1
|
Lawson JS, Williams TL. Extracellular vesicles in kidney disease - A veterinary perspective. Vet J 2024; 308:106247. [PMID: 39276847 DOI: 10.1016/j.tvjl.2024.106247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/21/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Extracellular vesicles (EVs) are membrane bound vesicles secreted from cells into the extracellular space which have an emerging role in both normal kidney physiology and the pathophysiology of kidney injury, predominantly as mediators of intercellular communication. EVs contain proteins and RNA cargo which reflect their cell of origin and can be isolated from the urine of cats and dogs. The majority of urinary EVs (uEVs) originate from the kidney, and both the uEV proteome and transcriptome have been investigated as sources of biomarkers of kidney disease. In addition to their possible diagnostic role, EVs may also have therapeutic potential, and veterinary species have been used as models to demonstrate the efficacy of exogenous EVs derived from mesenchymal stromal cells in the treatment of acute kidney injury. Furthermore, bioengineered EVs may represent a novel vehicle for the administration of drugs or therapeutic nucleic acids in kidney disease. This article reviews the biological functions of EVs within the kidney, techniques for their isolation, and their potential use as biomarkers and therapeutic agents, with particular focus on the potential significance to veterinary patients.
Collapse
Affiliation(s)
- Jack S Lawson
- The Royal Veterinary College, Hawkshead Ln, Brookmans Park, Hatfield AL9 7TA, UK.
| | - Timothy L Williams
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| |
Collapse
|
2
|
Korecka K, Gawin M, Pastuszka A, Partyka M, Koszutski T, Pietrowska M, Hyla-Klekot L. Proteomics of urinary small extracellular vesicles in early diagnosis of kidney diseases in children-expectations and limitations. Proteomics 2024; 24:e2300168. [PMID: 38213025 DOI: 10.1002/pmic.202300168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/13/2024]
Abstract
The primary function of the kidneys is to maintain systemic homeostasis (disruption of renal structure and function results in multilevel impairment of body function). Kidney diseases are characterized by a chronic, progressive course and may result in the development of chronic kidney disease (CKD). Evaluation of the composition of the proteome of urinary small extracellular vesicles (sEVs) as a so-called liquid biopsy is a promising new research direction. Knowing the composition of sEV could allow localization of cellular changes in specific sections of the nephron or the interstitial tissue before fixed changes, detectable only at an advanced stage of the disease, occur. Research is currently underway on the role of sEVs in the diagnosis and monitoring of many disease entities. Reports in the literature on the subject include: diabetic nephropathy, focal glomerulosclerosis in the course of glomerulopathies, renal fibrosis of various etiologies. Studies on pediatric patients are still few, involving piloting if small groups of patients without validation studies. Here, we review the literature addressing the use of sEV for diagnosis of the most common urinary disorders in children. We evaluate the clinical utility and define limitations of markers present in sEV as potential liquid biopsy.
Collapse
Affiliation(s)
- Klaudia Korecka
- Clinical Department of Paediatric Surgery and Urology, Medical University of Silesia in Katowice, Katowice, Poland
| | - Marta Gawin
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Agnieszka Pastuszka
- Clinical Department of Paediatric Surgery and Urology, Medical University of Silesia in Katowice, Katowice, Poland
| | - Mirosław Partyka
- Department of Laboratory Diagnostics, School of Medicine in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Tomasz Koszutski
- Clinical Department of Paediatric Surgery and Urology, Medical University of Silesia in Katowice, Katowice, Poland
| | - Monika Pietrowska
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Lidia Hyla-Klekot
- Clinical Department of Paediatric Surgery and Urology, Medical University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
3
|
Grange C, Dalmasso A, Cortez JJ, Spokeviciute B, Bussolati B. Exploring the role of urinary extracellular vesicles in kidney physiology, aging, and disease progression. Am J Physiol Cell Physiol 2023; 325:C1439-C1450. [PMID: 37842748 PMCID: PMC10861146 DOI: 10.1152/ajpcell.00349.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Extracellular vesicles (EVs), membranous vesicles present in all body fluids, are considered important messengers, carrying their information over long distance and modulating the gene expression profile of recipient cells. EVs collected in urine (uEVs) are mainly originated from the apical part of urogenital tract, following the urine flow. Moreover, bacterial-derived EVs are present within urine and may reflect the composition of microbiota. Consolidated evidence has established the involvement of uEVs in renal physiology, being responsible for glomerular and tubular cross talk and among different tubular segments. uEVs may also be involved in other physiological functions such as modulation of innate immunity, coagulation, or metabolic activities. Furthermore, it has been recently remonstrated that age, sex, endurance excise, and lifestyle may influence uEV composition and release, modifying their cargo. On the other hand, uEVs appear modulators of different urogenital pathological conditions, triggering disease progression. uEVs sustain fibrosis and inflammation processes, both involved in acute and chronic kidney diseases, aging, and stone formation. The molecular signature of uEVs collected from diseased patients can be of interest for understanding kidney physiopathology and for identifying diagnostic and prognostic biomarkers.
Collapse
Affiliation(s)
- Cristina Grange
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alessia Dalmasso
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Judiel John Cortez
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Beatrice Spokeviciute
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
4
|
Nørgård MØ, Svenningsen P. Acute Kidney Injury by Ischemia/Reperfusion and Extracellular Vesicles. Int J Mol Sci 2023; 24:15312. [PMID: 37894994 PMCID: PMC10607034 DOI: 10.3390/ijms242015312] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Acute kidney injury (AKI) is often caused by ischemia-reperfusion injury (IRI). IRI significantly affects kidney metabolism, which elicits pro-inflammatory responses and kidney injury. The ischemia/reperfusion of the kidney is associated with transient high mitochondrial-derived reactive oxygen species (ROS) production rates. Excessive mitochondrial-derived ROS damages cellular components and, together with other pathogenic mechanisms, elicits a range of acute injury mechanisms that impair kidney function. Mitochondrial-derived ROS production also stimulates epithelial cell secretion of extracellular vesicles (EVs) containing RNAs, lipids, and proteins, suggesting that EVs are involved in AKI pathogenesis. This literature review focuses on how EV secretion is stimulated during ischemia/reperfusion and how cell-specific EVs and their molecular cargo may modify the IRI process. Moreover, critical pitfalls in the analysis of kidney epithelial-derived EVs are described. In particular, we will focus on how the release of kidney epithelial EVs is affected during tissue analyses and how this may confound data on cell-to-cell signaling. By increasing awareness of methodological pitfalls in renal EV research, the risk of false negatives can be mitigated. This will improve future EV data interpretation regarding EVs contribution to AKI pathogenesis and their potential as biomarkers or treatments for AKI.
Collapse
Affiliation(s)
| | - Per Svenningsen
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark;
| |
Collapse
|
5
|
Palmer TC, Hunter RW. Using RNA-based therapies to target the kidney in cardiovascular disease. Front Cardiovasc Med 2023; 10:1250073. [PMID: 37868774 PMCID: PMC10587590 DOI: 10.3389/fcvm.2023.1250073] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
RNA-based therapies are currently used for immunisation against infections and to treat metabolic diseases. They can modulate gene expression in immune cells and hepatocytes, but their use in other cell types has been limited by an inability to selectively target specific tissues. Potential solutions to this targeting problem involve packaging therapeutic RNA molecules into delivery vehicles that are preferentially delivered to cells of interest. In this review, we consider why the kidney is a desirable target for RNA-based therapies in cardiovascular disease and discuss how such therapy could be delivered. Because the kidney plays a central role in maintaining cardiovascular homeostasis, many extant drugs used for preventing cardiovascular disease act predominantly on renal tubular cells. Moreover, kidney disease is a major independent risk factor for cardiovascular disease and a global health problem. Chronic kidney disease is projected to become the fifth leading cause of death by 2040, with around half of affected individuals dying from cardiovascular disease. The most promising strategies for delivering therapeutic RNA selectively to kidney cells make use of synthetic polymers and engineered extracellular vesicles to deliver an RNA cargo. Future research should focus on establishing the safety of these novel delivery platforms in humans, on developing palatable routes of administration and on prioritising the gene targets that are likely to have the biggest impact in cardiovascular disease.
Collapse
Affiliation(s)
- Trecia C. Palmer
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert W. Hunter
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Renal Medicine, Royal Infirmary ofEdinburgh, Edinburgh, United Kingdom
| |
Collapse
|
6
|
Oh S, Lee CM, Kwon SH. Extracellular Vesicle MicroRNA in the Kidney. Compr Physiol 2023; 13:4833-4850. [PMID: 37358511 PMCID: PMC11514415 DOI: 10.1002/cphy.c220023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Most cells in our body release membrane-bound, nano-sized particles into the extracellular milieu through cellular metabolic processes. Various types of macromolecules, reflecting the physiological and pathological status of the producing cells, are packaged into such so-called extracellular vesicles (EVs), which can travel over a distance to target cells, thereby transmitting donor cell information. The short, noncoding ribonucleic acid (RNA) called microRNA (miRNA) takes a crucial part in EV-resident macromolecules. Notably, EVs transferring miRNAs can induce alterations in the gene expression profiles of the recipient cells, through genetically instructed, base-pairing interaction between the miRNAs and their target cell messenger RNAs (mRNAs), resulting in either nucleolytic decay or translational halt of the engaged mRNAs. As in other body fluids, EVs released in urine, termed urinary EVs (uEVs), carry specific sets of miRNA molecules, which indicate either normal or diseased states of the kidney, the principal source of uEVs. Studies have therefore been directed to elucidate the contents and biological roles of miRNAs in uEVs and moreover to utilize the gene regulatory properties of miRNA cargos in ameliorating kidney diseases through their delivery via engineered EVs. We here review the fundamental principles of the biology of EVs and miRNA as well as our current understanding of the biological roles and applications of EV-loaded miRNAs in the kidney. We further discuss the limitations of contemporary research approaches, suggesting future directions to overcome the difficulties to advance both the basic biological understanding of miRNAs in EVs and their clinical applications in treating kidney diseases. © 2023 American Physiological Society. Compr Physiol 13:4833-4850, 2023.
