201
|
Wang Y, Lu J, Chen L, Bian H, Hu J, Li D, Xia C, Xu H. Tumor-Derived EV-Encapsulated miR-181b-5p Induces Angiogenesis to Foster Tumorigenesis and Metastasis of ESCC. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:421-437. [PMID: 32244169 PMCID: PMC7118284 DOI: 10.1016/j.omtn.2020.03.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/14/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023]
Abstract
Pathological angiogenesis is necessary for tumor development and metastasis. Tumor-derived extracellular vesicles (EVs) play an important role in mediating the crosstalk between cancer cells and vascular endothelial cells. To date, whether and how microRNAs (miRNAs) encapsulated in tumor-derived EVs affect angiogenesis in esophageal squamous cell carcinoma (ESCC) remains unclear. Here, we showed that miR-181b-5p, an angiogenesis-promoting miRNA of ESCC, can be transferred from ESCC cells to vascular endothelial cells via EVs. In addition, ESCC-derived EVs-miR-181b-5p dramatically induced angiogenesis by targeting PTEN and PHLPP2, and thereby facilitated tumor growth and metastasis. Moreover, miR-181b-5p was highly expressed in ESCC tissues and serum EVs. High miR-181b-5p expression level in ESCC patients was well predicted for poor overall survival. Our work suggests that intercellular crosstalk between tumor cells and vascular endothelial cells is mediated by tumor-derived EVs. miR-181b-5p-enriched EVs secreted from ESCC cells are involved in angiogenesis that control metastasis of ESCC, providing a potential diagnostic biomarker or drug target for ESCC patients.
Collapse
Affiliation(s)
- Ying Wang
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Jiqiang Lu
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Lin Chen
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Huan Bian
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Jialiang Hu
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Dongping Li
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Chunlei Xia
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Hanmei Xu
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
202
|
Bliss CM, Parsons AJ, Nachbagauer R, Hamilton JR, Cappuccini F, Ulaszewska M, Webber JP, Clayton A, Hill AV, Coughlan L. Targeting Antigen to the Surface of EVs Improves the In Vivo Immunogenicity of Human and Non-human Adenoviral Vaccines in Mice. Mol Ther Methods Clin Dev 2020; 16:108-125. [PMID: 31934599 PMCID: PMC6953706 DOI: 10.1016/j.omtm.2019.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/12/2019] [Indexed: 12/25/2022]
Abstract
Adenoviral (Ad) vectors represent promising vaccine platforms for infectious disease. To overcome pre-existing immunity to commonly used human adenovirus serotype 5 (Ad5), vectors based on rare species or non-human Ads are being developed. However, these vectors often exhibit reduced potency compared with Ad5, necessitating the use of innovative approaches to augment the immunogenicity of the encoded antigen (Ag). To achieve this, we engineered model Ag, enhanced green fluorescent protein (EGFP), for targeting to the surface of host-derived extracellular vesicles (EVs), namely exosomes. Exosomes are nano-sized EVs that play important roles in cell-to-cell communication and in regulating immune responses. Directed targeting of Ag to the surface of EVs/exosomes is achieved by "exosome display," through fusion of Ag to the C1C2 domain of lactadherin, a protein highly enriched in exosomes. Herein, we engineered chimpanzee adenovirus ChAdOx1 and Ad5-based vaccines encoding EGFP, or EGFP targeted to EVs (EGFP_C1C2), and compared vaccine immunogenicity in mice. We determined that exosome display substantially increases Ag-specific humoral immunity following intramuscular and intranasal vaccination, improving the immunological potency of both ChAdOx1 and Ad5. We propose that this Ag-engineering approach could increase the immunogenicity of diverse Ad vectors that exhibit desirable manufacturing characteristics, but currently lack the potency of Ad5.
Collapse
Affiliation(s)
- Carly M. Bliss
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Andrea J. Parsons
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jennifer R. Hamilton
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Federica Cappuccini
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, ORCRB Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Marta Ulaszewska
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, ORCRB Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Jason P. Webber
- Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 2XN, UK
| | - Aled Clayton
- Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 2XN, UK
| | - Adrian V.S. Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, ORCRB Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Lynda Coughlan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, ORCRB Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
203
|
Galley JD, Besner GE. The Therapeutic Potential of Breast Milk-Derived Extracellular Vesicles. Nutrients 2020; 12:nu12030745. [PMID: 32168961 PMCID: PMC7146576 DOI: 10.3390/nu12030745] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/04/2020] [Accepted: 03/08/2020] [Indexed: 12/21/2022] Open
Abstract
In the past few decades, interest in the therapeutic benefits of exosomes and extracellular vesicles (EVs) has grown exponentially. Exosomes/EVs are small particles which are produced and exocytosed by cells throughout the body. They are loaded with active regulatory and stimulatory molecules from the parent cell including miRNAs and enzymes, making them prime targets in therapeutics and diagnostics. Breast milk, known for years to have beneficial health effects, contains a population of EVs which may mediate its therapeutic effects. This review offers an update on the therapeutic potential of exosomes/EVs in disease, with a focus on EVs present in human breast milk and their remedial effect in the gastrointestinal disease necrotizing enterocolitis. Additionally, the relationship between EV miRNAs, health, and disease will be examined, along with the potential for EVs and their miRNAs to be engineered for targeted treatments.
Collapse
|
204
|
Comprehensive analysis of miRNA-gene regulatory network with clinical significance in human cancers. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1201-1212. [PMID: 32170623 DOI: 10.1007/s11427-019-9667-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022]
Abstract
microRNAs (miRNAs), particularly the exosomal miRNAs have been widely used as biomarkers and promising therapeutic targets in cancer. However, a comprehensive analysis of miRNA-gene regulatory network with clinical significance remains scarce. The emergence of high-throughput multi-omics data over large, well-characterized patient cohorts provides an unprecedented opportunity to address this problem. Herein, we performed a clinic-centered analysis to identify cancer-associated miRNAs, miRNA-target axis. We first calculated the correlation among miRNA, mRNA and 75 unique clinico-pathological characteristics (CPCs) in 26 cancer types, and established an online resource (4CR). Interestingly, we found that the high expression of several DNA methylation-related enzymes was associated with adverse outcomes of cancer patients, and these genes were regulated by a cluster of miRNAs. Furthermore, by integrating exosomal miRNA and mRNA databases, we identified exosomal miRNA biomarkers for non-invasive cancer surveillance and therapy monitoring. Finally, we explored the role of CPC-related miRNAs for therapeutic effect prediction of drugs based on their shared targets. Our analysis pipeline illustrated the significance of clinic-centered analysis in miRNA-gene pair identification and provided helpful clues for future cancer studies.
Collapse
|
205
|
Abstract
The study of extracellular vesicles (EVs) has the potential to identify unknown cellular and molecular mechanisms in intercellular communication and in organ homeostasis and disease. Exosomes, with an average diameter of ~100 nanometers, are a subset of EVs. The biogenesis of exosomes involves their origin in endosomes, and subsequent interactions with other intracellular vesicles and organelles generate the final content of the exosomes. Their diverse constituents include nucleic acids, proteins, lipids, amino acids, and metabolites, which can reflect their cell of origin. In various diseases, exosomes offer a window into altered cellular or tissue states, and their detection in biological fluids potentially offers a multicomponent diagnostic readout. The efficient exchange of cellular components through exosomes can inform their applied use in designing exosome-based therapeutics.
Collapse
Affiliation(s)
- Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- School of Bioengineering, Rice University, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Valerie S LeBleu
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
206
|
Nahand JS, Moghoofei M, Salmaninejad A, Bahmanpour Z, Karimzadeh M, Nasiri M, Mirzaei HR, Pourhanifeh MH, Bokharaei‐Salim F, Mirzaei H, Hamblin MR. Pathogenic role of exosomes and microRNAs in HPV-mediated inflammation and cervical cancer: A review. Int J Cancer 2020; 146:305-320. [PMID: 31566705 PMCID: PMC6999596 DOI: 10.1002/ijc.32688] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/02/2019] [Accepted: 09/10/2019] [Indexed: 12/24/2022]
Abstract
Cervical cancer (CC) is the fourth most common cause of cancer death in women. The most important risk factor for the development of CC is cervical infection with human papilloma virus (HPV). Inflammation is a protective strategy that is triggered by the host against pathogens such as viral infections that acts rapidly to activate the innate immune response. Inflammation is beneficial if it is brief and well controlled; however, if the inflammation is excessive or it becomes of chronic duration, it can produce detrimental effects. HPV proteins are involved, both directly and indirectly, in the development of chronic inflammation, which is a causal factor in the development of CC. However, other factors may also have a potential role in stimulating chronic inflammation. MicroRNAs (miRNAs) (a class of noncoding RNAs) are strong regulators of gene expression. They have emerged as key players in several biological processes, including inflammatory pathways. Abnormal expression of miRNAs may be linked to the induction of inflammation that occurs in CC. Exosomes are a subset of extracellular vesicles shed by almost all types of cells, which can function as cargo transfer vehicles. Exosomes contain proteins and genetic material (including miRNAs) derived from their parent cells and can potentially affect recipient cells. Exosomes have recently been recognized to be involved in inflammatory processes and can also affect the immune response. In this review, we discuss the role of HPV proteins, miRNAs and exosomes in the inflammation associated with CC.
Collapse
Affiliation(s)
- Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Arash Salmaninejad
- Drug Applied Research Center, Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Bahmanpour
- Department of Medical Genetics, Faculty of Medicine, Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Karimzadeh
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Nasiri
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Farah Bokharaei‐Salim
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, USA
| |
Collapse
|
207
|
Wang B, Wang ZM, Ji JL, Gan W, Zhang A, Shi HJ, Wang H, Lv L, Li Z, Tang T, Du J, Wang XH, Liu BC. Macrophage-Derived Exosomal Mir-155 Regulating Cardiomyocyte Pyroptosis and Hypertrophy in Uremic Cardiomyopathy. JACC Basic Transl Sci 2020; 5:148-166. [PMID: 32140622 PMCID: PMC7046511 DOI: 10.1016/j.jacbts.2019.10.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 02/08/2023]
Abstract
miR-155 was synthesized and loaded into exosomes in increased infiltration of macrophages in a uremic heart. The released exosomal fusion with the plasma membrane leads to the release of miR-155 into the cytosol and translational repression of forkhead transcription factors of the O class in cardiomyocytes. Macrophage-derived miR-155–containing exosomes promoted cardiomyocyte pyroptosis and uremic cardiomyopathy changes (cardiac hypertrophy and fibrosis) by directly targeting FoxO3a in uremic mice. Inhibiting secretion from macrophage-derived miR-155–containing exosomes represents a novel therapeutic strategy for the management of uremic cardiomyopathy.
miR-155 was synthesized and loaded into exosomes in increased infiltration of macrophages in a uremic heart. The released exosomal fusion with the plasma membrane leads to the release of miR-155 into the cytosol and translational repression of forkhead transcription factors of the O class (FoxO3a) in cardiomyocytes. Finally, macrophage-derived miR-155–containing exosomes promoted cardiomyocyte pyroptosis and uremic cardiomyopathy changes (cardiac hypertrophy and fibrosis) by directly targeting FoxO3a in uremic mice.
