biomarkers. Adv Clin Chem 2017; 78:103-122. [PMID: 28057185 DOI: 10.1016/bs.acc.2016.07.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Exosomes are released by most cells and can be isolated from all biofluids including urine. Exosomes are small vesicles formed as part of the endosomal pathway that contain cellular material surrounded by a lipid bilayer that can be traced to the plasma membrane. Exosomes are potentially a more targeted source of material for biomarker discovery than unfractionated urine, and provide diagnostic and pathophysiological information without an invasive tissue biopsy. Cytoplasmic contents including protein, mRNA, miRNA, and lipids have all been studied within the exosomal fraction. Many prospective urinary exosomal biomarkers have been successfully identified for a variety of kidney or genitourinary tract conditions; detection of systemic conditions may also be possible. Isolation and analysis of exosomes can be achieved by several approaches, although many require specialized equipment or involve lengthy protocols. The need for timely analysis in the clinical setting has driven considerable innovation with several promising options recently emerging. Consensus on exosome isolation, characterization, and normalization procedures would resolve critical clinical translational bottlenecks for existing candidate exosomal biomarkers and provide a template for additional discovery studies.
Collapse
|
137
|
Musante L, Tataruch-Weinert D, Kerjaschki D, Henry M, Meleady P, Holthofer H. Residual urinary extracellular vesicles in ultracentrifugation supernatants after hydrostatic filtration dialysis enrichment. J Extracell Vesicles 2016; 6:1267896. [PMID: 28326167 PMCID: PMC5328348 DOI: 10.1080/20013078.2016.1267896] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Indexed: 12/16/2022] Open
Abstract
Urinary extracellular vesicles (UEVs) appear an ideal source of biomarkers for kidney and urogenital diseases. The majority of protocols designed for their isolation are based on differential centrifugation steps. However, little is still known of the type and amount of vesicles left in the supernatant. Here we used an isolation protocol for UEVs which uses hydrostatic filtration dialysis as first pre-enrichment step, followed by differential centrifugation. Transmission electron microscopy (TEM), mass spectrometry (MS), western blot, ELISA assays and tuneable resistive pulse sensing (TRPS) were used to characterise and quantify UEVs in the ultracentrifugation supernatant. TEM showed the presence of a variety of small size vesicles in the supernatant while protein identification by MS matched accurately with the protein list available in Vesiclepedia. Screening and relative quantification for specific vesicle markers showed that the supernatant was preferentially positive for CD9 and TSG101. ELISA tests for quantification of exosome revealed that 14%, was left in the supernatant with a particle diameter of 110 nm and concentration of 1.54 × 1010/ml. Here we show a comprehensive characterisation of exosomes and other small size urinary vesicles which the conventional differential centrifugation protocol may lose.
Collapse
Affiliation(s)
- Luca Musante
- Centre for BioAnalytical Sciences, Dublin City University , Dublin 9 , Ireland
| | | | - Dontscho Kerjaschki
- Clinical Institute of Pathology, Medical University of Vienna , Vienna , Austria
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University , Dublin 9 , Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University , Dublin 9 , Ireland
| | - Harry Holthofer
- Freiburg Institute for Advanced Studies, Albert-Ludwigs University , Freiburg , Germany
| |
Collapse
|
138
|
Isolation and mass spectrometry analysis of urinary extraexosomal proteins. Sci Rep 2016; 6:36331. [PMID: 27805059 PMCID: PMC5090863 DOI: 10.1038/srep36331] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 10/06/2016] [Indexed: 12/20/2022] Open
Abstract
The aim of the present study was to develop a LC-MS/MS-based proteomic analysis method of urinary exosomal proteins that has the potential to discover disease biomarkers. In short, urinary exosomes from healthy subjects were isolated by immunocapture on magnetic beads, detected by immunofluorescence and TEM, trypsin digested directly on the beads for an accelerated time with no addition of detergents before performing an LC-MS analysis of the trypsinate. To our knowledge, this is the first proteomic analysis of proteins displayed on the outer surface of exosomes. The outer exosome proteome may contain proteins that are of higher biomarker value compared to soluble cargo protein as the proteins projecting into the extracellular milieu might be more directly involved in physiological functions of exosomes. The proteomic analysis identified 49 proteins that were considered significant; the majority is involved in carbohydrate and lipid metabolism or in immune responses. Thirty of the proteins are linked to diseases. The developed proteomic method exploiting urinary exosomes might be of great value in search for diagnostic or prognostic biomarkers of especially metabolic and immune-related diseases.