Collapse
Affiliation(s)
- Sekyung Oh
- Department of Medical Science, Catholic Kwandong University College of Medicine, Incheon 22711, South Korea
| | - Chang Min Lee
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, U.S.A
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, U.S.A
| |
Collapse
|
7
|
Erdbrügger U, Hoorn EJ, Le TH, Blijdorp CJ, Burger D. Extracellular Vesicles in Kidney Diseases: Moving Forward. KIDNEY360 2023; 4:245-257. [PMID: 36821616 PMCID: PMC10103258 DOI: 10.34067/kid.0001892022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/18/2022] [Indexed: 12/23/2022]
Abstract
Extracellular vesicles (EVs) are evolving as novel cell mediators, biomarkers, and therapeutic targets in kidney health and disease. They are naturally derived from cells both within and outside the kidney and carry cargo which mirrors the state of the parent cell. Thus, they are potentially more sensitive and disease-specific as biomarkers and messengers in various kidney diseases. Beside their role as novel communicators within the nephron, they likely communicate between different organs affected by various kidney diseases. Study of urinary EVs (uEVs) can help to fill current knowledge gaps in kidney diseases. However, separation and characterization are challenged by their heterogeneity in size, shape, and cargo. Fortunately, more sensitive and direct EV measuring tools are in development. Many clinical syndromes in nephrology from acute to chronic kidney and glomerular to tubular diseases have been studied. Yet, validation of biomarkers in larger cohorts is warranted and simpler tools are needed. Translation from in vitro to in vivo studies is also urgently needed. The therapeutic role of uEVs in kidney diseases has been studied extensively in rodent models of AKI. On the basis of the current exponential growth of EV research, the field of EV diagnostics and therapeutics is moving forward.
Collapse
Affiliation(s)
- Uta Erdbrügger
- Division of Nephrology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Ewout J. Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Thu H. Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Charles J. Blijdorp
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dylan Burger
- Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
8
|
Urinary extracellular vesicles: does cargo reflect tissue? Curr Opin Nephrol Hypertens 2022; 31:464-470. [PMID: 35894281 DOI: 10.1097/mnh.0000000000000822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To review recent developments in urinary extracellular vesicles (uEVs) to study kidney physiology and disease. RECENT FINDINGS Proteomic analysis in rats showed significant correlations between kidney and uEV protein abundances. Consistent with uEV biogenesis, these correlations were stronger for membrane-associated proteins than for e.g. soluble kinases or E3 ubiquitin ligases. When challenged with a high potassium diet, the physiologically predicted protein changes occurred both in kidney and uEVs, suggesting that analysis of uEVs might be utilized as a proxy or even replacement for tissue analysis. Although kidney-uEV correlations are more difficult to obtain in humans, analysis of uEV cargo from patients with inherited tubulopathies or with primary aldosteronism were also consistent with the predicted changes at the tissue level. The kidney appears to be the main source of uEVs, with a recent study showing that nephron mass determines uEV excretion rate. Therefore, a measure of nephron mass should be included for between-subject comparisons. SUMMARY The overall good correlation between kidney and uEV protein abundances renders uEVs an attractive noninvasive source of biomarkers for studying kidney physiology or disease. However, differences in per-protein kidney-uEV correlations and per-person uEV excretion rates should be considered in uEV biomarker studies.
Collapse
|
9
|
Blijdorp CJ, Burger D, Llorente A, Martens-Uzunova ES, Erdbrügger U. Extracellular Vesicles as Novel Players in Kidney Disease. J Am Soc Nephrol 2022; 33:467-471. [PMID: 35131841 PMCID: PMC8975061 DOI: 10.1681/asn.2021091232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Charles J. Blijdorp
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dylan Burger
- Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway,Department for Mechanical Electronics and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway
| | - Elena S. Martens-Uzunova
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Uta Erdbrügger
- Department of Medicine, Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
10
|
Kumar P, Zadjali F, Yao Y, Johnson D, Siroky B, Astrinidis A, Vogel P, Gross KW, Bissler JJ. Tsc2 mutation induces renal tubular cell nonautonomous disease. Genes Dis 2022; 9:187-200. [PMID: 35005118 PMCID: PMC8720703 DOI: 10.1016/j.gendis.2021.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/25/2021] [Accepted: 03/31/2021] [Indexed: 01/22/2023] Open
Abstract
TSC renal cystic disease is poorly understood and has no approved treatment. In a new principal cell-targeted murine model of Tsc cystic disease, the renal cystic epithelium is mostly composed of type A intercalated cells with an intact Tsc2 gene confirmed by sequencing, although these cells exhibit a Tsc-mutant disease phenotype. We used a newly derived targeted murine model in lineage tracing and extracellular vesicle (EV) characterization experiments and a cell culture model in EV characterization and cellular induction experiments to understand TSC cystogenesis. Using lineage tracing experiments, we found principal cells undergo clonal expansion but contribute very few cells to the cyst. We determined that cystic kidneys contain more interstitial EVs than noncystic kidneys, excrete fewer EVs in urine, and contain EVs in cyst fluid. Moreover, the loss of Tsc2 gene in EV-producing cells greatly changes the effect of EVs on renal tubular epithelium, such that the epithelium develops increased secretory and proliferative pathway activity. We demonstate that the mTORC1 pathway activity is independent form the EV production, and that the EV effects for a single cell line can vary significantly. TSC cystogenesis involves significant contribution from genetically intact cells conscripted to the mutant phenotype by mutant cell derived EVs.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - Fahad Zadjali
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Ying Yao
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - Daniel Johnson
- Molecular Bioinformatics Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Brian Siroky
- Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - Aristotelis Astrinidis
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| | - Peter Vogel
- Department of Veterinary Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - John J Bissler
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Pediatric Medicine Department, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
11
|
Blijdorp CJ, Hartjes TA, Wei K, van Heugten MH, Bovée DM, Budde RP, van de Wetering J, Hoenderop JG, van Royen ME, Zietse R, Severs D, Hoorn EJ. Nephron mass determines the excretion rate of urinary extracellular vesicles. J Extracell Vesicles 2022; 11:e12181. [PMID: 35064766 PMCID: PMC8783354 DOI: 10.1002/jev2.12181] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 11/15/2021] [Accepted: 12/08/2021] [Indexed: 12/20/2022] Open
Abstract
Urinary extracellular vesicles (uEVs) are emerging as non-invasive biomarkers for various kidney diseases, but it is unknown how differences in nephron mass impact uEV excretion. To address this, uEV excretion was measured before and after human kidney donor nephrectomy and rat nephrectomy. In male and female donors, uEVs were quantified in cell-free spot and 24-h urine samples using nanoparticle tracking analysis (NTA), EVQuant, and CD9-time-resolved fluorescence immunoassay. Female donors had significantly lower total kidney volume (TKV) and excreted 49% fewer uEVs than male donors. uEV excretion correlated positively with estimated glomerular filtration rate (eGFR), creatinine clearance, and TKV (R's between 0.6 and 0.7). uEV excretion rate could also be predicted from spot urines after multiplying spot uEV/creatinine by 24-h urine creatinine. Donor nephrectomy reduced eGFR by 36% ± 10%, but the excretion of uEVs by only 16% (CD9+ uEVs -37%, CD9- uEVs no decrease). Donor nephrectomy increased the podocyte marker WT-1 and the proximal tubule markers NHE3, NaPi-IIa, and cubilin in uEVs two- to four-fold when correcting for the nephrectomy. In rats, the changes in GFR and kidney weight correlated with the changes in uEV excretion rate (R = 0.46 and 0.60, P < 0.01). Furthermore, the estimated degree of hypertrophy matched the change in uEV excretion rate (1.4- to 1.5-fold after uninephrectomy and four-fold after 5/6th nephrectomy). Taken together, our data show that uEV excretion depends on nephron mass, and that nephrectomy reduces uEV excretion less than expected based on nephron loss due to compensatory hypertrophy. The major implication of our findings is that a measure for nephron mass or uEV excretion rate should be included when comparing uEV biomarkers between individuals.
Collapse
Affiliation(s)
- Charles J. Blijdorp
- Department of Internal Medicine, Division of Nephrology and TransplantationErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| | - Thomas A. Hartjes
- Department of PathologyErasmus Medical Center, University Medical Center RotterdamRotterdamThe Netherlands
| | - Kuang‐Yu Wei
- Department of Internal Medicine, Division of Nephrology and TransplantationErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| | - Martijn H. van Heugten
- Department of Internal Medicine, Division of Nephrology and TransplantationErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| | - Dominique M. Bovée
- Department of Internal Medicine, Division of Nephrology and TransplantationErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| | - Ricardo P.J. Budde
- Department of Radiology and Nuclear MedicineErasmus Medical Center, University Medical Center RotterdamRotterdamThe Netherlands
| | - Jacqueline van de Wetering
- Department of Internal Medicine, Division of Nephrology and TransplantationErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| | - Joost G.J. Hoenderop
- Department of PhysiologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Martin E. van Royen
- Department of PathologyErasmus Medical Center, University Medical Center RotterdamRotterdamThe Netherlands
| | - Robert Zietse
- Department of Internal Medicine, Division of Nephrology and TransplantationErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| | - David Severs
- Department of Internal Medicine, Division of Nephrology and TransplantationErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| | - Ewout J. Hoorn
- Department of Internal Medicine, Division of Nephrology and TransplantationErasmus MC, University Medical Center RotterdamRotterdamThe Netherlands
| |
Collapse
|
12
|
Yates AG, Pink RC, Erdbrügger U, Siljander PR, Dellar ER, Pantazi P, Akbar N, Cooke WR, Vatish M, Dias‐Neto E, Anthony DC, Couch Y. In sickness and in health: The functional role of extracellular vesicles in physiology and pathology in vivo: Part I: Health and Normal Physiology: Part I: Health and Normal Physiology. J Extracell Vesicles 2022; 11:e12151. [PMID: 35041249 PMCID: PMC8765331 DOI: 10.1002/jev2.12151] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
Previously thought to be nothing more than cellular debris, extracellular vesicles (EVs) are now known to mediate physiological and pathological functions throughout the body. We now understand more about their capacity to transfer nucleic acids and proteins between distant organs, the interaction of their surface proteins with target cells, and the role of vesicle-bound lipids in health and disease. To date, most observations have been made in reductionist cell culture systems, or as snapshots from patient cohorts. The heterogenous population of vesicles produced in vivo likely act in concert to mediate both beneficial and detrimental effects. EVs play crucial roles in both the pathogenesis of diseases, from cancer to neurodegenerative disease, as well as in the maintenance of system and organ homeostasis. This two-part review draws on the expertise of researchers working in the field of EV biology and aims to cover the functional role of EVs in physiology and pathology. Part I will outline the role of EVs in normal physiology.