Collapse
Affiliation(s)
- Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Ze-Mu Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jia-Ling Ji
- Department of Pediatric Nephrology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weihua Gan
- Department of Pediatric Nephrology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Aiqing Zhang
- Department of Pediatric Nephrology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao-Jie Shi
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Linli Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Zuolin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Taotao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaonan H Wang
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
208
|
Loria AD, Dattilo V, Santoro D, Guccione J, De Luca A, Ciaramella P, Pirozzi M, Iaccino E. Expression of Serum Exosomal miRNA 122 and Lipoprotein Levels in Dogs Naturally Infected by Leishmania infantum: A Preliminary Study. Animals (Basel) 2020; 10:ani10010100. [PMID: 31936232 PMCID: PMC7023135 DOI: 10.3390/ani10010100] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The immunopathogenesis of leishmaniasis is not completely understood. Exosomes are extracellular vesicles produced by most eukaryotic cells, containing various molecular constituents with biological effects (e.g., proteins, peptides, RNA). They play an important role in cell-to-cell signaling. Recently, exosomal microRNA were demonstrated to be able to regulate gene expression and protein production in mammalian cells, serving as potential biomarkers of disease. The microRNA miR-122 is a biomarker of hepatic damage widely studied in mice in the course of Leishmania infection. Leishmania organisms can interfere with miR-122 production leading to a dysfunction in cholesterol metabolism ensuring its proliferation in the infected host. In this study, we suggest that such a phenomenon may also occur in dogs affected by Leishmania infection. Abstract Current knowledge on the role of exosomal microRNA (miRNA) in canine leishmaniasis (CL), with particular regards to the interaction between miR-122 and lipid alterations, is limited. The aim of this study was to isolate/characterize exosomes in canine serum and evaluate the expression of miR-122 in ten healthy and ten leishmaniotic dogs. Serum exosomes were isolated using a polymer-based kit, ExoQuick® and characterized by flow cytometry and transmission electron microscopy, whereas miR-122-5p expression was evaluated by quantitative reverse-transcriptase polymerase chain reaction. A significant decreased expression of exosomal miR-122-5p, decreased serum levels of high-density lipoproteins, and increased serum levels of low-density lipoproteins were seen in leishmaniotic dogs when compared with healthy dogs. These results suggest that hepatic dysfunctions induced by the parasite interfere with lipoprotein status. The decreased expression of exosomal miR122 represents an additional effect of Leishmania infection in dogs as in people.
Collapse
Affiliation(s)
- Antonio Di Loria
- Department of Veterinary Medicine and Animal Productions, University Federico II, 80130 Napoli, Italy; (J.G.); (P.C.)
- Correspondence: (A.D.L.); (D.S.)
| | - Vincenzo Dattilo
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Domenico Santoro
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
- Correspondence: (A.D.L.); (D.S.)
| | - Jacopo Guccione
- Department of Veterinary Medicine and Animal Productions, University Federico II, 80130 Napoli, Italy; (J.G.); (P.C.)
| | - Adriana De Luca
- Department of Veterinary Medicine and Animal Productions, University Federico II, 80130 Napoli, Italy; (J.G.); (P.C.)
| | - Paolo Ciaramella
- Department of Veterinary Medicine and Animal Productions, University Federico II, 80130 Napoli, Italy; (J.G.); (P.C.)
| | - Marinella Pirozzi
- Institute of Protein Biochemistry, National Research Council, 88100 Napoli, Italy;
| | - Enrico Iaccino
- Department of Experimental and Clinical Medicine Magna Graecia University, 88100 Catanzaro, Italy;
| |
Collapse
|
209
|
Nasiri Kenari A, Cheng L, Hill AF. Methods for loading therapeutics into extracellular vesicles and generating extracellular vesicles mimetic-nanovesicles. Methods 2020; 177:103-113. [PMID: 31917274 DOI: 10.1016/j.ymeth.2020.01.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/05/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane bound vesicles released into the extracellular environment by eukaryotic and prokaryotic cells. EVs are enriched in active biomolecules and they can horizontally transfer cargo to recipient cells. In recent years EVs have demonstrated promising clinical applications due to their theragnostic potential. Although EVs have promising therapeutic potential, there are several challenges associated with using EVs before transition from the laboratory to clinical use. Some of these challenges include issues around low yield, isolation and purification methodologies, and efficient engineering (loading) of EVs with therapeutic cargo. Also, to achieve higher therapeutic efficiency, EV architecture and cargo may need to be manipulated prior to clinical application. Some of these issues have been addressed by developing biomimetic EVs. EV mimetic-nanovesicles (M-NVs) are a type of artificial EVs which can be generated from all cell types with comparable characteristics as EVs for an alternative therapeutic modality. In this review, we will discuss current techniques for modifying EVs and methodology used to generate and customize EV mimetic-nanovesicles.
Collapse
Affiliation(s)
- Amirmohammad Nasiri Kenari
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Australia
| | - Lesley Cheng
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Australia
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Australia.
| |
Collapse
|
210
|
Akerman AW, Blanding WM, Stroud RE, Nadeau EK, Mukherjee R, Ruddy JM, Zile MR, Ikonomidis JS, Jones JA. Elevated Wall Tension Leads to Reduced miR-133a in the Thoracic Aorta by Exosome Release. J Am Heart Assoc 2020; 8:e010332. [PMID: 30572760 PMCID: PMC6405702 DOI: 10.1161/jaha.118.010332] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Reduced miR‐133a was previously found to be associated with thoracic aortic (TA) dilation, as seen in aneurysm disease. Because wall tension increases with vessel diameter (Law of Laplace), this study tested the hypothesis that elevated tension led to the reduction of miR‐133a in the TA. Methods and Results Elevated tension (1.5 g; 150 mm Hg) applied to murine TA ex vivo reduced miR‐133a tissue abundance compared with TA held at normotension (0.7 g; 70 mm Hg). Cellular miR‐133a levels were reduced with biaxial stretch of isolated murine TA fibroblasts, whereas smooth muscle cells were not affected. Mechanisms contributing to the loss of miR‐133a abundance were further investigated in TA fibroblasts. Biaxial stretch did not reduce primary miR‐133a transcription and had no effect on the expression/abundance of 3 microRNA‐specific exoribonucleases. Remarkably, biaxial stretch increased exosome secretion, and exosomes isolated from TA fibroblasts contained more miR‐133a. Inhibition of exosome secretion prevented the biaxial stretch‐induced reduction of miR‐133a. Subsequently, 2 in vivo models of hypertension were used to determine the effect of elevated wall tension on miR‐133a abundance in the TA: wild‐type mice with osmotic pump–mediated angiotensin II infusion and angiotensin II–independent spontaneously hypertensive mice. Interestingly, the abundance of miR‐133a was decreased in TA tissue and increased in the plasma in both models of hypertension compared with a normotensive control group. Furthermore, miR‐133a was elevated in the plasma of hypertensive human subjects, compared with normotensive patients. Conclusions Taken together, these results identified exosome secretion as a tension‐sensitive mechanism by which miR‐133a abundance was reduced in TA fibroblasts.
Collapse
Affiliation(s)
- Adam W Akerman
- 1 Division of Cardiothoracic Surgery Department of Surgery Medical University of South Carolina Charleston SC.,4 Cardiothoracic Surgery Research University of North Carolina at Chapel Hill NC
| | - Walker M Blanding
- 1 Division of Cardiothoracic Surgery Department of Surgery Medical University of South Carolina Charleston SC
| | - Robert E Stroud
- 1 Division of Cardiothoracic Surgery Department of Surgery Medical University of South Carolina Charleston SC
| | - Elizabeth K Nadeau
- 1 Division of Cardiothoracic Surgery Department of Surgery Medical University of South Carolina Charleston SC
| | - Rupak Mukherjee
- 1 Division of Cardiothoracic Surgery Department of Surgery Medical University of South Carolina Charleston SC.,2 Research Service Ralph H. Johnson Veterans Affairs Medical Center Charleston SC
| | - Jean Marie Ruddy
- 3 Division of Vascular Surgery Medical University of South Carolina Charleston SC
| | - Michael R Zile
- 1 Division of Cardiothoracic Surgery Department of Surgery Medical University of South Carolina Charleston SC.,2 Research Service Ralph H. Johnson Veterans Affairs Medical Center Charleston SC
| | - John S Ikonomidis
- 4 Cardiothoracic Surgery Research University of North Carolina at Chapel Hill NC
| | - Jeffrey A Jones
- 1 Division of Cardiothoracic Surgery Department of Surgery Medical University of South Carolina Charleston SC.,2 Research Service Ralph H. Johnson Veterans Affairs Medical Center Charleston SC
| |
Collapse
|
211
|
Xiong Y, Tang Y, Fan F, Zeng Y, Li C, Zhou G, Hu Z, Zhang L, Liu Z. Exosomal hsa-miR-21-5p derived from growth hormone-secreting pituitary adenoma promotes abnormal bone formation in acromegaly. Transl Res 2020; 215:1-16. [PMID: 31469974 DOI: 10.1016/j.trsl.2019.07.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/01/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022]
Abstract
Growth hormone-secreting pituitary adenoma (GHPA), a benign endocrine tumor located in the base of the skull, results in acromegaly. In addition to the mass effect of the tumor itself in the sellar region, GHPA can lead to the overgrowth of almost every organ. Previous findings indicated that the processes underlying acromegaly were partly attributable to hyperactivity of the growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis. However, the mechanisms driving this syndrome remains largely unknown. Additionally, the roles of GHPA-derived exosomes, which contain functional microRNAs and proteins that manipulate target cell proliferation and differentiation in distal extremities, are also unknown. In this study, we demonstrated that GHPA exosomes promote bone formation in vitro and trabecula number in vivo. The mechanism of increased trabecula formation may be attributable to GHPA exosome-induced osteoblast proliferation via increased cell viability and DNA replication. We further discovered that exosomal hsa-miR-21-5p plays a distinct role from the GH/IGF-1 axis in these processes. Accordingly, the results of this study provide a novel mechanism whereby GHPA influences distal extremities and a new perspective for treating GHPA.
Collapse
Affiliation(s)
- Yuanyuan Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongjian Tang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fan Fan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Zeng
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Chuntao Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Gaofeng Zhou
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhongliang Hu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
212
|
Deng S, Zhou X, Xu J. Checkpoints Under Traffic Control: From and to Organelles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:431-453. [DOI: 10.1007/978-981-15-3266-5_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
213
|
Fridman E, Ginini L, Gil Z, Milman N. The Purification and Characterization of Exosomes from Macrophages. Methods Mol Biol 2020; 2184:77-90. [PMID: 32808219 DOI: 10.1007/978-1-0716-0802-9_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Macrophages play an essential role in diverse biological processes, from the immune response to inflammatory and neurodegenerative disorders, to various cancers. A macrophage subpopulation, known as tumor-associated macrophages (TAMs), has been shown to promote tumorigenesis, metastasis, and immune escape of cancer cells. Some of the pro-tumorigenic effects of TAMs are mediated via the secretion of nano-vesicles (exosomes) from macrophages to neighboring cells. In this chapter, we describe peritoneal macrophage isolation methods, polarization of TAMs, and purification and characterization of macrophage-derived exosomes.