Collapse
|
139
|
Mincheva-Nilsson L, Baranov V, Nagaeva O, Dehlin E. Isolation and Characterization of Exosomes from Cultures of Tissue Explants and Cell Lines. CURRENT PROTOCOLS IN IMMUNOLOGY 2016; 115:14.42.1-14.42.21. [PMID: 27801511 DOI: 10.1002/cpim.17] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exosomes are specialized, nanometer-sized extracellular vesicles of endosomal origin actively secreted into the extracellular space by a variety of cells under normal and pathological conditions. Exosomes have recently emerged as important intercellular communicators and modulators of diverse mechanisms and cellular responses. Characterization of their composition and function will open possibilities for new diagnostic methods and promising therapeutic approaches based on nanobiology. This unit provides a standard isolation procedure for purification of exosomes based on density gradient ultracentrifugation with sucrose. The process of isolating exosomes relies on obtaining proper source fluids/supernatants as well as qualitative and quantitative assessment of the isolated vesicles. The methodological procedures here can be divided in three parts: (1) pre-isolation procedures aiming to obtain fluids containing exosomes, with a focus on protocols for organ explants and cell cultures; (2) a procedure for exosome isolation with several gradient alternatives; and (3) post-isolation procedures for estimating the purity and yield of the exosomal fraction. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Lucia Mincheva-Nilsson
- Department of Clinical Microbiology, Division of Clinical Immunology, Umeå University, Umeå, Sweden
| | - Vladimir Baranov
- Department of Clinical Microbiology, Division of Clinical Immunology, Umeå University, Umeå, Sweden
| | - Olga Nagaeva
- Department of Clinical Microbiology, Division of Clinical Immunology, Umeå University, Umeå, Sweden
| | - Eva Dehlin
- Department of Clinical Microbiology, Division of Clinical Immunology, Umeå University, Umeå, Sweden
| |
Collapse
|
140
|
Puhka M, Nordberg ME, Valkonen S, Rannikko A, Kallioniemi O, Siljander P, Af Hällström TM. KeepEX, a simple dilution protocol for improving extracellular vesicle yields from urine. Eur J Pharm Sci 2016; 98:30-39. [PMID: 27771514 DOI: 10.1016/j.ejps.2016.10.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 10/11/2016] [Accepted: 10/18/2016] [Indexed: 12/22/2022]
Abstract
Urinary extracellular vesicles (EVs) are a promising source of biomarkers, which can be obtained in a non-invasive manner. However, the yield of EVs from urine samples may be insufficient for various analyses due to the entrapment of EVs by the Tamm-Horsfall protein (THP) meshwork. Here, we developed a simple dilution protocol to increase the urinary EV yield by disrupting the interaction between THP filaments and EVs with the help of alkaline pH and lowered ionic concentration. The integrity of the EVs and THP was assessed by electron microscopy. The effect of the protocol on the EV yield was quantified against an undiluted control by western blotting of four EV markers, nanoparticle tracking analysis and measuring of the RNA/miRNA concentration of the EV samples. The average EV yield from the dilution protocol was 2-7 fold the yield from the undiluted control i.e. increased by 130-624% as measured by western blotting and NTA. The yield increased most from samples with a high THP to EV ratio. The morphology and size range of the EVs were unaltered by the protocol. However, RNA/miRNA yields were the same as from the undiluted control and THP filaments could still be detected in EV samples. The dilution protocol, that we named KeepEX, provides a simple and efficient way to prevent loss of EVs thus increasing their yield from urine. Since KeepEX does not require individual adjustment of sample pH nor extra centrifugation steps, it could be used on its own or in combination with other EV purification protocols to improve EV isolation particularly from small urine volumes.