Collapse
Affiliation(s)
- Abi G. Yates
- Department of PharmacologyUniversity of OxfordOxfordUK
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandSt LuciaAustralia
| | - Ryan C. Pink
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Uta Erdbrügger
- Department of Medicine, Division of NephrologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Pia R‐M. Siljander
- Molecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Elizabeth R. Dellar
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Paschalia Pantazi
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - William R. Cooke
- Nuffield Department of Women's and Reproductive HealthUniversity of OxfordOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive HealthUniversity of OxfordOxfordUK
| | - Emmanuel Dias‐Neto
- Laboratory of Medical Genomics. A.C. Camargo Cancer CentreSão PauloBrazil
- Laboratory of Neurosciences (LIM‐27) Institute of PsychiatrySão Paulo Medical SchoolSão PauloBrazil
| | | | - Yvonne Couch
- Acute Stroke Programme ‐ Radcliffe Department of MedicineUniversity of OxfordJohn Radcliffe Hospital, HeadingtonOxfordUK
| |
Collapse
|
13
|
Myette RL, Burger D. Relapse in steroid-sensitive nephrotic syndrome: are extracellular vesicles the missing link? Am J Physiol Renal Physiol 2021; 321:F656-F658. [PMID: 34569254 DOI: 10.1152/ajprenal.00349.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Robert L Myette
- Division of Nephrology, Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada.,Kidney Research Centre, Department of Cellular and Molecular Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Dylan Burger
- Kidney Research Centre, Department of Cellular and Molecular Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
14
|
Török B, Fazekas CL, Szabó A, Zelena D. Epigenetic Modulation of Vasopressin Expression in Health and Disease. Int J Mol Sci 2021; 22:ijms22179415. [PMID: 34502322 PMCID: PMC8430944 DOI: 10.3390/ijms22179415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Vasopressin is a ubiquitous molecule playing an important role in a wide range of physiological processes thereby implicated in the pathomechanism of many disorders. Its effect is well characterized through V2 receptors, which regulates the water resorption in kidney, while its vasoconstrictory effect through V1a receptor also received a lot of attention in the maintenance of blood pressure during shock. However, the most striking is its central effect both through the V1b receptors in stress-axis regulation as well as through V1a receptors regulating many aspects of our behavior (e.g., social behavior, learning and memory). Vasopressin has been implicated in the development of depression, due to its connection with chronic stress, as well as schizophrenia because of its involvement in social interactions and memory processes. Epigenetic changes may also play a role in the development of these disorders. The possible mechanism includes DNA methylation, histone modification and/or micro RNAs, and these possible regulations will be in the focus of our present review.
Collapse
Affiliation(s)
- Bibiána Török
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (B.T.); (C.L.F.); (A.S.)
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Csilla Lea Fazekas
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (B.T.); (C.L.F.); (A.S.)
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Adrienn Szabó
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (B.T.); (C.L.F.); (A.S.)
- János Szentágothai School of Neurosciences, Semmelweis University, 1085 Budapest, Hungary
| | - Dóra Zelena
- Institute of Experimental Medicine, 1083 Budapest, Hungary; (B.T.); (C.L.F.); (A.S.)
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
- Correspondence:
| |
Collapse
|
15
|
Wu Q, Fenton RA. Urinary proteomics for kidney dysfunction: insights and trends. Expert Rev Proteomics 2021; 18:437-452. [PMID: 34187288 DOI: 10.1080/14789450.2021.1950535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Introduction: Kidney dysfunction poses a high burden on patients and health care systems. Early detection and accurate prediction of kidney disease progression remains a major challenge. Compared to existing clinical parameters, urinary proteomics has the potential to reveal molecular alterations within the kidney that may alter its function before the onset of clinical symptoms. Thus, urinary proteomics has greater prognostic potential for assessment of kidney dysfunction progression.Areas covered: Advances in urinary proteomics for major causes of kidney dysfunction are discussed. The application of urinary extracellular vesicles for studying kidney dysfunction are discussed. Technological advances in urinary proteomics are discussed. The literature was identified using a database search for titles containing 'proteom*' and 'urin*' and published within the past 5 years. Retrieved literature was manually filtered to retain kidney dysfunctions-related studies.Expert opinion: Despite major advances, diagnosis by urinary proteomics has not been fully applied in any clinical settings. This could be attributed to the complex nature of kidney diseases, in addition to the constraints on study power and feasibility of incorporating mass spectrometry techniques in daily routine analysis. Nevertheless, we are confident that advances in urinary proteomics will soon provide superior insights into kidney disease beyond existing clinical parameters.
Collapse
Affiliation(s)
- Qi Wu
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
16
|
Kumar P, Zadjali F, Yao Y, Siroky B, Astrinidis A, Gross KW, Bissler JJ. Tsc Gene Locus Disruption and Differences in Renal Epithelial Extracellular Vesicles. Front Physiol 2021; 12:630933. [PMID: 34262466 PMCID: PMC8273388 DOI: 10.3389/fphys.2021.630933] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/29/2021] [Indexed: 12/31/2022] Open
Abstract
In tuberous sclerosis complex (TSC), Tsc2 mutations are associated with more severe disease manifestations than Tsc1 mutations and the role of extracellular vesicles (EVs) in this context is not yet studied. We report a comparative analysis of EVs derived from isogenic renal cells except for Tsc1 or Tsc2 gene status and hypothesized that in spite of having similar physical characteristics, EVs modulate signaling pathways differently, thus leading to TSC heterogenicity. We used mouse inner medullary collecting duct (mIMCD3) cells with the Tsc1 (T1G cells) or Tsc2 (T2J cells) gene disrupted by CRISPR/CAS9. EVs were isolated from the cell culture media by size-exclusion column chromatography followed by detailed physical and chemical characterization. Physical characterization of EVs was accessed by tunable resistive pulse sensing and dynamic light scattering, revealing similar average sizes and zeta potentials (at pH 7.4) for EVs from mIMCD3 (123.5 ± 5.7 nm and −16.3 ± 2.1 mV), T1G cells (131.5 ± 8.3 nm and −19.8 ± 2.7 mV), and T2J cells (127.3 ± 4.9 nm and −20.2 ± 2.1 mV). EVs derived from parental mIMCD3 cells and both mutated cell lines were heterogeneous (>90% of EVs < 150 nm) in nature. Immunoblotting detected cilial Hedgehog signaling protein Arl13b; intercellular proteins TSG101 and Alix; and transmembrane proteins CD63, CD9, and CD81. Compared to Tsc2 deletion, Tsc1 deletion cells had reduced EV production and release rates. EVs from Tsc1 mutant cells altered mTORC1, autophagy, and β-catenin pathways differently than EVs from Tsc2-mutated cells. Quantitative PCR analysis revealed the down regulation of miR-212a-3p and miR-99a-5p in EVs from Tsc2-mutated cells compared to EVs from Tsc1-mutant cells. Thus, EV-derived miR-212-3p and mIR-99a-5p axes may represent therapeutic targets or biomarkers for TSC disease.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN, United States
| | - Fahad Zadjali
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN, United States.,Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Ying Yao
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN, United States
| | - Brian Siroky
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN, United States
| | - Aristotelis Astrinidis
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN, United States
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - John J Bissler
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN, United States.,Department of Pediatrics, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
17
|
Erdbrügger U, Blijdorp CJ, Bijnsdorp IV, Borràs FE, Burger D, Bussolati B, Byrd JB, Clayton A, Dear JW, Falcón‐Pérez JM, Grange C, Hill AF, Holthöfer H, Hoorn EJ, Jenster G, Jimenez CR, Junker K, Klein J, Knepper MA, Koritzinsky EH, Luther JM, Lenassi M, Leivo J, Mertens I, Musante L, Oeyen E, Puhka M, van Royen ME, Sánchez C, Soekmadji C, Thongboonkerd V, van Steijn V, Verhaegh G, Webber JP, Witwer K, Yuen PS, Zheng L, Llorente A, Martens‐Uzunova ES. Urinary extracellular vesicles: A position paper by the Urine Task Force of the International Society for Extracellular Vesicles. J Extracell Vesicles 2021; 10:e12093. [PMID: 34035881 PMCID: PMC8138533 DOI: 10.1002/jev2.12093] [Citation(s) in RCA: 188] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/26/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
Urine is commonly used for clinical diagnosis and biomedical research. The discovery of extracellular vesicles (EV) in urine opened a new fast-growing scientific field. In the last decade urinary extracellular vesicles (uEVs) were shown to mirror molecular processes as well as physiological and pathological conditions in kidney, urothelial and prostate tissue. Therefore, several methods to isolate and characterize uEVs have been developed. However, methodological aspects of EV separation and analysis, including normalization of results, need further optimization and standardization to foster scientific advances in uEV research and a subsequent successful translation into clinical practice. This position paper is written by the Urine Task Force of the Rigor and Standardization Subcommittee of ISEV consisting of nephrologists, urologists, cardiologists and biologists with active experience in uEV research. Our aim is to present the state of the art and identify challenges and gaps in current uEV-based analyses for clinical applications. Finally, recommendations for improved rigor, reproducibility and interoperability in uEV research are provided in order to facilitate advances in the field.