Collapse
Affiliation(s)
- Eran Fridman
- The Laboratory for Applied Cancer Research, Department of Otolaryngology, Head and Neck Surgery, The Head and Neck Center, Rambam Healthcare Campus, The Technion, Israel Institute of Technology, Haifa, Israel
| | - Lana Ginini
- The Laboratory for Applied Cancer Research, Department of Otolaryngology, Head and Neck Surgery, The Head and Neck Center, Rambam Healthcare Campus, The Technion, Israel Institute of Technology, Haifa, Israel
| | - Ziv Gil
- The Laboratory for Applied Cancer Research, Department of Otolaryngology, Head and Neck Surgery, The Head and Neck Center, Rambam Healthcare Campus, The Technion, Israel Institute of Technology, Haifa, Israel.
| | - Neta Milman
- The Laboratory for Applied Cancer Research, Department of Otolaryngology, Head and Neck Surgery, The Head and Neck Center, Rambam Healthcare Campus, The Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
214
|
Biemmi V, Milano G, Ciullo A, Cervio E, Burrello J, Dei Cas M, Paroni R, Tallone T, Moccetti T, Pedrazzini G, Longnus S, Vassalli G, Barile L. Inflammatory extracellular vesicles prompt heart dysfunction via TRL4-dependent NF-κB activation. Theranostics 2020; 10:2773-2790. [PMID: 32194834 PMCID: PMC7052909 DOI: 10.7150/thno.39072] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
Background: After myocardial infarction, necrotic cardiomyocytes release damage-associated proteins that stimulate innate immune pathways and macrophage tissue infiltration, which drives inflammation and myocardial remodeling. Circulating inflammatory extracellular vesicles play a crucial role in the acute and chronic phases of ischemia, in terms of inflammatory progression. In this study, we hypothesize that the paracrine effect mediated by these vesicles induces direct cytotoxicity in cardiomyocytes. Thus, we examined whether reducing the generation of inflammatory vesicles within the first few hours after the ischemic event ameliorates cardiac outcome at short and long time points. Methods: Myocardial infarction was induced in rats that were previously injected intraperitoneally with a chemical inhibitor of extracellular-vesicle biogenesis. Heart global function was assessed by echocardiography performed at 7, 14 and 28 days after MI. Cardiac outcome was also evaluated by hemodynamic analysis at sacrifice. Cytotoxic effects of circulating EV were evaluated ex-vivo in a Langendorff, system by measuring the level of cardiac troponin I (cTnI) in the perfusate. Mechanisms undergoing cytotoxic effects of EV derived from pro-inflammatory macrophages (M1) were studied in-vitro in primary rat neonatal cardiomyocytes. Results: Inflammatory response following myocardial infarction dramatically increased the number of circulating extracellular vesicles carrying alarmins such as IL-1α, IL-1β and Rantes. Reducing the boost in inflammatory vesicles during the acute phase of ischemia resulted in preserved left ventricular ejection fraction in vivo. Hemodynamic analysis confirmed functional recovery by displaying higher velocity of left ventricular relaxation and improved contractility. When added to the perfusate of isolated hearts, post-infarction circulating vesicles induced significantly more cell death in adult cardiomyocytes, as assessed by cTnI release, comparing to circulating vesicles isolated from healthy (non-infarcted) rats. In vitro inflammatory extracellular vesicles induce cell death by driving nuclear translocation of NF-κB into nuclei of cardiomyocytes. Conclusion: Our data suggest that targeting circulating extracellular vesicles during the acute phase of myocardial infarction may offer an effective therapeutic approach to preserve function of ischemic heart.
Collapse
Affiliation(s)
- Vanessa Biemmi
- Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Giuseppina Milano
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Dept. Cœur-Vaisseaux, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Alessandra Ciullo
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Elisabetta Cervio
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Jacopo Burrello
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Michele Dei Cas
- Department of Health Sciences of the University of Milan, Milan, Italy
| | - Rita Paroni
- Department of Health Sciences of the University of Milan, Milan, Italy
| | - Tiziano Tallone
- Cell and Biomedical Technologies Unit Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Tiziano Moccetti
- Department of Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Giovanni Pedrazzini
- Department of Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Sarah Longnus
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Giuseppe Vassalli
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
215
|
You Y, Borgmann K, Edara VV, Stacy S, Ghorpade A, Ikezu T. Activated human astrocyte-derived extracellular vesicles modulate neuronal uptake, differentiation and firing. J Extracell Vesicles 2019; 9:1706801. [PMID: 32002171 PMCID: PMC6968484 DOI: 10.1080/20013078.2019.1706801] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/06/2019] [Accepted: 12/16/2019] [Indexed: 01/01/2023] Open
Abstract
Astrocytes in the central nervous system (CNS) provide supportive neural functions and mediate inflammatory responses from microglia. Increasing evidence supports their critical roles in regulating brain homoeostasis in response to pro-inflammatory factors such as cytokines and pathogen/damage-associated molecular pattern molecules in infectious and neurodegenerative diseases. However, the underlying mechanisms of the trans-cellular communication are still unclear. Extracellular vesicles (EVs) can transfer a large diversity of molecules such as lipids, nucleic acids and proteins for cellular communications. The purpose of this study is to characterize the EVs cargo proteins derived from human primary astrocytes (ADEVs) under both physiological and pathophysiological conditions. ADEVs were isolated from human primary astrocytes after vehicle (CTL) or interleukin-1β (IL-1β) pre-treatment. Label-free quantitative proteomic profiling revealed a notable up-regulation of proteins including actin-associated molecules, integrins and major histocompatibility complex in IL-1β-ADEVs compared to CTL-ADEVs, which were involved in cellular metabolism and organization, cellular communication and inflammatory response. When fluorescently labelled ADEVs were added into primary cultured mouse cortical neurons, we found a significantly increased neuronal uptake of IL-1β-ADEVs compared to CTL-ADEVs. We further confirmed it is likely due to the enrichment of surface proteins in IL-1β-ADEVs, as IL-1β-ADEVs uptake by neurons was partially suppressed by a specific integrin inhibitor. Additionally, treatment of neurons with IL-1β-ADEVs also reduced neurite outgrowth, branching and neuronal firing. These findings provide insight for the molecular mechanism of the ADEVs' effects on neural uptake, neural differentiation and maturation, and its alteration in inflammatory conditions.
Collapse
Affiliation(s)
- Yang You
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Kathleen Borgmann
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Venkata Viswanadh Edara
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Satomi Stacy
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Anuja Ghorpade
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Tsuneya Ikezu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.,Neurology, Boston University School of Medicine, Boston, MA, USA.,Center for Systems Neuroscience, Boston University, Boston, MA, USA
| |
Collapse
|
216
|
Eliminating blood oncogenic exosomes into the small intestine with aptamer-functionalized nanoparticles. Nat Commun 2019; 10:5476. [PMID: 31792209 PMCID: PMC6889386 DOI: 10.1038/s41467-019-13316-w] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022] Open
Abstract
There are disease-causing biohazards in the blood that cannot be treated with modern medicines. Here we show that an intelligently designed safe biomaterial can precisely identify, tow and dump a targeted biohazard from the blood into the small intestine. Positively charged mesoporous silica nanoparticles (MSNs) functionalized with EGFR-targeting aptamers (MSN-AP) specifically recognize and bind blood-borne negatively charged oncogenic exosomes (A-Exo), and tow A-Exo across hepatobiliary layers and Oddi's sphincter into the small intestine. MSN-AP specifically distinguish and bind A-Exo from interfering exosomes in cell culture and rat and patient blood to form MSN-AP and A-Exo conjugates (MSN-Exo) that transverse hepatocytes, cholangiocytes, and endothelial monolayers via endocytosis and exocytosis mechanisms, although Kupffer cells have been shown to engulf some MSN-Exo. Blood MSN-AP significantly decreased circulating A-Exo levels, sequentially increased intestinal A-Exo and attenuated A-Exo-induced lung metastasis in mice. This study opens an innovative avenue to relocate blood-borne life-threatening biohazards to the intestine.
Collapse
|
217
|
Wu J, Gu J, Shen L, Fang D, Zou X, Cao Y, Wang S, Mao L. Exosomal MicroRNA-155 Inhibits Enterovirus A71 Infection by Targeting PICALM. Int J Biol Sci 2019; 15:2925-2935. [PMID: 31853228 PMCID: PMC6909958 DOI: 10.7150/ijbs.36388] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Enterovirus A71 (EV-A71) causes hand, foot, and mouth disease (HFMD) that is associated with neurological complications. Researchers have shown that exosomes containing host cellular microRNA (miRNA) can modulate the recipient's cellular response during viral infection. However, it is unclear how exosomal miRNAs regulate this response during EV-A71 infection. In this study, we used an exosomal miRNA chip to show that microRNA-155 (miR-155) was markedly enriched in exosomes after EV-A71 infection. Moreover, exosomal miR-155 efficaciously inhibited EV-A71 infection by targeting phosphatidylinositol clathrin assembly protein (PICALM) in recipient cells. Importantly, we confirmed that exosomal miR-155 reduced EV-A71 infection severity in vivo. Additionally, miR-155 levels in throat swabs from EV-A71-infected patients were higher than in those from healthy individuals. Collectively, our findings provide evidence that exosomal miR-155 plays a role in host-pathogen interactions by mediating EV-A71 infection via the repression of PICALM; these results provide insights into the regulatory mechanisms of viral infection.
Collapse
Affiliation(s)
- Jing Wu
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jiaqi Gu
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Li Shen
- Clinical Laboratory, Zhenjiang Center for Disease Control and Prevention, Jiangsu, China
| | - Daihua Fang
- Clinical Laboratory, Xuzhou Children's Hospital, Xuzhou, China
| | - Xinran Zou
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuwen Cao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Lingxiang Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| |
Collapse
|
218
|
Pérez PS, Romaniuk MA, Duette GA, Zhao Z, Huang Y, Martin-Jaular L, Witwer KW, Théry C, Ostrowski M. Extracellular vesicles and chronic inflammation during HIV infection. J Extracell Vesicles 2019; 8:1687275. [PMID: 31998449 PMCID: PMC6963413 DOI: 10.1080/20013078.2019.1687275] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/16/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
Inflammation is a hallmark of HIV infection. Among the multiple stimuli that can induce inflammation in untreated infection, ongoing viral replication is a primary driver. After initiation of effective combined antiretroviral therapy (cART), HIV replication is drastically reduced or halted. However, even virologically controlled patients may continue to have abnormal levels of inflammation. A number of factors have been proposed to cause inflammation in HIV infection: among others, residual (low-level) HIV replication, production of HIV protein or RNA in the absence of replication, microbial translocation from the gut to the circulation, co-infections, and loss of immunoregulatory responses. Importantly, chronic inflammation in HIV-infected individuals increases the risk for a number of non-infectious co-morbidities, including cancer and cardiovascular disease. Thus, achieving a better understanding of the underlying mechanisms of HIV-associated inflammation in the presence of cART is of utmost importance. Extracellular vesicles have emerged as novel actors in intercellular communication, involved in a myriad of physiological and pathological processes, including inflammation. In this review, we will discuss the role of extracellular vesicles in the pathogenesis of HIV infection, with particular emphasis on their role as inducers of chronic inflammation.