Collapse
Affiliation(s)
- M Puhka
- Institute for Molecular Medicine Finland (FIMM), Helsinki Urological Biobank, University of Helsinki, Finland.
| | - M-E Nordberg
- Institute for Molecular Medicine Finland (FIMM), Helsinki Urological Biobank, University of Helsinki, Finland
| | - S Valkonen
- EV Group, Division of Biochemistry and Biotechnology, Department of Biosciences and Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Finland; Finnish Red Cross Blood Service, Helsinki, Finland
| | - A Rannikko
- Institute for Molecular Medicine Finland (FIMM), Helsinki Urological Biobank, University of Helsinki, Finland; Helsinki University Central Hospital, Department of Urology, Helsinki, Finland
| | - O Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Urological Biobank, University of Helsinki, Finland
| | - P Siljander
- EV Group, Division of Biochemistry and Biotechnology, Department of Biosciences and Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Finland
| | - T M Af Hällström
- Institute for Molecular Medicine Finland (FIMM), Helsinki Urological Biobank, University of Helsinki, Finland; Orion Corporation, Orion Pharma, Orionintie 1, P.O. Box 65, 02101 Espoo, Finland
| |
Collapse
|
141
|
Proteome Profiling of Urinary Exosomes Identifies Alpha 1-Antitrypsin and H2B1K as Diagnostic and Prognostic Biomarkers for Urothelial Carcinoma. Sci Rep 2016; 6:34446. [PMID: 27686150 PMCID: PMC5043375 DOI: 10.1038/srep34446] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/14/2016] [Indexed: 11/09/2022] Open
Abstract
MALDI-TOF spectrometry has not been used for urinary exosome analysis. We used it for determining UC biomarkers. From 2012 to 2015, we enrolled 129 consecutive patients with UC and 62 participants without UC. Exosomes from their urine were isolated, and analyzed through MALDI-TOF spectrometry. Immunohistochemical (IHC) analysis of another 122 UC and 26 non-UC tissues was conducted to verify the discovered biomarkers. Two peaks at m/z 5593 (fragmented peptide of alpha-1-antitrypsin; sensitivity, 50.4%; specificity, 96.9%) and m/z 5947 (fragmented peptide of histone H2B1K sensitivity, 62.0%; specificity, 92.3%) were identified as UC diagnosis exosome biomarkers. UC patients with detectable histone H2B1K showed 2.29- and 3.11-fold increased risks of recurrence and progression, respectively, compared with those with nondetectable histone H2B1K. Verification results of IHC staining revealed significantly higher expression of alpha 1-antitrypsin (p = 0.038) and H2B1K (p = 0.005) in UC tissues than in normal tissues. The expression of alpha 1-antitrypsin and H2B1K in UC tissues was significantly correlated with UC grades (p < 0.05). Urinary exosome proteins alpha 1-antitrypsin and histone H2B1K, which are identified through MALDI-TOF analysis, could facilitate rapid diagnosis and prognosis of UC.