Collapse
|
18
|
Mazzariol M, Camussi G, Brizzi MF. Extracellular Vesicles Tune the Immune System in Renal Disease: A Focus on Systemic Lupus Erythematosus, Antiphospholipid Syndrome, Thrombotic Microangiopathy and ANCA-Vasculitis. Int J Mol Sci 2021; 22:ijms22084194. [PMID: 33919576 PMCID: PMC8073859 DOI: 10.3390/ijms22084194] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 01/02/2023] Open
Abstract
Extracellular vesicles (EV) are microparticles released in biological fluids by different cell types, both in physiological and pathological conditions. Owing to their ability to carry and transfer biomolecules, EV are mediators of cell-to-cell communication and are involved in the pathogenesis of several diseases. The ability of EV to modulate the immune system, the coagulation cascade, the angiogenetic process, and to drive endothelial dysfunction plays a crucial role in the pathophysiology of both autoimmune and renal diseases. Recent studies have demonstrated the involvement of EV in the control of renal homeostasis by acting as intercellular signaling molecules, mediators of inflammation and tissue regeneration. Moreover, circulating EV and urinary EV secreted by renal cells have been investigated as potential early biomarkers of renal injury. In the present review, we discuss the recent findings on the involvement of EV in autoimmunity and in renal intercellular communication. We focused on EV-mediated interaction between the immune system and the kidney in autoimmune diseases displaying common renal damage, such as antiphospholipid syndrome, systemic lupus erythematosus, thrombotic microangiopathy, and vasculitis. Although further studies are needed to extend our knowledge on EV in renal pathology, a deeper investigation of the impact of EV in kidney autoimmune diseases may also provide insight into renal biological processes. Furthermore, EV may represent promising biomarkers of renal diseases with potential future applications as diagnostic and therapeutic tools.
Collapse
|
19
|
Extracellular RNA in kidney disease: moving slowly but surely from bench to bedside. Clin Sci (Lond) 2021; 134:2893-2895. [PMID: 33185689 PMCID: PMC7672254 DOI: 10.1042/cs20201092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
We have known for just over a decade that functional RNA is shuttled between cells (Nat. Cell Biol. (2007) 9, 654–659). In that short time, there have been countless reports of extracellular RNA (exRNA) and extracellular vesicles (EVs) participating in diverse biological processes in development (Dev. Cell (2017) 40, 95–103), homoeostasis (Nature (2017) 542, 450–455) and disease (Nature (2017) 546, 498–503). Unsurprisingly – as these disciplines are still in their infancies – most of this work is still in the ‘discovery biology’ phase. However, exRNA and EVs show promise as disease biomarkers and could be harnessed in novel therapies.
Collapse
|
20
|
Urinary MicroRNAs in Environmental Health: Biomarkers of Emergent Kidney Injury and Disease. Curr Environ Health Rep 2021; 7:101-108. [PMID: 32166731 DOI: 10.1007/s40572-020-00271-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW There is a critical need for sensitive biomarkers of renal disease and progression. Micro(mi)RNAs are attractive as next-generation biomarkers in kidney disease, particularly as urine miRNAs can inform kidney function and cellular integrity. This review summarizes recent epidemiologic and toxicologic advances using urinary miRNAs and exosomal miRNAs as novel biomarkers of chemical exposure and of kidney damage and disease. RECENT FINDINGS Urine miRNA biomarkers offer improved stability over protein in stored samples, relative ease of collection and quantitation, and conserved sequence homology across species. Particularly in the case of emergent environmental health threats such as chronic kidney disease of unknown origin, urinary miRNAs hold promise as biomarkers of disease and/or exposure. We present evidence to address scientific knowledge gaps, comment on the relevance of urine-derived miRNAs in environmental health research, and discuss limitations and recommendations for future directions needed to advance miRNA biomarker strategies.
Collapse
|
21
|
Panfoli I, Granata S, Candiano G, Verlato A, Lombardi G, Bruschi M, Zaza G. Analysis of urinary exosomes applications for rare kidney disorders. Expert Rev Proteomics 2021; 17:735-749. [PMID: 33395324 DOI: 10.1080/14789450.2020.1866993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Exosomes are nanovesicles that play important functions in a variety of physiological and pathological conditions. They are powerful cell-to-cell communication tool thanks to the protein, mRNA, miRNA, and lipid cargoes they carry. They are also emerging as valuable diagnostic and prognostic biomarker sources. Urinary exosomes carry information from all the cells of the urinary tract, downstream of the podocyte. Rare kidney diseases are a subset of an inherited diseases whose genetic diagnosis can be unclear, and presentation can vary due to genetic, epigenetic, and environmental factors. Areas covered: In this review, we focus on a group of rare and often neglected kidney diseases, for which we have sufficient available literature data on urinary exosomes. The analysis of their content can help to comprehend pathological mechanisms and to identify biomarkers for diagnosis, prognosis, and therapeutic targets. Expert opinion: The foreseeable large-scale application of system biology approach to the profiling of exosomal proteins as a source of renal disease biomarkers will be also useful to stratify patients with rare kidney diseases whose penetrance, phenotypic presentation, and age of onset vary sensibly. This can ameliorate the clinical management.
Collapse
Affiliation(s)
- Isabella Panfoli
- Department of Pharmacy-DIFAR, University of Genoa , Genoa, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini , Genoa, Italy
| | - Alberto Verlato
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Gianmarco Lombardi
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini , Genoa, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| |
Collapse
|
22
|
Raby KL, Horsely H, McCarthy-Boxer A, Norman JT, Wilson PD. Urinary exosome proteomic profiling defines stage-specific rapid progression of autosomal dominant polycystic kidney disease and tolvaptan efficacy. BBA ADVANCES 2021; 1:100013. [PMID: 37082007 PMCID: PMC10074914 DOI: 10.1016/j.bbadva.2021.100013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 11/17/2022] Open
Abstract
ADPKD is the most common genetic disease of the kidney leading to end-stage renal disease necessitating renal replacement therapy at any time between the 1st and 8th decades of life due to widely variable rates of disease progression. This presents significant patient anxiety and a significant prognostic and therapeutic challenge. Tolvaptan is the only approved drug licensed to slow ADPKD progression by reducing renal cystic expansion but side-effects can limit its efficacy. To address the need to identify new biomarkers to monitor progression of ADPKD and to evaluate the therapeutic effects of Tolvaptan, proteomic analysis was conducted on defined (40-100nm) urinary exosomes isolated from ADPKD patients phenotyped and clinically monitored over a 10-year period. Comparative Gene Ontology analysis of Tandem Mass Tag labelled mass spectrometry-derived protein profiles from urinary exosomes from ADPKD patients with rapid (>10ml/min/5 years decline in estimated glomerular filtration rate) versus slow progression showed distinctive patterns of pathway up-regulation. Clear discrimination between rapid and slowly-progressive profiles were seen in all stages functional decline in ADPKD patients whether with mild (>70ml/min), moderate (50-69ml/min) or severe (<49ml/min) disease at onset. Discriminatory pathways and proteins included Notch-, integrin- and growth factor-signalling; microtubular kinase, vesicular proteins and epidermal growth factor substrates. Confocal microscopy of fluorescently-labelled normal versus ADPKD epithelial cell-derived exosomes in vitro also identified ADPKD-dependent abnormalities in intracellular vesicular trafficking and implicated changes in ADPKD-dependent exosome secretion and target cell uptake as factors underlying urinary exosome excretion biomarker properties. Comparative proteomic analysis of urinary exosomal proteins in individual patients before and after treatment with Tolvaptan for 4 years also identified distinct patterns of pathway modification dependent on the degree of effectiveness of the therapeutic response. Up-regulation of Wnt-pathway and vesicular proteins were characteristic of urinary exosomes from ADPKD patients with good responses to Tolvaptan while upregulation of angiogenesis pathways and additional molecular forms of vasopressin receptor AVPR2 were characteristic in urinary exosomes of ADPKD patients with poor responses. Taken together, these studies conclude that proteomic profiling of urinary exosome biomarkers provides a specific, sensitive and practical non-invasive method to identify and monitor the rate of disease progression and the effects of Tolvaptan therapy in individual ADPKD patients. This provides a means to identify those patients most likely to benefit maximally from therapy and to progress towards a personalization of ADPKD prognosis and management.