Collapse
Affiliation(s)
- Paula Soledad Pérez
- Instituto INBIRS, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | | | - Gabriel A. Duette
- Instituto INBIRS, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Zezhou Zhao
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yiyao Huang
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lorena Martin-Jaular
- INSERM U932, Institut Curie Centre de Recherche, PSL Research University, Paris, France
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clotilde Théry
- INSERM U932, Institut Curie Centre de Recherche, PSL Research University, Paris, France
| | - Matías Ostrowski
- Instituto INBIRS, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| |
Collapse
|
219
|
Zhong X, Gao W, Wu R, Liu H, Ge J. Dendritic cell exosome‑shuttled miRNA146a regulates exosome‑induced endothelial cell inflammation by inhibiting IRAK‑1: A feedback control mechanism. Mol Med Rep 2019; 20:5315-5323. [PMID: 31638185 DOI: 10.3892/mmr.2019.10749] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/04/2019] [Indexed: 11/06/2022] Open
Abstract
Activation of endothelial cells is the first step of atherosclerosis. The current authors have previously reported that exosomes from mature dendritic cells (mDC‑exo) participate in endothelial inflammation and atherosclerosis through membrane tumor necrosis factor‑α mediated the nuclear factor (NF)‑κB signaling pathway. However, whether mDC‑exo shuttled microRNAs (miRNAs/miRs) play a role in endothelial inflammation remains unknown. In this study, mDC‑exo were co‑cultured with human umbilical vein endothelial cells (HUVECs) and the expression of adhesion molecules, such as vascular cell adhesion molecule‑1, intercellular adhesion molecule‑1 and E‑Selectin was investigated. Then the expression of miRNAs in DC‑exo was explored and the role of miR‑146a in endothelial inflammation was investigated. mDC‑exos were first demonstrated to increase endothelial expression of adhesion molecules through a quick activation of the NF‑κB signaling pathway. Then it was demonstrated that HUVECs resistant to a second stimulation after the first stimulation by mDC‑exo. A set of miRNAs were targeted and their expression in HUVECs stimulated with mDC‑exo was measured. Finally, it was confirmed that mDC‑exo shuttles miR‑146a into HUVECs and the shuttled miR‑146a contributes to protect HUVECs from a second stimulation through inhibiting interleukin‑1 receptor‑associated kinase. These data suggest a negative feedback loop of inflammation regulation by DC‑exo.
Collapse
Affiliation(s)
- Xin Zhong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
| | - Wei Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
| | - Runda Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
| | - Haibo Liu
- Department of Cardiovascular Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, P.R. China
| |
Collapse
|
220
|
Liu R, Li X, Zhu W, Wang Y, Zhao D, Wang X, Gurley EC, Liang G, Chen W, Lai G, Pandak WM, Lippman HR, Bajaj JS, Hylemon PB, Zhou H. Cholangiocyte-Derived Exosomal Long Noncoding RNA H19 Promotes Hepatic Stellate Cell Activation and Cholestatic Liver Fibrosis. Hepatology 2019; 70:1317-1335. [PMID: 30985008 PMCID: PMC6783323 DOI: 10.1002/hep.30662] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/29/2019] [Indexed: 12/16/2022]
Abstract
Activation of hepatic stellate cells (HSCs) represents the primary driving force to promote the progression of chronic cholestatic liver diseases. We previously reported that cholangiocyte-derived exosomal long noncoding RNA-H19 (lncRNA-H19) plays a critical role in promoting cholestatic liver injury. However, it remains unclear whether cholangiocyte-derived lncRNA-H19 regulates HSC activation, which is the major focus of this study. Both bile duct ligation (BDL) and Mdr2 knockout (Mdr2-/- ) mouse models were used. Wild-type and H19maternalΔExon1/+ (H19KO) mice were subjected to BDL. Mdr2-/- H19maternalΔExon1/+ (DKO) mice were generated. Exosomes isolated from cultured mouse and human cholangiocytes or mouse serum were used for in vivo transplantation and in vitro studies. Fluorescence-labeled exosomes and flow cytometry were used to monitor exosome uptake by hepatic cells. Collagen gel contraction and bromodeoxyuridine assays were used to determine the effect of exosomal-H19 on HSC activation and proliferation. Mouse and human primary sclerosing cholangitis (PSC)/primary biliary cholangitis (PBC) liver samples were analyzed by real-time PCR, western blot analysis, histology, and immunohistochemistry. The results demonstrated that hepatic H19 level was closely correlated with the severity of liver fibrosis in both mouse models and human patients with PSC and PBC. H19 deficiency significantly protected mice from liver fibrosis in BDL and Mdr2-/- mice. Transplanted cholangiocyte-derived H19-enriched exosomes were rapidly and preferentially taken up by HSCs and HSC-derived fibroblasts, and promoted liver fibrosis in BDL-H19KO mice and DKO mice. H19-enriched exosomes enhanced transdifferentiation of cultured mouse primary HSCs and promoted proliferation and matrix formation in HSC-derived fibroblasts. Conclusion: Cholangiocyte-derived exosomal H19 plays a critical role in the progression of cholestatic liver fibrosis by promoting HSC differentiation and activation and represents a potential diagnostic biomarker and therapeutic target for cholangiopathies.
Collapse
Affiliation(s)
- Runping Liu
- Department of Microbiology and Immunology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, USA;,Division of Gastroenterology, Hepatology and Nutrition and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University
| | - Xiaojiaoyang Li
- Department of Microbiology and Immunology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, USA;,Division of Gastroenterology, Hepatology and Nutrition and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University
| | - Weiwei Zhu
- Department of Microbiology and Immunology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, USA;,School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanyan Wang
- Department of Microbiology and Immunology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, USA;,School of Pharmaceutical Science, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Derrick Zhao
- Department of Microbiology and Immunology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, USA;,Division of Gastroenterology, Hepatology and Nutrition and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University
| | - Xuan Wang
- Department of Microbiology and Immunology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, USA;,Division of Gastroenterology, Hepatology and Nutrition and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University
| | - Emily C. Gurley
- Department of Microbiology and Immunology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, USA;,Division of Gastroenterology, Hepatology and Nutrition and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University
| | - Guang Liang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weidong Chen
- School of Pharmaceutical Science, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Guanhua Lai
- Department of Pathology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - William M Pandak
- Division of Gastroenterology, Hepatology and Nutrition and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University
| | - H. Robert Lippman
- Department of Pathology and Laboratory Medicine, McGuire Veterans Affairs Medical Center, Richmond, Virginia, USA
| | - Jasmohan S. Bajaj
- Division of Gastroenterology, Hepatology and Nutrition and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University
| | - Phillip B. Hylemon
- Department of Microbiology and Immunology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, USA;,Division of Gastroenterology, Hepatology and Nutrition and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University
| | - Huiping Zhou
- Department of Microbiology and Immunology and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University, Richmond, Virginia, USA;,Division of Gastroenterology, Hepatology and Nutrition and McGuire Veterans Affairs Medical Center, Virginia Commonwealth University;,Address correspondence to: Huiping Zhou, Ph.D, Department of Microbiology & Immunology, Virginia Commonwealth University, McGuire Veterans Affairs Medical Center, 1220 East Broad Street, MMRB-5044, Richmond, VA 23298-0678, USA, Tel: 804-828-6817; Fax: 804-828-0676,
| |
Collapse
|
221
|
Balaphas A, Meyer J, Sadoul R, Morel P, Gonelle-Gispert C, Bühler LH. Extracellular vesicles: Future diagnostic and therapeutic tools for liver disease and regeneration. Liver Int 2019; 39:1801-1817. [PMID: 31286675 DOI: 10.1111/liv.14189] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 06/06/2019] [Accepted: 07/01/2019] [Indexed: 02/13/2023]
Abstract
Extracellular vesicles are membrane fragments that can be produced by all cell types. Interactions between extracellular vesicles and various liver cells constitute an emerging field in hepatology and recent evidences have established a role for extracellular vesicles in various liver diseases and physiological processes. Extracellular vesicles originating from liver cells are implicated in intercellular communication and fluctuations of specific circulating extracellular vesicles could constitute new diagnostic tools. In contrast, extracellular vesicles derived from progenitor cells interact with hepatocytes or non-parenchymal cells, thereby protecting the liver from various injuries and promoting liver regeneration. Our review focuses on recent developments investigating the role of various types of extracellular vesicles in acute and chronic liver diseases as well as their potential use as biomarkers and therapeutic tools.
Collapse
Affiliation(s)
- Alexandre Balaphas
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Jeremy Meyer
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Rémy Sadoul
- Université Grenoble Alpes, Institut des Neurosciences, Grenoble, France
| | - Philippe Morel
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| | - Leo Hans Bühler
- Division of Digestive Surgery, University Hospitals of Geneva, Geneva, Switzerland
- Surgical Research Unit, University Hospitals of Geneva, Geneva, Switzerland
- Geneva Medical School, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
222
|
Human Immunodeficiency Virus (HIV) Infection and Use of Illicit Substances Promote Secretion of Semen Exosomes that Enhance Monocyte Adhesion and Induce Actin Reorganization and Chemotactic Migration. Cells 2019; 8:cells8091027. [PMID: 31484431 PMCID: PMC6770851 DOI: 10.3390/cells8091027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/09/2019] [Accepted: 08/15/2019] [Indexed: 12/23/2022] Open
Abstract
Semen exosomes (SE) from HIV-uninfected (HIV−) individuals potently inhibit HIV infection in vitro. However, morphological changes in target cells in response to SE have not been characterized or have the effect of HIV infection or the use of illicit substances, specifically psychostimulants, on the function of SE been elucidated. The objective of this study was to evaluate the effect of HIV infection, psychostimulant use, and both together on SE-mediated regulation of monocyte function. SE were isolated from semen of HIV− and HIV-infected (HIV+) antiretroviral therapy (ART)-naive participants who reported either using or not using psychostimulants. The SE samples were thus designated as HIV−Drug−, HIV−Drug+, HIV+Drug−, and HIV+Drug+. U937 monocytes were treated with different SEs and analyzed for changes in transcriptome, morphometrics, actin reorganization, adhesion, and chemotaxis. HIV infection and/or use of psychostimulants had minimal effects on the physical characteristics of SE. However, different SEs had diverse effects on the messenger RNA signature of monocytes and rapidly induced monocyte adhesion and spreading. SE from HIV infected or psychostimulants users but not HIV−Drug− SE, stimulated actin reorganization, leading to the formation of filopodia-like structures and membrane ruffles containing F-actin and vinculin that in some cases were colocalized. All SE stimulated monocyte chemotaxis to HIV secretome and activated the secretion of matrix metalloproteinases, a phenotype exacerbated by HIV infection and psychostimulant use. SE-directed regulation of cellular morphometrics and chemotaxis depended on the donor clinical status because HIV infection and psychostimulant use altered SE function. Although our inclusion criteria specified the use of cocaine, humans are poly-drug and alcohol users and our study participants used psychostimulants, marijuana, opiates, and alcohol. Thus, it is possible that the effects observed in this study may be due to one of these other substances or due to an interaction between different substances.