Collapse
|
142
|
Øverbye A, Skotland T, Koehler CJ, Thiede B, Seierstad T, Berge V, Sandvig K, Llorente A. Identification of prostate cancer biomarkers in urinary exosomes. Oncotarget 2016. [PMID: 26196085 PMCID: PMC4745805 DOI: 10.18632/oncotarget.4851] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Exosomes have recently appeared as a novel source of non-invasive cancer biomarkers since tumour-specific molecules can be found in exosomes isolated from biological fluids. We have here investigated the proteome of urinary exosomes by using mass spectrometry to identify proteins differentially expressed in prostate cancer patients compared to healthy male controls. In total, 15 control and 16 prostate cancer samples of urinary exosomes were analyzed. Importantly, 246 proteins were differentially expressed in the two groups. The majority of these proteins (221) were up-regulated in exosomes from prostate cancer patients. These proteins were analyzed according to specific criteria to create a focus list that contained 37 proteins. At 100% specificity, 17 of these proteins displayed individual sensitivities above 60%. Even though several of these proteins showed high sensitivity and specificity for prostate cancer as individual biomarkers, combining them in a multi-panel test has the potential for full differentiation of prostate cancer from non-disease controls. The highest sensitivity, 94%, was observed for transmembrane protein 256 (TM256; chromosome 17 open reading frame 61). LAMTOR proteins were also distinctly enriched with very high specificity for patient samples. TM256 and LAMTOR1 could be used to augment the sensitivity to 100%. Other prominent proteins were V-type proton ATPase 16 kDa proteolipid subunit (VATL), adipogenesis regulatory factor (ADIRF), and several Rab-class members and proteasomal proteins. In conclusion, this study clearly shows the potential of using urinary exosomes in the diagnosis and clinical management of prostate cancer.
Collapse
Affiliation(s)
- Anders Øverbye
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway.,Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway.,Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Christian J Koehler
- The Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Bernd Thiede
- The Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Therese Seierstad
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Viktor Berge
- Department of Urology, Oslo University Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway.,Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway.,Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
143
|
De Palma G, Sallustio F, Schena FP. Clinical Application of Human Urinary Extracellular Vesicles in Kidney and Urologic Diseases. Int J Mol Sci 2016; 17:ijms17071043. [PMID: 27376269 PMCID: PMC4964419 DOI: 10.3390/ijms17071043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/16/2016] [Accepted: 06/22/2016] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) have been isolated in different body fluids, including urine. The cargo of urinary EVs is composed of nucleic acids and proteins reflecting the physiological and possibly pathophysiological state of cells lining the nephron and the urinary tract. Urinary EVs have been confirmed to contain low amounts of various types of RNA that play a role in intercellular communication by transferring genetic information. This communication through EV RNAs includes both continuation of normal physiological processes and conditioning in disease mechanisms. Although proteins included in urinary EVs represent only 3% of the whole-urine proteome, urinary EVs can influence cells in the renal epithelia not only by delivering RNA cargo, but also by delivering a wide range of proteins. Since urine is a readily available biofluid, the discovery of EVs has opened a new field of biomarker research. The potential use of urinary EV RNAs and proteins as diagnostic biomarkers for various kidney and urologic diseases is currently being explored. Here, we review recent studies that deal in identifying biomarker candidates for human kidney and urologic diseases using urinary EVs and might help to understand the pathophysiology.
Collapse
Affiliation(s)
- Giuseppe De Palma
- C.A.R.S.O. Consortium, University of Bari, Valenzano 70010, Italy.
- Schena Foundation-European Research Center for Kidney Diseases, Valenzano 70010, Italy.
| | - Fabio Sallustio
- Department of Emergency and Organ Transplantation, University of Bari, DETO, Bari 70124, Italy.