Collapse
Affiliation(s)
| | | | | | | | - Patricia D. Wilson
- Corresponding author at: University College London, Department of Renal Medicine, 2 Floor, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, United Kingdom.
| |
Collapse
|
23
|
Matthews O, Morrison EE, Tranter JD, Starkey Lewis P, Toor IS, Srivastava A, Sargeant R, Rollison H, Matchett KP, Kendall TJ, Gray GA, Goldring C, Park K, Denby L, Dhaun N, Bailey MA, Henderson NC, Williams D, Dear JW. Transfer of hepatocellular microRNA regulates cytochrome P450 2E1 in renal tubular cells. EBioMedicine 2020; 62:103092. [PMID: 33232872 PMCID: PMC7689533 DOI: 10.1016/j.ebiom.2020.103092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Extracellular microRNAs enter kidney cells and modify gene expression. We used a Dicer-hepatocyte-specific microRNA conditional-knock-out (Dicer-CKO) mouse to investigate microRNA transfer from liver to kidney. METHODS Dicerflox/flox mice were treated with a Cre recombinase-expressing adenovirus (AAV8) to selectively inhibit hepatocyte microRNA production (Dicer-CKO). Organ microRNA expression was measured in health and following paracetamol toxicity. The functional consequence of hepatic microRNA transfer was determined by measuring the expression and activity of cytochrome P450 2E1 (target of the hepatocellular miR-122), and by measuring the effect of serum extracellular vesicles (ECVs) on proximal tubular cell injury. In humans with liver injury we measured microRNA expression in urinary ECVs. A murine model of myocardial infarction was used as a non-hepatic model of microRNA release. FINDINGS Dicer-CKO mice demonstrated a decrease in kidney miR-122 in the absence of other microRNA changes. During hepatotoxicity, miR-122 increased in kidney tubular cells; this was abolished in Dicer-CKO mice. Depletion of hepatocyte microRNA increased kidney cytochrome P450 2E1 expression and activity. Serum ECVs from mice with hepatotoxicity increased proximal tubular cell miR-122 and prevented cisplatin toxicity. miR-122 increased in urinary ECVs during human hepatotoxicity. Transfer of microRNA was not restricted to liver injury -miR-499 was released following cardiac injury and correlated with an increase in the kidney. INTERPRETATION Physiological transfer of functional microRNA to the kidney is increased by liver injury and this signalling represents a new paradigm for understanding the relationship between liver injury and renal function. FUNDING Kidney Research UK, Medical Research Scotland, Medical Research Council.
Collapse
Affiliation(s)
- Olivia Matthews
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Emma E Morrison
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - John D Tranter
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Philip Starkey Lewis
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, United Kingdom
| | - Iqbal S Toor
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Abhishek Srivastava
- AstraZeneca, Clinical Pharmacology & Safety Sciences Department, Biopharmaceuticals Science Unit, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge, CB4 0FZ. United Kingdom
| | - Rebecca Sargeant
- AstraZeneca, Clinical Pharmacology & Safety Sciences Department, Biopharmaceuticals Science Unit, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge, CB4 0FZ. United Kingdom
| | - Helen Rollison
- AstraZeneca, Clinical Pharmacology & Safety Sciences Department, Biopharmaceuticals Science Unit, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge, CB4 0FZ. United Kingdom
| | - Kylie P Matchett
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Timothy J Kendall
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Gillian A Gray
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Chris Goldring
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, United Kingdom
| | - Kevin Park
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, United Kingdom
| | - Laura Denby
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Neeraj Dhaun
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Matthew A Bailey
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom; MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, United Kingdom
| | - Dominic Williams
- AstraZeneca, Clinical Pharmacology & Safety Sciences Department, Biopharmaceuticals Science Unit, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge, CB4 0FZ. United Kingdom
| | - James W Dear
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom.
| |
Collapse
|
24
|
Li M, Jiang M, Meng J, Tao L. Exosomes: Carriers of Pro-Fibrotic Signals and Therapeutic Targets in Fibrosis. Curr Pharm Des 2020; 25:4496-4509. [PMID: 31814552 DOI: 10.2174/1381612825666191209161443] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
Abstract
Exosomes are nano-sized extracellular vesicles that are released by a variety of cells. Exosomes contain cargo from cells they derived, including lipids, proteins and nucleic acids. The bilayer lipid membrane structure of exosomes protects these contents from degradation, allowing them for intercellular communication. The role of exosomes in fibrotic diseases is increasingly being valued. Exosomes, as carriers of profibrotic signals, are involved in the development of fibrotic diseases, and also regulate fibrosis by transmitting signals that inhibit fibrosis or inflammation. Exosomes mobilize and activate a range of effector cells by targeted delivery of bioactive information. Exosomes can also reflect the condition of cells, tissues and organisms, and thus become potential biomarkers of fibrotic diseases. Exosomes from bone marrow stem cells support biological signaling that regulates and inhibits fibrosis and thus initially used in the treatment of fibrotic diseases. This article briefly summarizes the role of exosomes in the pathogenesis and treatment of fibrotic diseases and raises some issues that remain to be resolved.
Collapse
Affiliation(s)
- Mengyu Li
- Department of Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, 932 Lushans Rd, Yuela, Changsha, Hunan, China.,Organ Fibrosis Research Center, Central South University, 932 Lushans Rd, Yuela, Changsha, Hunan, China
| | - Mao Jiang
- Department of Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, 932 Lushans Rd, Yuela, Changsha, Hunan, China.,Organ Fibrosis Research Center, Central South University, 932 Lushans Rd, Yuela, Changsha, Hunan, China
| | - Jie Meng
- Department of Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, 932 Lushans Rd, Yuela, Changsha, Hunan, China.,Organ Fibrosis Research Center, Central South University, 932 Lushans Rd, Yuela, Changsha, Hunan, China
| | - Lijian Tao
- Organ Fibrosis Research Center, Central South University, 932 Lushans Rd, Yuela, Changsha, Hunan, China.,Department of Nephrology, Xiangya Hospital, Central South University, 932 Lushans Rd, Yuela, Changsha, Hunan, China
| |
Collapse
|
25
|
Ishige F, Hoshino I, Iwatate Y, Chiba S, Arimitsu H, Yanagibashi H, Nagase H, Takayama W. MIR1246 in body fluids as a biomarker for pancreatic cancer. Sci Rep 2020; 10:8723. [PMID: 32457495 PMCID: PMC7250935 DOI: 10.1038/s41598-020-65695-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 05/08/2020] [Indexed: 01/28/2023] Open
Abstract
Pancreatic cancer is an aggressive tumor associated with poor survival, and early detection is important to improve patient outcomes. In the present study, we examined MIR1246 expression as a biomarker of pancreatic cancer. Total RNA was extracted from serum, urine and saliva samples from healthy subjects (n = 30) and patients with pancreatic cancer (n = 41, stage 0–IV). The MIR1246 level in each fluid was analyzed by quantitative reverse transcription-polymerase chain reaction. Significantly higher MIR1246 expression in serum and urine was observed in patients with cancer than in healthy controls. A significant positive correlation was found between serum and urine MIR1246 expression (r = 0.34). Receiver operating characteristic curves were constructed for MIR1246 in all three body fluids. The area under the curve for serum MIR1246 was 0.87 (sensitivity, 92.3%; specificity, 73.3%), and that for urine MIR1246 was 0.90 (sensitivity, 90.2%; specificity, 83.3%). With a cut-off of the control group’s mean plus twice the standard deviation, the sensitivities of MIR1246 in serum and urine for pancreatic cancer were 60.9 and 58.5%, respectively. Combining both serum and urine MIR1246 expression yielded a sensitivity of 85%. These results indicate that MIR246 may be a useful diagnostic biomarker for pancreatic cancer.
Collapse
Affiliation(s)
- Fumitaka Ishige
- Division of Hepatobiliarypancreatic Surgery, Chiba Cancer Center, Chuo-ku, Chiba, Japan
| | - Isamu Hoshino
- Division of Gastrointestinal Surgery, Chiba Cancer Center, Chuo-ku, Chiba, Japan.
| | - Yosuke Iwatate
- Division of Hepatobiliarypancreatic Surgery, Chiba Cancer Center, Chuo-ku, Chiba, Japan
| | - Satoshi Chiba
- Division of Hepatobiliarypancreatic Surgery, Chiba Cancer Center, Chuo-ku, Chiba, Japan
| | - Hidehito Arimitsu
- Division of Hepatobiliarypancreatic Surgery, Chiba Cancer Center, Chuo-ku, Chiba, Japan
| | - Hiroo Yanagibashi
- Division of Hepatobiliarypancreatic Surgery, Chiba Cancer Center, Chuo-ku, Chiba, Japan
| | - Hiroki Nagase
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chuo-ku, Chiba, Japan
| | - Wataru Takayama
- Division of Hepatobiliarypancreatic Surgery, Chiba Cancer Center, Chuo-ku, Chiba, Japan
| |
Collapse
|
26
|
Georgatzakou HT, Pavlou EG, Papageorgiou EG, Papassideri IS, Kriebardis AG, Antonelou MH. The Multi-Faced Extracellular Vesicles in the Plasma of Chronic Kidney Disease Patients. Front Cell Dev Biol 2020; 8:227. [PMID: 32351956 PMCID: PMC7174738 DOI: 10.3389/fcell.2020.00227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/17/2020] [Indexed: 11/13/2022] Open
Abstract
Extracellular vesicles (EVs) are membrane-enclosed nanoparticles released by most cells in body fluids and extracellular matrix. They function as signal transducers in intercellular communication, contributing to the maintenance of cell and tissue integrity. EVs biogenesis is deregulated in various pathologies, in structural and functional connection to the pathophysiology of donor cells. Consequently, EVs are considered diagnostic and monitoring factors in many diseases. Despite consensus as to their activity in promoting coagulation and inflammation, there is evidence suggesting protective roles for EVs in stress states. Chronic kidney disease (CKD) patients are at high risk of developing cardiovascular defects. The pathophysiology, comorbidities, and treatment of CKD may individually and in synergy affect extracellular vesiculation in the kidney, endothelium, and blood cells. Oxidative and mechanical stresses, chronic inflammation, and deregulation of calcium and phosphate homeostasis are established stressors of EV release. EVs may affect the clinical severity of CKD by transferring biological response modifiers between renal, vascular, blood, and inflammatory cells. In this Review, we focus on EVs circulating in the plasma of CKD patients. We highlight some recent advances in the understanding of their biogenesis, the effects of dialysis, and pharmacological treatments on them and their potential impact on thrombosis and vascular defects. The strong interest of the scientific community to this exciting field of research may reveal hidden pieces in the pathophysiology of CKD and thus, innovative ways to treat it. Overcoming gaps in EV biology and technical difficulties related to their size and heterogeneity will define the success of the project.