Collapse
|
223
|
Zhang C, Guo F, Chang M, Zhou Z, Yi L, Gao C, Huang X, Huan J. Exosome-delivered syndecan-1 rescues acute lung injury via a FAK/p190RhoGAP/RhoA/ROCK/NF-κB signaling axis and glycocalyx enhancement. Exp Cell Res 2019; 384:111596. [PMID: 31487506 DOI: 10.1016/j.yexcr.2019.111596] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/30/2019] [Accepted: 09/01/2019] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) is characterized by protein-rich pulmonary edema, critical hypoxemia, and influx of pro-inflammatory cytokines and cells. There are currently no effective pharmacon therapies in clinical practice. Syndecan-1 in endothelial cells has potential to protect barrier function of endothelium and suppress inflammation response. Thus, the present study was to identify whether exosomes with encapsulation of syndecan-1 could achieve ideal therapeutic effects in ALI. Exosomes were isolated from the conditional medium of lentivirus-transfected mouse pulmonary microvascular endothelial cells (MPMVECs) and characterized by nanoparticle tracking analysis (NTA), transmission electron microscopy (TEM), and western blotting. ALI mouse models were induced via intratracheal administration of lipopolysaccharide (LPS) and treated with exosomes. Lung edema, inflammation, and glycocalyx thickness were examined. The possible mechanism was verified by immunoblotting in MPMVECs. The purified exosomes included SDC1-high-Exos and SDC1-low-Exos which loaded with up-regulated syndecan-1 and down-regulated syndecan-1 respectively. Compared with SDC1-low-Exos, administration of SDC1-high-Exos could ameliorate lung edema and inflammation, attenuate number of cells and protein levels in bronchoalveolar lavage fluid (BALF), and preserve glycocalyx. Furthermore, SDC1-high-Exos also mitigated the expression of pro-inflammatory cytokines such as IL-1β, TNF-α, and IL-6 following LPS challenge. In MPMVECs, SDC1-high-Exos decreased stress fiber formation and ameliorated monolayer hyper-permeability after LPS stimulation. Western blotting analysis demonstrated that FAK/p190RhoGAP/RhoA/ROCK/NF-κB signaling pathway may be involved in LPS-induced ALI. In conclusion, SDC1-high-Exos play a pivotal role in ameliorating LPS-stimulated ALI models and may be served as a potential therapeutic agent for clinical application in the future.
Collapse
Affiliation(s)
- Chuankai Zhang
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China
| | - Feng Guo
- Department of Plastic Surgery, Affiliated Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai, China
| | - Mengling Chang
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China
| | - Zengding Zhou
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China
| | - Lei Yi
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China
| | - Chengjin Gao
- Emergency Department, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiaoqin Huang
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China.
| | - Jingning Huan
- Department of Burn and Plastic Surgery, Shanghai Jiao Tong University, School of Medicine, Rui Jin Hospital, Shanghai, 230022, China.
| |
Collapse
|
224
|
Zhang Y, Wang F, Zhang H, Wang H, Liu Y. Multivalency Interface and g-C3N4 Coated Liquid Metal Nanoprobe Signal Amplification for Sensitive Electrogenerated Chemiluminescence Detection of Exosomes and Their Surface Proteins. Anal Chem 2019; 91:12100-12107. [DOI: 10.1021/acs.analchem.9b03427] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Yimeng Zhang
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Feng Wang
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Huixin Zhang
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Hongye Wang
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Yang Liu
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| |
Collapse
|
225
|
Bandera A, Taramasso L, Bozzi G, Muscatello A, Robinson JA, Burdo TH, Gori A. HIV-Associated Neurocognitive Impairment in the Modern ART Era: Are We Close to Discovering Reliable Biomarkers in the Setting of Virological Suppression? Front Aging Neurosci 2019; 11:187. [PMID: 31427955 PMCID: PMC6687760 DOI: 10.3389/fnagi.2019.00187] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/10/2019] [Indexed: 11/24/2022] Open
Abstract
The prevalence of the most severe forms of HIV-associated neurocognitive disorders (HAND) is decreasing due to worldwide availability and high efficacy of antiretroviral treatment (ART). However, several grades of HIV-related cognitive impairment persist with effective ART and remain a clinical concern for people with HIV (PWH). The pathogenesis of these cognitive impairments has yet to be fully understood and probably multifactorial. In PWH with undetectable peripheral HIV-RNA, the presence of viral escapes in cerebrospinal fluid (CSF) might explain a proportion of cases, but not all. Many other mechanisms have been hypothesized to be involved in disease progression, in order to identify possible therapeutic targets. As potential indicators of disease staging and progression, numerous biomarkers have been used to characterize and implicate chronic inflammation in the pathogenesis of neuronal injuries, such as certain phenotypes of activated monocytes/macrophages, in the context of persistent immune activation. Despite none of them being disease-specific, the correlation of several CSF cellular biomarkers to HIV-induced neuronal damage has been investigated. Furthermore, recent studies have been evaluating specific microRNA (miRNA) profiles in the CSF of PWH with neurocognitive impairment (NCI). The aim of the present study is to review the body of evidence on different biomarkers use in research and clinical settings, focusing on PWH on ART with undetectable plasma HIV-RNA.
Collapse
Affiliation(s)
- Alessandra Bandera
- Infectious Disease Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milano, Milan, Italy
| | - Lucia Taramasso
- Infectious Disease Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Infectious Diseases Clinic, Department of Health Sciences, School of Medical and Pharmaceutical Sciences, Policlinico Hospital San Martino, University of Genova (DISSAL), Genova, Italy
| | - Giorgio Bozzi
- Infectious Disease Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonio Muscatello
- Infectious Disease Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jake A Robinson
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Tricia H Burdo
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Andrea Gori
- Infectious Disease Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milano, Milan, Italy
| |
Collapse
|
226
|
Kaminski VDL, Ellwanger JH, Chies JAB. Extracellular vesicles in host-pathogen interactions and immune regulation - exosomes as emerging actors in the immunological theater of pregnancy. Heliyon 2019; 5:e02355. [PMID: 31592031 PMCID: PMC6771614 DOI: 10.1016/j.heliyon.2019.e02355] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 06/30/2019] [Accepted: 08/19/2019] [Indexed: 02/06/2023] Open
Abstract
This review correlates and summarizes the role of the maternal-fetal interface in the immune tolerance of the fetus and the processes that lead to infection avoidance, emphasizing the participation of exosomes and other extracellular vesicles in both situations. Exosomes are released into the extracellular medium by several cell types and are excellent carriers of biomolecules. Host-derived exosomes and the transport of pathogen-derived molecules by exosomes impact infections in different ways. The interactions of exosomes with the maternal immune system are pivotal to a favorable gestational outcome. In this review, we highlight the potential role of exosomes in the establishment of an adequate milieu that enables embryo implantation and discuss the participation of exosomes released at the maternal-fetal interface during the establishment of an immune-privileged compartment for fetal development. The placenta is a component where important strategies are used to minimize the risk of infection. To present a contrast, we also discuss possible mechanisms used by pathogens to cross the maternal-fetal interface. We review the processes, mechanisms, and potential consequences of dysregulation in all of the abovementioned phenomena. Basic information about exosomes and their roles in viral immune evasion is also presented. The interactions between extracellular vesicles and bacteria, fungi, parasites and proteinaceous infectious agents are addressed. The discovery of the placental microbiota and the implications of this new microbiota are also discussed, and current proposals that explain fetal/placental colonization by both pathogenic and commensal microbes are addressed. The comprehension of such interactions will help us to understand the immune dynamics of human pregnancy and the mechanisms of immune evasion used by different pathogens.
Collapse
Affiliation(s)
| | | | - José Artur Bogo Chies
- Laboratório de Imunobiologia e Imunogenética, Programa de Pós-Graduação em Genética e Biologia Molecular, Departamento de Genética, Universidade Federal do Rio Grande do Sul – UFRGS, Porto Alegre, RS, Brazil
| |
Collapse
|
227
|
Yin Z, Ma T, Huang B, Lin L, Zhou Y, Yan J, Zou Y, Chen S. Macrophage-derived exosomal microRNA-501-3p promotes progression of pancreatic ductal adenocarcinoma through the TGFBR3-mediated TGF-β signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:310. [PMID: 31307515 PMCID: PMC6631643 DOI: 10.1186/s13046-019-1313-x] [Citation(s) in RCA: 246] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 07/04/2019] [Indexed: 12/13/2022]
Abstract
Background Exosomes from cancer cells or immune cells, carrying bio-macromolecules or microRNAs (miRNAs), participate in tumor pathogenesis and progression by modulating microenvironment. Our study aims to investigate the role of these microRNA-501-3p (miR-501-3p) containing exosomes derived from tumor-associated macrophage (TAM) in the progression of pancreatic ductal adenocarcinoma (PDAC). Methods Firstly, the function of TAM recruitment in PDAC tissues was assessed, followed by identification of the effects of M2 macrophage-derived exosomes on PDAC cell activities and tumor formation and metastasis in mice. In silico analysis was conducted to predict differentially expressed genes and regulatory miRNAs related to PDAC treated with macrophages, which determined miR-501-3p and TGFBR3 for subsequent experiments. Next, gain- and loss-of-function experiments were performed to examine their role in PDAC progression with the involvement of the TGF-β signaling pathway. Results TAM recruitment in PDAC tissues was associated with metastasis. Highly expressed miR-501-3p was observed in PDAC tissues and TAM-derived exosomes. Both M2 macrophage-derived exosomes and miR-501-3p promoted PDAC cell migration and invasion, as well as tumor formation and metastasis in nude mice. MiR-501-3p was verified to target TGFBR3. PDAC cells presented with down-regulated TGFBR3, which was further decreased in response to M2 macrophage treatment. TGF-β signaling pathway activation was implicated in the promotion of miR-501-3p in PDAC development. The suppression of macrophage-derived exosomal miR-501-3p resulted in the inhibition of tumor formation and metastasis in vivo. Conclusion M2 macrophage-derived exosomal miR-501-3p inhibits tumor suppressor TGFBR3 gene and facilitates the development of PDAC by activating the TGF-β signaling pathway, which provides novel targets for the molecular treatment of PDAC. Electronic supplementary material The online version of this article (10.1186/s13046-019-1313-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zi Yin
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| | - Tingting Ma
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Bowen Huang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Lehang Lin
- Medical Research Center, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Yu Zhou
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Jinhai Yan
- Pathology Department, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Yiping Zou
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China
| | - Sheng Chen
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, No. 106, Zhongshan Er Road, Guangzhou, 510080, Guangdong Province, People's Republic of China.