| | | |
Collapse
|
144
|
Welton JL, Brennan P, Gurney M, Webber JP, Spary LK, Carton DG, Falcón-Pérez JM, Walton SP, Mason MD, Tabi Z, Clayton A. Proteomics analysis of vesicles isolated from plasma and urine of prostate cancer patients using a multiplex, aptamer-based protein array. J Extracell Vesicles 2016; 5:31209. [PMID: 27363484 PMCID: PMC4929354 DOI: 10.3402/jev.v5.31209] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 04/11/2016] [Accepted: 04/17/2016] [Indexed: 12/28/2022] Open
Abstract
Proteomics analysis of biofluid-derived vesicles holds enormous potential for discovering non-invasive disease markers. Obtaining vesicles of sufficient quality and quantity for profiling studies has, however, been a major problem, as samples are often replete with co-isolated material that can interfere with the identification of genuine low abundance, vesicle components. Here, we used a combination of ultracentrifugation and size-exclusion chromatography to isolate and analyse vesicles of plasma or urine origin. We describe a sample-handling workflow that gives reproducible, quality vesicle isolations sufficient for subsequent protein profiling. Using a semi-quantitative aptamer-based protein array, we identified around 1,000 proteins, of which almost 400 were present at comparable quantities in plasma versus urine vesicles. Significant differences were, however, apparent with elements like HSP90, integrin αVβ5 and Contactin-1 more prevalent in urinary vesicles, while hepatocyte growth factor activator, prostate-specific antigen–antichymotrypsin complex and many others were more abundant in plasma vesicles. This was also applied to a small set of specimens collected from men with metastatic prostate cancer, highlighting several proteins with the potential to indicate treatment refractory disease. The study provides a practical platform for furthering protein profiling of vesicles in prostate cancer, and, hopefully, many other disease scenarios.
Collapse
Affiliation(s)
- Joanne Louise Welton
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom.,Cardiff School of Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
| | - Paul Brennan
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Mark Gurney
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Jason Paul Webber
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Lisa Kate Spary
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - David Gil Carton
- Metabolomics Unit, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain
| | | | - Sean Peter Walton
- Department of Computer Science, College of Science, Swansea University, United Kingdom
| | - Malcolm David Mason
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Zsuzsanna Tabi
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom
| | - Aled Clayton
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom.,Velindre Cancer Centre, Cardiff, United Kingdom;
| |
Collapse
|
145
|
Morrison EE, Bailey MA, Dear JW. Renal extracellular vesicles: from physiology to clinical application. J Physiol 2016; 594:5735-5748. [PMID: 27104781 DOI: 10.1113/jp272182] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/15/2016] [Indexed: 12/27/2022] Open
Abstract
Urinary extracellular vesicles (uEVs) are released from all regions of the kidney's nephron and from other cells that line the urinary tract. Extracellular vesicles retain proteomic and transcriptomic markers specific to their cell of origin and so represent a potential reservoir for kidney disease biomarker discovery. Exosomes, a subtype of uEVs, are distinguished from other vesicles by features related to their biogenesis within cells: mature multi-vesicular bodies fuse with the cellular membrane to liberate exosomes into the extracellular space. uEVs represent a novel cell signalling mechanism because they can be shuttled to a recipient cell and, through a number of proposed mechanisms, affect the recipient cell's proteome and function. Here we review the current evidence for uEV signalling along the nephron, their role in health and disease of the kidney, and their potential for clinical translation as biomarkers and therapeutics.
Collapse
Affiliation(s)
- E E Morrison
- University/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - M A Bailey
- University/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
| | - J W Dear
- University/BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
146
|
Santucci L, Bruschi M, Candiano G, Lugani F, Petretto A, Bonanni A, Ghiggeri GM. Urine Proteome Biomarkers in Kidney Diseases. I. Limits, Perspectives, and First Focus on Normal Urine. Biomark Insights 2016; 11:41-8. [PMID: 26997865 PMCID: PMC4795486 DOI: 10.4137/bmi.s26229] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/29/2015] [Accepted: 12/30/2015] [Indexed: 11/05/2022] Open
Abstract
Urine proteome is a potential source of information in renal diseases, and it is considered a natural area of investigation for biomarkers. Technology developments have markedly increased the power analysis on urinary proteins, and it is time to confront methodologies and results of major studies on the topics. This is a first part of a series of reviews that will focus on the urine proteome as a site for detecting biomarkers of renal diseases; the theme of the first review concerns methodological aspects applied to normal urine. Main issues are techniques for urine pretreatment, separation of exosomes, use of combinatorial peptide ligand libraries, mass spectrometry approaches, and analysis of data sets. Available studies show important differences, suggesting a major confounding effect of the technologies utilized for analysis. The objective is to obtain consensus about which approaches should be utilized for studying urine proteome in renal diseases.