Collapse
Affiliation(s)
- Hara T Georgatzakou
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica, Athens, Greece
| | - Efthimia G Pavlou
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica, Athens, Greece
| | - Effie G Papageorgiou
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica, Athens, Greece
| | - Issidora S Papassideri
- Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Anastasios G Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, School of Health & Welfare Sciences, University of West Attica, Athens, Greece
| | - Marianna H Antonelou
- Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| |
Collapse
|
27
|
Dahiya B, Khan A, Mor P, Kamra E, Singh N, Gupta KB, Sheoran A, Sreenivas V, Mehta PK. Detection of Mycobacterium tuberculosis lipoarabinomannan and CFP-10 (Rv3874) from urinary extracellular vesicles of tuberculosis patients by immuno-PCR. Pathog Dis 2020; 77:5565047. [PMID: 31549171 DOI: 10.1093/femspd/ftz049] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 09/05/2019] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs), the small circulating vesicles released from urine samples of tuberculosis (TB) patients, contain a pool of biomarkers. We recently detected Mycobacterium tuberculosis lipoarabinomannan (LAM) and CFP-10 (Rv3874) biomarkers from the urinary EVs of pulmonary TB (PTB) and extrapulmonary TB (EPTB) patients by immuno-polymerase chain reaction (I-PCR) assay and the results were compared with the analogous enzyme-linked immunosorbent assay (ELISA). The detection limits of both purified LAM and CFP-10 were determined to be 1 fg/mL with I-PCR, which was 106 times lower than ELISA. Detection of LAM and CFP-10 biomarkers in urinary EVs of TB patients by I-PCR showed superiority over ELISA. Notably, LAM I-PCR revealed sensitivities of 74.3 and 67.9% in PTB (n = 74) and EPTB (n = 53) patients, respectively, with specificities of 91.5-92.8% (n = 116). Moreover, the sensitivities attained with LAM I-PCR were significantly higher (P < 0.01) than with CFP-10 I-PCR. After further improving the sensitivity and specificity of the assay, our I-PCR based on LAM detection in urinary EVs may be used as an adjunct test for rapid diagnosis of TB.
Collapse
Affiliation(s)
- Bhawna Dahiya
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak-124001 (Haryana), India
| | - Anish Khan
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak-124001 (Haryana), India
| | - Preeti Mor
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak-124001 (Haryana), India
| | - Ekta Kamra
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak-124001 (Haryana), India
| | - Netrapal Singh
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak-124001 (Haryana), India
| | - Krishna B Gupta
- Department of TB & Respiratory Medicine, University of Health Sciences (UHS), Rohtak-124001, India
| | - Abhishek Sheoran
- Department of Statistics, Ramanujan College, University of Delhi, New Delhi-110019, India
| | - Vishnubhatla Sreenivas
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Promod K Mehta
- Centre for Biotechnology, Maharshi Dayanand University (MDU), Rohtak-124001 (Haryana), India
| |
Collapse
|
28
|
Rigalli JP, Barros ER, Sommers V, Bindels RJM, Hoenderop JGJ. Novel Aspects of Extracellular Vesicles in the Regulation of Renal Physiological and Pathophysiological Processes. Front Cell Dev Biol 2020; 8:244. [PMID: 32351960 PMCID: PMC7174565 DOI: 10.3389/fcell.2020.00244] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/23/2020] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EV) are nanosized particles released by a large variety of cells. They carry molecules such as proteins, RNA and lipids. While urinary EVs have been longer studied as a source of biomarkers for renal and non-renal disorders, research on EVs as regulatory players of renal physiological and pathological processes has experienced an outbreak recently in the past decade. In general, the microenvironment and (patho)physiological state of the donor cells affect the cargo of the EVs released, which then determines the effect of these EVs once they reach a target cell. For instance, EVs released by renal epithelial cells modulate the expression and function of water and solute transporting proteins in other cells. Also, EVs have been demonstrated to regulate renal organogenesis and blood flow. Furthermore, a dual role of EVs promoting, but also counteracting, disease has also been reported. EVs released by renal tubular cells can reach fibroblasts, monocytes, macrophages, T cells and natural killer cells, thus influencing the pathogenesis and progression of renal disorders like acute kidney injury and fibrosis, nephrolithiasis, renal transplant rejection and renal cancer, among others. On the contrary, EVs may also exert a cytoprotective role upon renal damage and promote recovery of renal function. In the current review, a systematic summary of the key studies from the past 5 years addressing the role of EVs in the modulation of renal physiological and pathophysiological processes is provided, highlighting open questions and discussing the potential of future research.
Collapse
Affiliation(s)
- Juan Pablo Rigalli
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Eric Raul Barros
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Vera Sommers
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
29
|
Hill N, Michell DL, Ramirez-Solano M, Sheng Q, Pusey C, Vickers KC, Woollard KJ. Glomerular endothelial derived vesicles mediate podocyte dysfunction: A potential role for miRNA. PLoS One 2020; 15:e0224852. [PMID: 32214346 PMCID: PMC7098579 DOI: 10.1371/journal.pone.0224852] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 02/26/2020] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNA) are shown to be involved in the progression of several types of kidney diseases. Podocytes maintain the integrity of the glomerular basement membrane. Extracellular vesicles (EV) are important in cell-to-cell communication as they can transfer cellular content between cells, including miRNA. However, little is known about how extracellular signals from the glomerular microenvironment regulate podocyte activity. Using a non-contact transwell system, communication between glomerular endothelial cells (GEnC) and podocytes was characterised in-vitro. Identification of transferred EV-miRNAs from GEnC to podocytes was performed using fluorescence cell tracking and miRNA mimetics. To represent kidney disease, podocyte molecular profiling and functions were analysed after EV treatments derived from steady state or activated GEnC. Our data shows activation of GEnC alters EV-miRNA loading, but activation was not found to alter EV secretion. EV delivery of miRNA to recipient podocytes altered cellular miRNA abundance and effector functions in podocytes, including decreased secretion of VEGF and increased mitochondrial stress which lead to altered cellular metabolism and cytoskeletal rearrangement. Finally, results support our hypothesis that miRNA-200c-3p is transfered by EVs from GEnC to podocytes in response to activation, ultimately leading to podocyte dysfunction.
Collapse
Affiliation(s)
- N. Hill
- Department of Medicine, Centre for Inflammatory Disease, Imperial College London, London, United Kingdom
| | - D. L. Michell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - M. Ramirez-Solano
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Q. Sheng
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - C. Pusey
- Department of Medicine, Centre for Inflammatory Disease, Imperial College London, London, United Kingdom
| | - K. C. Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - K. J. Woollard
- Department of Medicine, Centre for Inflammatory Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
30
|
The role of extracellular vesicles in renal fibrosis. Cell Death Dis 2019; 10:367. [PMID: 31068572 PMCID: PMC6506498 DOI: 10.1038/s41419-019-1605-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 12/15/2022]
Abstract
As a particularly important mediator of intercellular communication, extracellular vesicles (EVs) have been proved to be extensively involved in various system diseases over the past two decades, including in renal diseases. As is well-known, renal fibrosis is the common pathological process of any ongoing renal disease or adaptive repair of kidney injury based on current knowledge. Although much work has been performed focusing on EVs in various renal diseases, the role of EVs in renal fibrosis has not been described in detail and summarized. In this review, we provide a brief overview of the definition, classification and biological process of EVs. Then, the potential mechanisms of EVs in renal fibrosis are illustrated. Lastly, recent advances in EVs and the implications of EVs for diagnosis and therapy in renal fibrosis disease are introduced. We look forward to a more comprehensive understanding of EVs in renal fibrosis, which could be a boon to patients with renal fibrosis disease.
Collapse
|
31
|
Dominguez JM, Dominguez JH, Xie D, Kelly KJ. Human extracellular microvesicles from renal tubules reverse kidney ischemia-reperfusion injury in rats. PLoS One 2018; 13:e0202550. [PMID: 30148844 PMCID: PMC6110463 DOI: 10.1371/journal.pone.0202550] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 08/06/2018] [Indexed: 11/22/2022] Open
Abstract
Hypoxic acute kidney injury, a major unresolved problem, initiates, or aggravates, renal functional and structural decline. There is no treatment for hypoxic acute renal injury and its sequelae. We tested the hypothesis that human kidney tubular cells, or their extracellular vesicles (exosomes), prevent renal injury when infused intravenously 24 hours after 50 minutes of bilateral renal ischemia in Nude rats. Cells and their exosomes were from harvested human kidneys declined for transplantation. Injections of either cells or exosomes, given after 24 and 48 hours of reperfusion, preserved renal function and structure in both treatment groups. However, exosomes were superior to cells; and maintained renal vascular and epithelial networks, prevented renal oxidant stress, and apoptosis; and restrained activation of pro-inflammatory and pro-fibrogenic pathways. Exosomes worked in 24 hours, consistent with functional rather than regenerative activity. Comprehensive proteomic analysis identified 6152 renal proteins from all cellular compartments; and 628 were altered by ischemia at all cell levels, while 377 were significantly improved by exosome infusions. We conclude that renal damage from severe ischemia was broad, and human renal exosomes prevented most protein alterations. Thus, exosomes seem to acutely correct a critical and consequential abnormality during reperfusion. In their absence, renal structure and cells transition to a chronic state of fibrosis and extensive renal cell loss.