| |
Collapse
|
228
|
Zhou W, Woodson M, Sherman MB, Neelakanta G, Sultana H. Exosomes mediate Zika virus transmission through SMPD3 neutral Sphingomyelinase in cortical neurons. Emerg Microbes Infect 2019; 8:307-326. [PMID: 30866785 PMCID: PMC6455149 DOI: 10.1080/22221751.2019.1578188] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The harmful effects of ZIKA virus (ZIKV) infection are reflected by severe neurological manifestations such as microcephaly in neonates and other complications associated with Guillain-Barré syndrome in adults. The transmission dynamics of ZIKV in or between neurons, or within the developing brains of the foetuses are not fully understood. Using primary cultures of murine cortical neurons, we show that ZIKV uses exosomes as mediators of viral transmission between neurons. Cryo-electron microscopy showed heterogeneous population of neuronal exosomes with a size range of 30–200 nm. Increased production of exosomes from neuronal cells was noted upon ZIKV infection. Neuronal exosomes contained both ZIKV viral RNA and protein(s) that were highly infectious to naïve cells. RNaseA and neutralizing antibodies treatment studies suggest the presence of viral RNA/proteins inside exosomes. Exosomes derived from time- and dose-dependent incubations showed increasing viral loads suggesting higher packaging and delivery of ZIKV RNA and proteins. Furthermore, we noted that ZIKV induced both activity and gene expression of neutral Sphingomyelinase (nSMase)-2/SMPD3, an important molecule that regulates production and release of exosomes. Silencing of SMPD3 in neurons resulted in reduced viral burden and transmission through exosomes. Treatment with SMPD3 specific inhibitor GW4869, significantly reduced ZIKV loads in both cortical neurons and in exosomes derived from these neuronal cells. Taken together, our results suggest that ZIKV modulates SMPD3 activity in cortical neurons for its infection and transmission through exosomes perhaps leading to severe neuronal death that may result in neurological manifestations such as microcephaly in the developing embryonic brains.
Collapse
Affiliation(s)
- Wenshuo Zhou
- a Department of Biological Sciences, Center for Molecular Medicine , Old Dominion University , Norfolk , VA , USA
| | - Michael Woodson
- b Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , TX , USA
| | - Michael B Sherman
- b Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , TX , USA.,c Sealy Center for Structural Biology and Molecular Biophysics , University of Texas Medical Branch , Galveston , TX , USA
| | - Girish Neelakanta
- a Department of Biological Sciences, Center for Molecular Medicine , Old Dominion University , Norfolk , VA , USA
| | - Hameeda Sultana
- a Department of Biological Sciences, Center for Molecular Medicine , Old Dominion University , Norfolk , VA , USA.,d Department of Medicine, Division of Infectious Diseases and International Health , University of Virginia School of Medicine , Charlottesville , VA , USA
| |
Collapse
|
229
|
Maemura T, Fukuyama S, Sugita Y, Lopes TJS, Nakao T, Noda T, Kawaoka Y. Lung-Derived Exosomal miR-483-3p Regulates the Innate Immune Response to Influenza Virus Infection. J Infect Dis 2019; 217:1372-1382. [PMID: 29373693 DOI: 10.1093/infdis/jiy035] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 01/18/2018] [Indexed: 01/21/2023] Open
Abstract
Exosomes regulate cell-cell communication by transferring functional proteins and RNAs between cells. Here, to clarify the function of exosomes during influenza virus infection, we characterized lung-derived exosomal microRNAs (miRNAs). Among the detected miRNAs, miR-483-3p was present at high levels in bronchoalveolar lavage fluid (BALF) exosomes during infection of mice with various strains of influenza virus, and miR-483-3p transfection potentiated gene expression of type I interferon and proinflammatory cytokine upon viral infection of MLE-12 cells. RNF5, a regulator of the RIG-I signaling pathway, was identified as a target gene of miR-483-3p. Moreover, we found that CD81, another miR-483-3p target, functions as a negative regulator of RIG-I signaling in MLE-12 cells. Taken together, this study indicates that BALF exosomal miRNAs may mediate the antiviral and inflammatory response to influenza virus infection.
Collapse
Affiliation(s)
- Tadashi Maemura
- Division of Virology, Department of Microbiology and Immunology, Japan.,Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Japan
| | - Satoshi Fukuyama
- Division of Virology, Department of Microbiology and Immunology, Japan
| | - Yukihiko Sugita
- Molecular Cryo-Electron Microscopy Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| | - Tiago J S Lopes
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Japan
| | - Tomomi Nakao
- Division of Virology, Department of Microbiology and Immunology, Japan
| | - Takeshi Noda
- Laboratory of Ultrastructural Virology, Institute for Frontier Life and Medical Sciences, Kyoto University, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Japan.,Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Japan.,Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Japan
| |
Collapse
|
230
|
Borghesan M, Fafián-Labora J, Eleftheriadou O, Carpintero-Fernández P, Paez-Ribes M, Vizcay-Barrena G, Swisa A, Kolodkin-Gal D, Ximénez-Embún P, Lowe R, Martín-Martín B, Peinado H, Muñoz J, Fleck RA, Dor Y, Ben-Porath I, Vossenkamper A, Muñoz-Espin D, O'Loghlen A. Small Extracellular Vesicles Are Key Regulators of Non-cell Autonomous Intercellular Communication in Senescence via the Interferon Protein IFITM3. Cell Rep 2019; 27:3956-3971.e6. [PMID: 31242426 PMCID: PMC6613042 DOI: 10.1016/j.celrep.2019.05.095] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/04/2019] [Accepted: 05/22/2019] [Indexed: 12/25/2022] Open
Abstract
Senescence is a cellular phenotype present in health and disease, characterized by a stable cell-cycle arrest and an inflammatory response called senescence-associated secretory phenotype (SASP). The SASP is important in influencing the behavior of neighboring cells and altering the microenvironment; yet, this role has been mainly attributed to soluble factors. Here, we show that both the soluble factors and small extracellular vesicles (sEVs) are capable of transmitting paracrine senescence to nearby cells. Analysis of individual cells internalizing sEVs, using a Cre-reporter system, show a positive correlation between sEV uptake and senescence activation. We find an increase in the number of multivesicular bodies during senescence in vivo. sEV protein characterization by mass spectrometry (MS) followed by a functional siRNA screen identify interferon-induced transmembrane protein 3 (IFITM3) as being partially responsible for transmitting senescence to normal cells. We find that sEVs contribute to paracrine senescence.
Collapse
Affiliation(s)
- Michela Borghesan
- Epigenetics & Cellular Senescence Group, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK; Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Juan Fafián-Labora
- Epigenetics & Cellular Senescence Group, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK; Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Olga Eleftheriadou
- Epigenetics & Cellular Senescence Group, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK; Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Paula Carpintero-Fernández
- Epigenetics & Cellular Senescence Group, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK; Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Marta Paez-Ribes
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, Hutchison/MRC Research Centre, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Gema Vizcay-Barrena
- Centre for Ultrastructure Imaging, King's College London, London SE1 1UL, UK
| | - Avital Swisa
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Dror Kolodkin-Gal
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Pilar Ximénez-Embún
- Proteomics Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain; ProteoRed-ISCIII, Autonomous University of Madrid Campus, Cantoblanco, Madrid 28049, Spain
| | - Robert Lowe
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Belen Martín-Martín
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Hector Peinado
- Microenvironment and Metastasis Group, Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), Madrid 28029, Spain
| | - Javier Muñoz
- Proteomics Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain; ProteoRed-ISCIII, Autonomous University of Madrid Campus, Cantoblanco, Madrid 28049, Spain
| | - Roland A Fleck
- Centre for Ultrastructure Imaging, King's College London, London SE1 1UL, UK
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ittai Ben-Porath
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Anna Vossenkamper
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Daniel Muñoz-Espin
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, Hutchison/MRC Research Centre, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Ana O'Loghlen
- Epigenetics & Cellular Senescence Group, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK; Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK.
| |
Collapse
|
231
|
Slonchak A, Clarke B, Mackenzie J, Amarilla AA, Setoh YX, Khromykh AA. West Nile virus infection and interferon alpha treatment alter the spectrum and the levels of coding and noncoding host RNAs secreted in extracellular vesicles. BMC Genomics 2019; 20:474. [PMID: 31182021 PMCID: PMC6558756 DOI: 10.1186/s12864-019-5835-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are small membrane vesicles secreted by the cells that mediate intercellular transfer of molecules and contribute to transduction of various signals. Viral infection and action of pro-inflammatory cytokines has been shown to alter molecular composition of EV content. Transfer of antiviral proteins by EVs is thought to contribute to the development of inflammation and antiviral state. Altered incorporation of selected host RNAs into EVs in response to infection has also been demonstrated for several viruses, but not for WNV. Considering the medical significance of flaviviruses and the importance of deeper knowledge about the mechanisms of flavivirus-host interactions we assessed the ability of West Nile virus (WNV) and type I interferon (IFN), the main cytokine regulating antiviral response to WNV, to alter the composition of EV RNA cargo. RESULTS We employed next generation sequencing to perform transcriptome-wide profiling of RNA cargo in EVs produced by cells infected with WNV or exposed to IFN-alpha. RNA profile of EVs secreted by uninfected cells was also determined and used as a reference. We found that WNV infection significantly changed the levels of certain host microRNAs (miRNAs), small noncoding RNAs (sncRNAs) and mRNAs incorporated into EVs. Treatment with IFN-alpha also altered miRNA and mRNA profiles in EV but had less profound effect on sncRNAs. Functional classification of RNAs differentially incorporated into EVs upon infection and in response to IFN-alpha treatment demonstrated association of enriched in EVs mRNAs and miRNAs with viral processes and pro-inflammatory pathways. Further analysis revealed that WNV infection and IFN-alpha treatment changed the levels of common and unique mRNAs and miRNAs in EVs and that IFN-dependent and IFN-independent processes are involved in regulation of RNA sorting into EVs during infection. CONCLUSIONS WNV infection and IFN-alpha treatment alter the spectrum and the levels of mRNAs, miRNAs and sncRNAs in EVs. Differentially incorporated mRNAs and miRNAs in EVs produced in response to WNV infection and to IFN-alpha treatment are associated with viral processes and host response to infection. WNV infection affects composition of RNA cargo in EVs via IFN-dependent and IFN-independent mechanisms.