Collapse
Affiliation(s)
- Laura Santucci
- Laboratory on Physiopathology of Uremia, Istituto Giannina Gaslini, Genova, Italy
| | - Maurizio Bruschi
- Laboratory on Physiopathology of Uremia, Istituto Giannina Gaslini, Genova, Italy
| | - Giovanni Candiano
- Laboratory on Physiopathology of Uremia, Istituto Giannina Gaslini, Genova, Italy
| | - Francesca Lugani
- Nephrology, Dialysis and Transplantation, Istituto Giannina Gaslini, Genova, Italy
| | - Andrea Petretto
- Laboratory of Mass Spectrometry - Core Facility, Istituto Giannina Gaslini, Genova, Italy
| | - Alice Bonanni
- Nephrology, Dialysis and Transplantation, Istituto Giannina Gaslini, Genova, Italy
| | - Gian Marco Ghiggeri
- Laboratory on Physiopathology of Uremia, Istituto Giannina Gaslini, Genova, Italy.; Nephrology, Dialysis and Transplantation, Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
147
|
Salih M, Demmers JA, Bezstarosti K, Leonhard WN, Losekoot M, van Kooten C, Gansevoort RT, Peters DJM, Zietse R, Hoorn EJ. Proteomics of Urinary Vesicles Links Plakins and Complement to Polycystic Kidney Disease. J Am Soc Nephrol 2016; 27:3079-3092. [PMID: 26940098 DOI: 10.1681/asn.2015090994] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/19/2015] [Indexed: 12/21/2022] Open
Abstract
Novel therapies in autosomal dominant polycystic kidney disease (ADPKD) signal the need for markers of disease progression or response to therapy. This study aimed to identify disease-associated proteins in urinary extracellular vesicles (uEVs), which include exosomes, in patients with ADPKD. We performed quantitative proteomics on uEVs from healthy controls and patients with ADPKD using a labeled approach and then used a label-free approach with uEVs of different subjects (healthy controls versus patients with ADPKD versus patients with non-ADPKD CKD). In both experiments, 30 proteins were consistently more abundant (by two-fold or greater) in ADPKD-uEVs than in healthy- and CKD-uEVs. Of these proteins, we selected periplakin, envoplakin, villin-1, and complement C3 and C9 for confirmation because they were also significantly overrepresented in pathway analysis and were previously implicated in ADPKD pathogenesis. Immunoblotting confirmed higher abundances of the selected proteins in uEVs from three independent groups of patients with ADPKD. Whereas uEVs of young patients with ADPKD and preserved kidney function already had higher levels of complement, only uEVs of patients with advanced stages of ADPKD had increased levels of villin-1, periplakin, and envoplakin. Furthermore, all five proteins correlated positively with total kidney volume. Analysis in kidney tissue from mice with kidney-specific, tamoxifen-inducible Pkd1 deletion demonstrated higher expression in more severe stages of the disease and correlation with kidney weight for each protein of interest. In summary, proteomic analysis of uEVs identified plakins and complement as disease-associated proteins in ADPKD. These proteins are new candidates for evaluation as biomarkers or targets for therapy in ADPKD.