Collapse
Affiliation(s)
- James M. Dominguez
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Jesus H. Dominguez
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
- Roudebush VA Medical Center, Indianapolis, IN, United States of America
- * E-mail:
| | - Danhui Xie
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - K. J. Kelly
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
- Roudebush VA Medical Center, Indianapolis, IN, United States of America
| |
Collapse
|
32
|
Abbasian N, Herbert KE, Pawluczyk I, Burton JO, Bevington A. Vesicles bearing gifts: the functional importance of micro-RNA transfer in extracellular vesicles in chronic kidney disease. Am J Physiol Renal Physiol 2018; 315:F1430-F1443. [PMID: 30110570 DOI: 10.1152/ajprenal.00318.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), including microparticles (MPs) and exosomes (EXOs), are derived from a wide range of mammalian cells including blood platelets, endothelial cells, and kidney cells and can be detected in body fluids including blood and urine. While EVs are well established as diagnostic markers under pathophysiological and stress conditions, there is also mounting evidence of their functional significance as vehicles for communication between cells mediated by the presence of nucleic acids, especially microRNAs (miRs), encapsulated in the EVs. miRs regulate gene expression, are transported both in MPs and EXOs, and exert profound effects in the kidney. Here we review current understanding of the links between EVs and miRs, discuss the importance of miRs in kidney disease, and shed light on the role of EVs in transferring miRs through the circulation among the renal, vascular, and inflammatory cell populations that are functionally important in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Nima Abbasian
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - Karl E Herbert
- Department of Cardiovascular Sciences, University of Leicester, and Leicester National Institute of Health Research Cardiovascular Biomedical Research Unit , Leicester , United Kingdom
| | - Izabella Pawluczyk
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| | - James O Burton
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom.,John Walls Renal Unit, University Hospitals of Leicester , Leicester , United Kingdom
| | - Alan Bevington
- Department of Infection, Immunity, and Inflammation, University of Leicester , Leicester , United Kingdom
| |
Collapse
|
33
|
Xu WC, Qian G, Liu AQ, Li YQ, Zou HQ. Urinary Extracellular Vesicle: A Potential Source of Early Diagnostic and Therapeutic Biomarker in Diabetic Kidney Disease. Chin Med J (Engl) 2018; 131:1357-1364. [PMID: 29786051 PMCID: PMC5987509 DOI: 10.4103/0366-6999.232801] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE Diabetic kidney disease (DKD) has become one of the major causes of end-stage renal disease. Urinary extracellular vesicles (uEVs) contain rich biological information which could be the ideal source for noninvasive biomarkers of DKD. This review discussed the potential early diagnostic and therapeutic values of proteins and microRNAs in uEVs in DKD. DATA SOURCES This review was based articles published in PubMed, Embase, Cochrane, and Google Scholar databases up to November 20, 2017, with the following keywords: "Diabetic kidney disease", "Extracellular vesicle", and "Urine". STUDY SELECTION Relevant articles were carefully reviewed, with no exclusions applied to the study design and publication type. RESULTS There is no "gold standard" technology to separate and/or purify uEVs. The uEVs contain a variety of proteins and RNAs and participate in the physiological and pathological processes of the kidney. UEVs, especially urinary exosomes, may be useful biomarkers for early diagnosis and treatment to DKD. Furthermore, the uEVs has been used as a therapeutic target for DKD. CONCLUSION Proteins and nucleic acids in uEVs represent promising biomarker for the diagnosis and treatment of DKD.
Collapse
Affiliation(s)
- Wei-Cheng Xu
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Ge Qian
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Ai-Qun Liu
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Yong-Qiang Li
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - He-Qun Zou
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
34
|
Schneider DJ, Speth JM, Penke LR, Wettlaufer SH, Swanson JA, Peters-Golden M. Mechanisms and modulation of microvesicle uptake in a model of alveolar cell communication. J Biol Chem 2017; 292:20897-20910. [PMID: 29101235 PMCID: PMC5743066 DOI: 10.1074/jbc.m117.792416] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 10/27/2017] [Indexed: 12/31/2022] Open
Abstract
Extracellular vesicles, including exosomes and shed microvesicles (MVs), can be internalized by recipient cells to modulate function. Although the mechanism by which extracellular vesicles are internalized is incompletely characterized, it is generally considered to involve endocytosis and an initial surface-binding event. Furthermore, modulation of uptake by microenvironmental factors is largely unstudied. Here, we used flow cytometry, confocal microscopy, and pharmacologic and molecular targeting to address these gaps in knowledge in a model of pulmonary alveolar cell-cell communication. Alveolar macrophage-derived MVs were fully internalized by alveolar epithelial cells in a time-, dose-, and temperature-dependent manner. Uptake was dependent on dynamin and actin polymerization. However, it was neither saturable nor dependent on clathrin or receptor binding. Internalization was enhanced by extracellular proteins but was inhibited by cigarette smoke extract via oxidative disruption of actin polymerization. We conclude that MV internalization occurs via a pathway more consistent with fluid-phase than receptor-dependent endocytosis and is subject to bidirectional modulation by relevant pathologic perturbations.
Collapse
Affiliation(s)
| | | | - Loka R Penke
- From the Division of Pulmonary and Critical Care Medicine
| | | | - Joel A Swanson
- Department of Microbiology and Immunology, and
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Marc Peters-Golden
- From the Division of Pulmonary and Critical Care Medicine,
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
35
|
Lv LL, Wu WJ, Feng Y, Li ZL, Tang TT, Liu BC. Therapeutic application of extracellular vesicles in kidney disease: promises and challenges. J Cell Mol Med 2017; 22:728-737. [PMID: 29083099 PMCID: PMC5783839 DOI: 10.1111/jcmm.13407] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized, membrane‐bound vesicles released from different cells. Recent studies have revealed that EVs may participate in renal tissue damage and regeneration through mediating inter‐nephron communication. Thus, the potential use of EVs as therapeutic vector has gained considerable interest. In this review, we will discuss the basic characteristics of EVs and its role in nephron cellular communication. Then, the application of EVs as therapeutic vector based on its natural content or as carriers of drug, in acute and chronic kidney injury, was discussed. Finally, perspectives and challenges of EVs in therapy of kidney disease were described.
Collapse
Affiliation(s)
- Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Wei-Jun Wu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Ye Feng
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
36
|
Holub JM. Small Scaffolds, Big Potential: Developing Miniature Proteins as Therapeutic Agents. Drug Dev Res 2017; 78:268-282. [PMID: 28799168 DOI: 10.1002/ddr.21408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022]
Abstract
Preclinical Research Miniature proteins are a class of oligopeptide characterized by their short sequence lengths and ability to adopt well-folded, three-dimensional structures. Because of their biomimetic nature and synthetic tractability, miniature proteins have been used to study a range of biochemical processes including fast protein folding, signal transduction, catalysis and molecular transport. Recently, miniature proteins have been gaining traction as potential therapeutic agents because their small size and ability to fold into defined tertiary structures facilitates their development as protein-based drugs. This research overview discusses emerging developments involving the use of miniature proteins as scaffolds to design novel therapeutics for the treatment and study of human disease. Specifically, this review will explore strategies to: (i) stabilize miniature protein tertiary structure; (ii) optimize biomolecular recognition by grafting functional epitopes onto miniature protein scaffolds; and (iii) enhance cytosolic delivery of miniature proteins through the use of cationic motifs that facilitate endosomal escape. These objectives are discussed not only to address challenges in developing effective miniature protein-based drugs, but also to highlight the tremendous potential miniature proteins hold for combating and understanding human disease. Drug Dev Res 78 : 268-282, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Justin M Holub
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, USA
| |
Collapse
|
37
|
Abstract
Extracellular vesicles, such as exosomes and microvesicles, are host cell-derived packages of information that allow cell-cell communication and enable cells to rid themselves of unwanted substances. The release and uptake of extracellular vesicles has important physiological functions and may also contribute to the development and propagation of inflammatory, vascular, malignant, infectious and neurodegenerative diseases. This Review describes the different types of extracellular vesicles, how they are detected and the mechanisms by which they communicate with cells and transfer information. We also describe their physiological functions in cellular interactions, such as in thrombosis, immune modulation, cell proliferation, tissue regeneration and matrix modulation, with an emphasis on renal processes. We discuss how the detection of extracellular vesicles could be utilized as biomarkers of renal disease and how they might contribute to disease processes in the kidney, such as in acute kidney injury, chronic kidney disease, renal transplantation, thrombotic microangiopathies, vasculitides, IgA nephropathy, nephrotic syndrome, urinary tract infection, cystic kidney disease and tubulopathies. Finally, we consider how the release or uptake of extracellular vesicles can be blocked, as well as the associated benefits and risks, and how extracellular vesicles might be used to treat renal diseases by delivering therapeutics to specific cells.
Collapse
Affiliation(s)
- Diana Karpman
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Klinikgatan 28, 22184 Lund, Sweden
| | - Anne-Lie Ståhl
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Klinikgatan 28, 22184 Lund, Sweden
| | - Ida Arvidsson
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Klinikgatan 28, 22184 Lund, Sweden
| |
Collapse
|
38
|
Abstract
Extracellular vesicles are a heterogeneous population of microparticles released by virtually all living cells which have been recently widely investigated in different biological fields. They are typically composed of two primary types (exosomes and microvesicles) and are recently commanding increasing attention as mediators of cellular signaling. Indeed, these vesicles can affect recipient cells by carrying and delivering complex cargos of biomolecules (including proteins, lipids and nucleic acids), protected from enzymatic degradation in the environment. Their importance has been demonstrated in the pathophysiology of several organs, in particular in kidney, where different cell types secrete extracellular vesicles that mediate their communication with downstream urinary tract cells. Over the past few years, evidence has been shown that vesicles participate in kidney development and normal physiology. Moreover, EVs are widely demonstrated to be implicated in cellular signaling during renal regenerative and pathological processes. Although many EV mechanisms are still poorly understood, in particular in kidney, the discovery of their role could help to shed light on renal biological processes which are so far elusive. Lastly, extracellular vesicles secreted by renal cells gather in urine, thus becoming a great resource for disease or recovery markers and a promising non-invasive diagnostic instrument for renal disease. In the present review, we discuss the most recent findings on the role of extracellular vesicles in renal physiopathology and their potential implication in diagnosis and therapy.