Collapse
Affiliation(s)
- Andrii Slonchak
- The Australian Infectious Diseases Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, MBS building 76, Cooper Rd, St Lucia, QLD, 4072, Australia
| | - Brian Clarke
- The Australian Infectious Diseases Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, MBS building 76, Cooper Rd, St Lucia, QLD, 4072, Australia
- The Pirbright Institute, Ash Rd, Pirbright, Surrey, GU24 GNF, UK
| | - Jason Mackenzie
- The Peter Doherty Institute for Infection and Immunity, Department of Microbiology and Immunology, The University of Melbourne, 792 Elizabeth Street, Melbourne, VIC, 3000, Australia
| | - Alberto Anastacio Amarilla
- The Australian Infectious Diseases Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, MBS building 76, Cooper Rd, St Lucia, QLD, 4072, Australia
| | - Yin Xiang Setoh
- The Australian Infectious Diseases Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, MBS building 76, Cooper Rd, St Lucia, QLD, 4072, Australia
| | - Alexander A Khromykh
- The Australian Infectious Diseases Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, MBS building 76, Cooper Rd, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
232
|
Shi Y, Du L, Lv D, Li H, Shang J, Lu J, Zhou L, Bai L, Tang H. Exosomal Interferon-Induced Transmembrane Protein 2 Transmitted to Dendritic Cells Inhibits Interferon Alpha Pathway Activation and Blocks Anti-Hepatitis B Virus Efficacy of Exogenous Interferon Alpha. Hepatology 2019; 69:2396-2413. [PMID: 30723923 PMCID: PMC6593428 DOI: 10.1002/hep.30548] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 01/30/2019] [Indexed: 02/05/2023]
Abstract
The negative regulators in the interferon (IFN) signaling pathway inhibit intrahepatic immune response, resulting in suboptimal therapeutic response to IFNα treatment in chronic hepatitis B (CHB) patients. Identifying the key negative factors and elucidating the regulating mechanism are essential for improving anti-HBV (hepatitis B virus) efficacy of IFNα. From the Gene Expression Omnibus (GEO) database, we downloaded and analyzed gene expression profiles of CHB patients with different responses to IFNα (GSE54747), and found that innate immune status was associated with the IFNα-based therapeutic response in CHB patients. Through PCR array, we found higher baseline level of IFN-induced transmembrane protein 2 (IFITM2) mRNA and lower baseline level of IFNα mRNA in peripheral blood mononuclear cells (PBMCs) of CHB patients with suboptimal response to IFNα treatment. Increased IFITM2 protein was also found in the serum of IFNα nonresponsive patients. With further experiments, we found that overexpressing IFITM2 in Huh7 cells suppressed endogenous IFNα synthesis by inhibiting phosphorylation of extracellular signal-regulated kinase (ERK), TANK-binding kinase 1 (TBK1), and interferon regulatory factor 3 (IRF3); knocking out IFITM2 enhanced activation of the endogenous IFNα synthesis pathway, exhibiting better inhibition on HBV replication. We also found that IFITM2 protein was shuttled by exosomes to dendritic cells (DCs), the main source of endogenous IFNα. Exosome-mediated transport of IFITM2 inhibited synthesis of endogenous IFNα in DCs whereas the inhibitory effect was abolished when IFITM2 was knocked out. Furthermore, we demonstrated that both palmitoylation inhibitor and mutation on 70/71 sites of IFITM2 protein influenced its incorporation into exosomes. Mutated IFITM2 protein increased the effect of IFNα against HBV. Conclusion: Exosome-mediated transport of IFITM2 to DCs inhibits IFNα pathway activation and blocks anti-HBV efficacy of exogenous IFNα. The findings provide an explanation to the suboptimal response of CHB patients to IFNα treatment.
Collapse
Affiliation(s)
- Ying Shi
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China HospitalSichuan UniversityChengduChina,Center of Infectious DiseasesWest China Hospital of Sichuan UniversityChengduChina
| | - Lingyao Du
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China HospitalSichuan UniversityChengduChina,Center of Infectious DiseasesWest China Hospital of Sichuan UniversityChengduChina
| | - Duoduo Lv
- Center of Infectious DiseasesWest China Hospital of Sichuan UniversityChengduChina
| | - Hong Li
- Center of Infectious DiseasesWest China Hospital of Sichuan UniversityChengduChina
| | - Jin Shang
- Center of Infectious DiseasesWest China Hospital of Sichuan UniversityChengduChina
| | - Jiajie Lu
- Center of Infectious DiseasesWest China Hospital of Sichuan UniversityChengduChina
| | - Lingyun Zhou
- Center of Infectious DiseasesWest China Hospital of Sichuan UniversityChengduChina
| | - Lang Bai
- Center of Infectious DiseasesWest China Hospital of Sichuan UniversityChengduChina
| | - Hong Tang
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China HospitalSichuan UniversityChengduChina,Center of Infectious DiseasesWest China Hospital of Sichuan UniversityChengduChina
| |
Collapse
|
233
|
Exosomes Modulate the Viral Replication and Host Immune Responses in HBV Infection. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2103943. [PMID: 31275965 PMCID: PMC6558633 DOI: 10.1155/2019/2103943] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/21/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022]
Abstract
Although current diagnosis and treatment of hepatitis B virus (HBV) infection can maintain viral suppression, new therapies need to be invented to sustain off-treatment virologic suppression and reduce side effects. Exosomes act as intercellular communicators to facilitate direct transfer of proteins, lipids, and nucleic acids between cells in vitro and in vivo. Pioneering work has demonstrated that exosomal cargos changed markedly during HBV infection. An improved understanding of the functions of exosomes during HBV infection could lead to a powerful new strategy for preventing and treating HBV. In this review, we point out the role of exosomes in HBV infection: (1) exosomes could directly participate in HBV replication; (2) exosomes modulate immune response during HBV infections; (3) exosomal RNAs and proteins might be selected as novel biomarkers for the diagnosis of HBV infections; and (4) exosomes can also be designed as vaccines.
Collapse
|
234
|
Ouattara LA, Anderson SM, Doncel GF. Seminal exosomes and HIV-1 transmission. Andrologia 2019; 50:e13220. [PMID: 30569645 PMCID: PMC6378409 DOI: 10.1111/and.13220] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 11/16/2018] [Accepted: 11/22/2018] [Indexed: 12/11/2022] Open
Abstract
Exosomes are endosomal‐derived membrane‐confined nanovesicles secreted by many (if not all) cell types and isolated from every human bodily fluid examined up to now including plasma, semen, vaginal secretions and breast milk. Exosomes are thought to represent a new player in cell‐to‐cell communication pathways and immune regulation, and be involved in many physiological and pathological processes. Susceptibility to HIV‐1 infection can be impacted by exosomes, while HIV‐1 pathogenesis can alter exosomal function and composition. Exosomes isolated from semen and vaginal fluid of healthy individuals can inhibit HIV‐1 infection and/or potently block viral transfer in vitro. However, the role of exosomes in HIV‐1 transmission and progression is not fully understood yet and some studies show conflicting results, mainly for exosomes isolated from plasma and breast milk. Determining the composition of exosomes from infected donors and studying their interaction with HIV‐1 in vitro compared to exosomes isolated from uninfected donors will provide insights into the role exosomes play in HIV‐1 transmission during sexual intercourse and breastfeeding.
Collapse
|
235
|
Jeon H, Lee J, Lee S, Kang SK, Park SJ, Yoo SM, Lee MS. Extracellular Vesicles From KSHV-Infected Cells Stimulate Antiviral Immune Response Through Mitochondrial DNA. Front Immunol 2019; 10:876. [PMID: 31068945 PMCID: PMC6491682 DOI: 10.3389/fimmu.2019.00876] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/05/2019] [Indexed: 12/24/2022] Open
Abstract
Kaposi's Sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma, which is the most common cancer in acquired immune deficiency syndrome patients. KSHV contains a variety of immunoregulatory proteins. There have been many studies on the modulation of antiviral response by these immunoregulatory proteins of KSHV. However, the antiviral effects of extracellular vesicles (EVs) during de novo KSHV infection have not been investigated to our best knowledge. In this study, we showed that KSHV-infected cells induce interferon-stimulated genes (ISGs) response but not type I interferon in uninfected bystander cells using EVs. mRNA microarray analysis showed that ISGs and IRF-activating genes were prominently activated in EVs from KSHV-infected cells (KSHV EVs)-treated human endothelial cells, which were validated by RT-qPCR and western blot analysis. We also found that this response was not associated with cell death or apoptosis by virus infection. Mechanistically, the cGAS-STING pathway was linked with these KSHV EVs-mediated ISGs expressions, and mitochondrial DNA on the surface of KSHV EVs was one of the causative factors. Besides, KSHV EVs-treated cells showed lower infectivity for KSHV and viral replication activity than mock EVs-treated cells. Our results indicate that EVs from KSHV-infected cells could be an initiating factor for the innate immune response against viral infection, which may be critical to understanding the microenvironment of virus-infected cells.
Collapse
Affiliation(s)
- Hyungtaek Jeon
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Jisu Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Suhyuk Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Su-Kyung Kang
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Sang June Park
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Seung-Min Yoo
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| | - Myung-Shin Lee
- Department of Microbiology and Immunology, Eulji University School of Medicine, Daejeon, South Korea
| |
Collapse
|
236
|
Chen J, Jin L, Yan M, Yang Z, Wang H, Geng S, Gong Z, Liu G. Serum Exosomes from Newborn Piglets Restrict Porcine Epidemic Diarrhea Virus Infection. J Proteome Res 2019; 18:1939-1947. [PMID: 30983354 DOI: 10.1021/acs.jproteome.9b00195] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Exosomes are vehicles in the body fluid that participate in many biological processes, especially immune responses. In this study, we employed comparative proteome analysis to investigate the roles of serum exosomes during viral infection in neonates using porcine epidemic diarrhea virus (PEDV), a devastating enteric virus in newborn piglets, as a model virus. Serum exosomes were first isolated from newborn piglets infected with PEDV or mock-infected newborn piglets, followed by label-free LC-MS/MS-based comparative quantitative proteomic analysis. Among the 441 detected proteins, 10 complement proteins were found in the serum exosomes, and significantly decreased expression levels of the C3, C6, and CFB complements were measured in PEDV-infected serum exosomes compared to those in mock-infected serum exosomes. After confirmation by Western blot, we then investigated the function of these exosomes in PEDV infection and discovered that exosomes from mock-infected newborn piglets restricted PEDV infection. However, this inhibition disappeared after the exosomes were heat-inactivated, suggesting that complements are key antiviral molecules. Our findings improve the understanding of antiviral responses mediated by exosomes in neonatal piglets and facilitate the discovery of novel antiviral drugs.