Collapse
Affiliation(s)
- Mahdi Salih
- Department of Internal Medicine, Division of Nephrology & Transplantation, and
| | - Jeroen A Demmers
- Proteomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Karel Bezstarosti
- Proteomics Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | - Cees van Kooten
- Nephrology, Leiden University Medical Center, Leiden, The Netherlands; and
| | - Ron T Gansevoort
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Robert Zietse
- Department of Internal Medicine, Division of Nephrology & Transplantation, and
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology & Transplantation, and
| | | |
Collapse
|
148
|
Wachalska M, Koppers-Lalic D, van Eijndhoven M, Pegtel M, Geldof AA, Lipinska AD, van Moorselaar RJ, Bijnsdorp IV. Protein Complexes in Urine Interfere with Extracellular Vesicle Biomarker Studies. J Circ Biomark 2016; 5:4. [PMID: 28936252 PMCID: PMC5548314 DOI: 10.5772/62579] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/18/2016] [Indexed: 02/03/2023] Open
Abstract
Urine exosomes (extracellular vesicles; EVs) contain (micro)RNA (miRNA) and protein biomarkers that are useful for the non-invasive diagnosis of various urological diseases. However, the urinary Tamm-Horsfall protein (THP) complex, which forms at reduced temperatures, may affect EV isolation and may also lead to contamination by other molecules including microRNAs (miRNAs). Therefore, we compared the levels of three miRNAs within the purified EV fraction and THP- protein-network. Urine was collected from healthy donors and EVs were isolated by ultracentrifugation (UC), two commercial kits or sepharose size-exclusion chromatography (SEC). SEC enables the separation of EVs from protein-complexes in urine. After UC, the isolation of EV-miRNA was compared with two commercial kits. The EV isolation efficiency was evaluated by measuring the EV protein markers, Alix and TSG101, CD63 by Western blotting, or miR-375, miR-204 and miR-21 by RT-qPCR. By using commercial kits, EV isolation resulted in either low yields or dissimilar miRNA levels. Via SEC, the EVs were separated from the protein-complex fraction. Importantly, a different ratio was observed between the three miRNAs in the protein fraction compared to the EV fraction. Thus, protein-complexes within urine may influence EV-biomarker studies. Therefore, the characterization of the isolated EV fraction is important to obtain reproducible results.
Collapse
Affiliation(s)
- Magda Wachalska
- Department of Urology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology UG-MUG, University of Gdańsk, Gdańsk, Poland
| | - Danijela Koppers-Lalic
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Neurosurgery, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Michiel Pegtel
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert A Geldof
- Department of Urology, VU University Medical Center, Amsterdam, The Netherlands
| | - Andrea D Lipinska
- Department of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology UG-MUG, University of Gdańsk, Gdańsk, Poland
| | | | - Irene V Bijnsdorp
- Department of Urology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
149
|
Turco AE, Lam W, Rule AD, Denic A, Lieske JC, Miller VM, Larson JJ, Kremers WK, Jayachandran M. Specific renal parenchymal-derived urinary extracellular vesicles identify age-associated structural changes in living donor kidneys. J Extracell Vesicles 2016; 5:29642. [PMID: 26837814 PMCID: PMC4737715 DOI: 10.3402/jev.v5.29642] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 12/21/2022] Open
Abstract
Non-invasive tests to identify age and early disease-associated pathology within the kidney are needed. Specific populations of urinary extracellular vesicles (EVs) could potentially be used for such a diagnostic test. Random urine samples were obtained from age- and sex-stratified living kidney donors before kidney donation. A biopsy of the donor kidney was obtained at the time of transplantation to identify nephron hypertrophy (larger glomerular volume, cortex per glomerulus and mean profile tubular area) and nephrosclerosis (% fibrosis, % glomerulosclerosis and arteriosclerosis). Renal parenchymal-derived EVs in cell-free urine were quantified by digital flow cytometry. The relationship between these EV populations and structural pathology on the kidney biopsy was assessed. Clinical characteristics of the kidney donors (n=138, age range: 20–70 years, 50% women) were within the normative range. Overall, urine from women contained more EVs than that from men. The number of exosomes, juxtaglomerular cells and podocyte marker–positive EVs decreased (p<0.05) with increasing age. There were fewer total EVs as well as EVs positive for mesangial cell, parietal cell, descending limb of Henle's loop (simple squamous epithelium), collecting tubule-intercalated cell and monocyte chemoattractant protein-1 markers (p<0.05) in persons with nephron hypertrophy. The number of EVs positive for intercellular adhesion molecule-1, juxtaglomerular cell, podocyte, parietal cell, proximal tubular epithelial cell, distal tubular epithelial cell and collecting duct cells were fewer (p<0.05) in persons with nephrosclerosis. EVs carrying markers of cells from the renal pelvis epithelium did not associate with any indices of nephron hypertrophy or nephrosclerosis. Therefore, specific populations of EVs derived from cells of the glomerulus and nephron associate with underlying kidney structural changes. Further validation of these findings in other cohorts is needed to determine their clinical utility.