Collapse
Affiliation(s)
| | - Chiara Gai
- Stem Cell Laboratory, Department of Medical Sciences, University of TurinTurin, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of TurinTurin, Italy
| | - Giovanni Camussi
- Stem Cell Laboratory, Department of Medical Sciences, University of TurinTurin, Italy
| |
Collapse
|
39
|
Barros ER, Carvajal CA. Urinary Exosomes and Their Cargo: Potential Biomarkers for Mineralocorticoid Arterial Hypertension? Front Endocrinol (Lausanne) 2017; 8:230. [PMID: 28951728 PMCID: PMC5599782 DOI: 10.3389/fendo.2017.00230] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/23/2017] [Indexed: 12/13/2022] Open
Abstract
Arterial hypertension (AHT) currently affects approximately 40% of adults worldwide, and its pathological mechanisms are mainly related to renal, vascular, and endocrine systems. Steroid hormones as aldosterone and cortisol are highly relevant to human endocrine physiology, and also to endocrine hypertension. Pathophysiological conditions, such as primary aldosteronism, affect approximately 10% of patients diagnosed with AHT and are secondary to a high production of aldosterone, increasing the risk also for cardiovascular damage and heart diseases. Excess of aldosterone or cortisol increases the activity of the mineralocorticoid receptor (MR) in epithelial and non-epithelial cells. Current research in this field highlights the potential regulatory mechanisms of the MR pathway, including pre-receptor regulation of the MR (action of 11BHSD2), MR activating proteins, and the downstream genes/proteins sensitive to MR (e.g., epithelial sodium channel, NCC, NKCC2). Mineralocorticoid AHT is present in 15-20% of hypertensive subjects, but the mechanisms associated to this condition have been poorly described, due mainly to the absence of reliable biomarkers. In this way, steroids, peptides, and lately urinary exosomes are thought to be potential reporters of biological processes. This review highlight exosomes and their cargo as potential biomarkers of metabolic changes associated to mineralocorticoid AHT. Recent reports have shown the presence of RNA, microRNAs, and proteins in urinary exosomes, which could be used as biomarkers in physiological and pathophysiological conditions. However, more studies are needed in order to benefit from exosomes and the exosomal cargo as a diagnostic tool in mineralocorticoid AHT.
Collapse
Affiliation(s)
- Eric R. Barros
- Center of Translational Endocrinology (CETREN), Faculty of Medicine, Endocrinology Department, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian A. Carvajal
- Center of Translational Endocrinology (CETREN), Faculty of Medicine, Endocrinology Department, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Cristian A. Carvajal,
| |
Collapse
|
40
|
Rivoli L, Vliegenthart ADB, de Potter CMJ, van Bragt JJMH, Tzoumas N, Gallacher P, Farrah TE, Dhaun N, Dear JW. The effect of renal dysfunction and haemodialysis on circulating liver specific miR-122. Br J Clin Pharmacol 2016; 83:584-592. [PMID: 27650800 DOI: 10.1111/bcp.13136] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 08/26/2016] [Accepted: 09/16/2016] [Indexed: 12/31/2022] Open
Abstract
AIMS microRNA-122 (miR-122) is a hepatotoxicity biomarker with utility in the management of paracetamol overdose and in drug development. Renal dysfunction and haemodialysis have been associated with a reduction in circulating microRNA. The objective of this study was to determine their effect on miR-122. METHODS Blood samples were collected from 17 patients with end-stage renal disease (ESRD) on haemodialysis, 22 healthy controls, 30 patients with chronic kidney disease (CKD) and 15 patients post-kidney transplantation. All had normal standard liver function tests. Samples from ESRD patients were collected immediately pre- and post-haemodialysis. Serum alanine transaminase activity (ALT), miR-122 and miR-885 (liver enriched) were compared. RESULTS Circulating miR-122 was substantially reduced in ESRD patients pre-haemodialysis compared with the other groups (19.0-fold lower than healthy controls; 21.7-fold lower than CKD). Haemodialysis increased miR-122 from a median value of 6.7 × 103 (2.3 × 103 -1.4 × 104 ) to 1.6 × 104 (5.4 × 103 -3.2 × 104 ) copies ml-1 . The increase in miR-122 did not correlate with dialysis adequacy. miR-122 was reduced in the argonaute 2 bound fraction pre-haemodialysis; this fraction was increased post-dialysis. There was no change in miR-122 associated with extracellular vesicles. miR-885 was also reduced in ESRD patients (4-fold compared to healthy subjects) and increased by haemodialysis. CONCLUSION miR-122 is substantially lower in ESRD compared to healthy controls, patients with CKD and transplanted patients. Haemodialysis increases the concentration of miR-122. These data need to be considered when interpreting liver injury using miR-122 in patients with ESRD on dialysis, and specific reference ranges that define normal in this setting may need to be developed.
Collapse
Affiliation(s)
- Laura Rivoli
- Edinburgh University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh
| | - A D Bastiaan Vliegenthart
- Edinburgh University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh
| | - Carmelita M J de Potter
- Edinburgh University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh
| | - Job J M H van Bragt
- Edinburgh University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh
| | - Nikolaos Tzoumas
- Edinburgh University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh
| | - Peter Gallacher
- Edinburgh University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh
| | - Tariq E Farrah
- Edinburgh University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh
| | - Neeraj Dhaun
- Edinburgh University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh
| | - James W Dear
- Edinburgh University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, Edinburgh
| |
Collapse
|
41
|
Schneider DJ, Speth JM, Peters-Golden M. Signed, Sealed, Delivered: Microenvironmental Modulation of Extracellular Vesicle-Dependent Immunoregulation in the Lung. Front Cell Dev Biol 2016; 4:94. [PMID: 27626032 PMCID: PMC5004409 DOI: 10.3389/fcell.2016.00094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/18/2016] [Indexed: 12/26/2022] Open
Abstract
Unconventional secretion and subsequent uptake of molecular cargo via extracellular vesicles (EVs) is an important mechanism by which cells can exert paracrine effects. While this phenomenon has been widely characterized in the context of their ability to promote inflammation, less is known about the ability of EVs to transfer immunosuppressive cargo. Maintenance of normal physiology in the lung requires suppression of potentially damaging inflammatory responses to the myriad of insults to which it is continually exposed. Recently, our laboratory has reported the ability of alveolar macrophages (AMs) to secrete suppressors of cytokine signaling (SOCS) proteins within microvesicles (MVs) and exosomes (Exos). Uptake of these EVs by alveolar epithelial cells (AECs) resulted in inhibition of pro-inflammatory STAT activation in response to cytokines. Moreover, AM packaging of SOCS within EVs could be rapidly tuned in response to exogenous or AEC-derived substances. In this article we will highlight gaps in knowledge regarding microenvironmental modulation of cargo packaging and utilization as well as EV secretion and uptake. Advances in these areas are critical for improving understanding of intercellular communication in the immune system and for therapeutic application of artificial vesicles aimed at treatment of diseases characterized by dysregulated inflammation.
Collapse
Affiliation(s)
- Daniel J Schneider
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School USA
| | - Jennifer M Speth
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical SchoolUSA; Graduate Program in Immunology, University of Michigan Medical SchoolUSA
| |
Collapse
|
42
|
Extracellular vesicles in renal tissue damage and regeneration. Eur J Pharmacol 2016; 790:83-91. [PMID: 27375075 DOI: 10.1016/j.ejphar.2016.06.058] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/10/2016] [Accepted: 06/30/2016] [Indexed: 12/22/2022]
Abstract
Extracellular vesicles (EVs) appear as important actors in cell-to-cell communication. EV content is characterized by proteins and RNA species that dynamically reflect cell and tissue state. Urinary EVs in particular may act in inter-nephron communication with possible beneficial or detrimental effects. Increasing interest is addressed to the pharmacological properties of EVs as a cell-free therapy, since several of the effects crAQ/tgqcedited to stem cells have been recapitulated by administration of their EVs. Preclinical data in models of renal damage indicate a general regenerative potential of EVs derived from mesenchymal stromal cells of different sources, including bone marrow, fetal tissues, urine and kidney. In this review we will discuss the results on the effect of EVs in repair of acute and chronic renal injury, and the mechanisms involved. In addition, we will analyse the strategies for EV pharmacological applications in renal regenerative medicine and limits and benefits involved.
Collapse
|
43
|
Aquaporins in Urinary Extracellular Vesicles (Exosomes). Int J Mol Sci 2016; 17:ijms17060957. [PMID: 27322253 PMCID: PMC4926490 DOI: 10.3390/ijms17060957] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 01/05/2023] Open
Abstract
Since the successful characterization of urinary extracellular vesicles (uEVs) by Knepper’s group in 2004, these vesicles have been a focus of intense basic and translational research worldwide, with the aim of developing novel biomarkers and therapeutics for renal disease. Along with these studies, there is growing evidence that aquaporins (AQPs), water channel proteins, in uEVs have the potential to be diagnostically useful. In this review, we highlight current knowledge of AQPs in uEVs from their discovery to clinical application.
Collapse
|
44
|
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: 3.6] [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
|
45
|
Cell biology: Intercellular communication in the kidney. Nat Rev Nephrol 2016; 12:314. [PMID: 27067527 DOI: 10.1038/nrneph.2016.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|