Collapse
Affiliation(s)
- Anne E Turco
- Department of Physiology & Biomedical Engineering, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Wing Lam
- Department of Physiology & Biomedical Engineering, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Department of Internal Medicine, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Aleksandar Denic
- Division of Nephrology and Hypertension, Department of Internal Medicine, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - John C Lieske
- Division of Nephrology and Hypertension, Department of Internal Medicine, College of Medicine, Mayo Clinic, Rochester, MN, USA.,Laboratory Medicine and Pathology, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Virginia M Miller
- Department of Physiology & Biomedical Engineering, College of Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Surgery, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joseph J Larson
- Department of Health Sciences Research, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Walter K Kremers
- Department of Health Sciences Research, College of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Muthuvel Jayachandran
- Department of Physiology & Biomedical Engineering, College of Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Surgery, College of Medicine, Mayo Clinic, Rochester, MN, USA;
| |
Collapse
|
150
|
Tutakhel OAZ, Jeleń S, Valdez-Flores M, Dimke H, Piersma SR, Jimenez CR, Deinum J, Lenders JW, Hoenderop JGJ, Bindels RJM. Alternative splice variant of the thiazide-sensitive NaCl cotransporter: a novel player in renal salt handling. Am J Physiol Renal Physiol 2016; 310:F204-16. [DOI: 10.1152/ajprenal.00429.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/31/2015] [Indexed: 11/22/2022] Open
Abstract
The thiazide-sensitive NaCl cotransporter (NCC) is an important pharmacological target in the treatment of hypertension. The human SLC12A3 gene, encoding NCC, gives rise to three isoforms. Only the third isoform has been extensively investigated. The aim of the present study was, therefore, to establish the abundance and localization of the almost identical isoforms 1 and 2 (NCC1/2) in the human kidney and to determine their functional properties and regulation in physiological conditions. Immunohistochemical analysis of NCC1/2 in the human kidney revealed that NCC1/2 localizes to the apical plasma membrane of the distal convoluted tubule. Importantly, NCC1/2 mRNA constitutes ∼44% of all NCC isoforms in the human kidney. Functional analysis performed in the Xenopus laevis oocyte revealed that thiazide-sensitive 22Na+ transport of NCC1 was significantly increased compared with NCC3. Mimicking a constitutively active phosphorylation site at residue 811 (S811D) in NCC1 further augmented Na+ transport, while a nonphosphorylatable variant (S811A) of NCC1 prevented this enhanced response. Analysis of human urinary exosomes demonstrated that water loading in human subjects significantly reduces the abundance of NCC1/2 in urinary exosomes. The present study highlights that previously underrepresented NCC1/2 is a fully functional thiazide-sensitive NaCl-transporting protein. Being significantly expressed in the kidney, it may constitute a unique route of renal NaCl reabsorption and could, therefore, play an important role in blood pressure regulation.
Collapse
Affiliation(s)
- Omar A. Z. Tutakhel
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sabina Jeleń
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marco Valdez-Flores
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Sander R. Piersma
- OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Connie R. Jimenez
- OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jaap Deinum
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jacques W. Lenders
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany; and
| | - Joost G. J. Hoenderop
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - René J. M. Bindels
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|