Collapse
Affiliation(s)
- Jianing Chen
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , Gansu 730046 , China
| | - Li Jin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , Gansu 730046 , China
| | - Miaomiao Yan
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , Gansu 730046 , China
| | - Ze Yang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , Gansu 730046 , China.,The First Affiliated Hospital of Lanzhou University , Lanzhou , Gansu 730000 , China
| | - Haiwen Wang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , Gansu 730046 , China
| | - Shuxian Geng
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , Gansu 730046 , China.,School of Veterinary Medicine , Gansu Agricultural University , Lanzhou , Gansu 730070 , China
| | - Zhenli Gong
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , Gansu 730046 , China
| | - Guangliang Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute , Chinese Academy of Agricultural Sciences , Lanzhou , Gansu 730046 , China
| |
Collapse
|
237
|
Sheveleva ON, Domaratskaya EI, Payushina OV. Extracellular Vesicles and Prospects of Their Use for Tissue Regeneration. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2019. [DOI: 10.1134/s1990747818040104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
238
|
Chen H, Kasagi S, Chia C, Zhang D, Tu E, Wu R, Zanvit P, Goldberg N, Jin W, Chen W. Extracellular Vesicles from Apoptotic Cells Promote TGFβ Production in Macrophages and Suppress Experimental Colitis. Sci Rep 2019; 9:5875. [PMID: 30971739 PMCID: PMC6458171 DOI: 10.1038/s41598-019-42063-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 12/12/2018] [Indexed: 11/09/2022] Open
Abstract
The clearance of apoptotic cells is an essential process to maintain homeostasis of immune system, which is regulated by immunoregulatory cytokines such as TGFβ. We show here that Extracellular Vesicles (EVs) were highly released from apoptotic cells, and contributed to macrophage production of TGFβ in vitro and in vivo. We further elucidated mechanistically that phosphatidylserine in EVs was a key triggering-factor, and transcription factor FOXO3 was a critical mediator for apoptotic EV-induced TGFβ in macrophages. Importantly, we found that macrophages pre-exposed to EVs exhibited an anti-inflammatory phenotype. More strikingly, administration of EVs in vivo promotes Tregs differentiation and suppresses Th1 cell response, and ameliorates experimental colitis. Thus, apoptotic-EV-based treatment might be a promising therapeutic approach for human autoimmune disease.
Collapse
Affiliation(s)
- Hua Chen
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA
| | - Shimpei Kasagi
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA
| | - Cheryl Chia
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA
| | - Dunfang Zhang
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA
| | - Eric Tu
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA
| | - Ruiqing Wu
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA
| | - Peter Zanvit
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA
| | - Nathan Goldberg
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA
| | - Wenwen Jin
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA
| | - WanJun Chen
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
239
|
Wang J, Teng Y, Zhao G, Li F, Hou A, Sun B, Kong W, Gao F, Cai L, Jiang C. Exosome-Mediated Delivery of Inducible miR-423-5p Enhances Resistance of MRC-5 Cells to Rabies Virus Infection. Int J Mol Sci 2019; 20:ijms20071537. [PMID: 30934732 PMCID: PMC6479321 DOI: 10.3390/ijms20071537] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 12/19/2022] Open
Abstract
The human diploid cell line Medical Research Council -5 (MRC-5) is commonly utilized for vaccine development. Although a rabies vaccine developed in cultured MRC-5 cells exists, the poor susceptibility of MRC-5 cells to the rabies virus (RABV) infection limits the potential yield of this vaccine. The underlying mechanism of MRC-5 cell resistance to RABV infection remains unknown. In this study, we demonstrate that viral infection increased exosomal release from MRC-5 cells; conversely, blocking exosome release promoted RABV infection in MRC-5 cells. Additionally, RABV infection up-regulated microRNA (miR)-423-5p expression in exosomes, resulting in feedback inhibition of RABV replication by abrogating the inhibitory effect of suppressor of cytokine signaling 3 (SOCS3) on type I interferon (IFN) signaling. Furthermore, intercellular delivery of miR-423-5p by exosomes inhibited RABV replication in MRC-5 cells. We also show that RABV infection increased IFN-β production in MRC-5 cells and that blocking the type I IFN receptor promoted RABV infection. In conclusion, MRC-5 cells were protected from RABV infection by the intercellular delivery of exosomal miR-423-5p and the up-regulation of IFN-β. These findings reveal novel antiviral mechanisms in MRC-5 cells against RABV infection. miR-423-5p, exosomes, and IFN signaling pathways may therefore be potential targets for improving MRC-5 cell-based rabies vaccine production.
Collapse
Affiliation(s)
- Jingyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
| | - Yawei Teng
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
| | - Guanshu Zhao
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
| | - Fang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
| | - Ali Hou
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Bo Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Feng Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Linjun Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| |
Collapse
|
240
|
Cerebrospinal fluid extracellular vesicles and neurofilament light protein as biomarkers of central nervous system injury in HIV-infected patients on antiretroviral therapy. AIDS 2019; 33:615-625. [PMID: 30557159 PMCID: PMC6399073 DOI: 10.1097/qad.0000000000002121] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective: The relationship of cerebrospinal fluid (CSF) extracellular vesicles to neurocognitive impairment (NCI) in HIV-infected individuals is unclear. Here, we characterize CSF extracellular vesicles and their association with central nervous system (CNS) injury related biomarkers [neurofilament light (NFL), S100B, neopterin] and NCI in HIV-positive individuals on combination antiretroviral therapy (cART). Design: A cross-sectional and longitudinal study of CSF samples from HIV-positive individuals on cART. Methods: NFL, S100B and neopterin were measured by ELISA in 190 CSF samples from 112 individuals (67 HIV-positive and 45 HIV-negative). CSF extracellular vesicles were isolated and characterized by electron microscopy, nanoparticle tracking analysis, immunoblotting for exosome markers (CD9, CD63, CD81, FLOT-1) and ELISA for HLA-DR. Results: HIV-positive individuals had median age 52 years, 67% with suppressed plasma viral load (< 50 copies/ml), median CD4+ nadir 66 cells/μl and CD4+ cell count 313 cells/μl. CSF NFL, S100B and neopterin levels were higher in HIV-positive vs. HIV-negative individuals, and nonsuppressed vs. suppressed HIV-positive individuals. Although CSF NFL and S100B levels were higher in NCI vs. unimpaired HIV-positive individuals (P < 0.05), only NFL was associated with NCI in adjusted models (P < 0.05). CSF extracellular vesicles were increased in HIV-positive vs. HIV-negative individuals, and NCI vs. unimpaired HIV-positive individuals (P < 0.05), and correlated positively with NFL (P < 0.001). HLA-DR was enriched in CSF extracellular vesicles from HIV-positive individuals with NCI (P < 0.05), suggesting that myeloid cells are a potential source of CSF extracellular vesicles during HIV infection. Conclusion: Increased CSF extracellular vesicles correlate with neuronal injury biomarker NFL in cART-treated HIV-positive individuals with neurocognitive impairment, suggesting potential applications as novel biomarkers of CNS injury.
Collapse
|
241
|
Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Inhibit Endometrial Cancer Cell Proliferation and Migration through Delivery of Exogenous miR-302a. Stem Cells Int 2019; 2019:8108576. [PMID: 31001342 PMCID: PMC6437733 DOI: 10.1155/2019/8108576] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/30/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023] Open
Abstract
MicroRNAs (miRNAs) are potential therapeutic targets in endometrial cancer, but the difficulties associated with their delivery to tumor target cells have hampered their applications. Human umbilical cord mesenchymal stem cells (hUCMSCs) have a well-recognized tumor-homing ability, emphasizing the capacity of tumor-targeted delivery of extracellular vesicles. hUCMSCs release extracellular vesicles rich in miRNAs, which play a vital role in intercellular communication. The purpose of this study was to verify a potential tumor suppressor microRNA, miR-302a, and engineered hUCMSC extracellular vesicles enriched with miR-302a for therapy of endometrial cancer. Here, we observed that miR-302a was significantly downregulated in endometrial cancer tissues when compared with adjacent tissues. Overexpression of miR-302a in endometrial cancer cells robustly suppressed cell proliferation and migration. Meanwhile, the proliferation and migration were significantly inhibited in endometrial cancer cells when cultured with miR-302a-loaded extracellular vesicles derived from hUCMSCs. Importantly, our data showed that engineered extracellular vesicles rich in miR-302 significantly inhibited the expression of cyclin D1 and suppressed AKT signaling pathway in endometrial cancer cells. These results suggested that exogenous miR-302a delivered by hUCMSC-derived extracellular vesicles has exciting potential as an effective anticancer therapy.
Collapse
|
242
|
Song H, Tan G, Yang Y, Cui A, Li H, Li T, Wu Z, Yang M, Lv G, Chi X, Niu J, Zhu K, Crispe IN, Su L, Tu Z. Hepatitis B Virus-Induced Imbalance of Inflammatory and Antiviral Signaling by Differential Phosphorylation of STAT1 in Human Monocytes. THE JOURNAL OF IMMUNOLOGY 2019; 202:2266-2275. [PMID: 30842274 DOI: 10.4049/jimmunol.1800848] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 02/12/2019] [Indexed: 12/14/2022]
Abstract
It is not clear how hepatitis B virus (HBV) modulates host immunity during chronic infection. In addition to the key mediators of inflammatory response in viral infection, monocytes also express a high-level IFN-stimulated gene, CH25H, upon response to IFN-α exerting an antiviral effect. In this study, the mechanism by which HBV manipulates IFN signaling in human monocytes was investigated. We observed that monocytes from chronic hepatitis B patients express lower levels of IFN signaling/stimulated genes and higher levels of inflammatory cytokines compared with healthy donors. HBV induces monocyte production of inflammatory cytokines via TLR2/MyD88/NF-κB signaling and STAT1-Ser727 phosphorylation and inhibits IFN-α-induced stat1, stat2, and ch25h expression through the inhibition of STAT1-Tyr701 phosphorylation and in an IL-10-dependent, partially autocrine manner. Further, we found that enhancement of STAT1 activity with a small molecule (2-NP) rescued HBV-mediated inhibition of IFN signaling and counteracted the induction of inflammatory cytokines. In conclusion, HBV contributes to the monocyte inflammatory response but inhibits their IFN-α/β responsiveness to impair antiviral innate immunity. These effects are mediated via differential phosphorylation of Tyr701 and Ser727 of STAT1.
Collapse
Affiliation(s)
- Hongxiao Song
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Guangyun Tan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yang Yang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - An Cui
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Haijun Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Tianyang Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhihui Wu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Miaomiao Yang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xiumei Chi
- Institute of Liver Diseases, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Junqi Niu
- Institute of Liver Diseases, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Ian Nicholas Crispe
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.,Department of Pathology, University of Washington, Seattle, WA 98195; and
| | - Lishan Su
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.,Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Zhengkun Tu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China; .,Institute of Liver Diseases, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
243
|
Li I, Nabet BY. Exosomes in the tumor microenvironment as mediators of cancer therapy resistance. Mol Cancer 2019; 18:32. [PMID: 30823926 PMCID: PMC6397467 DOI: 10.1186/s12943-019-0975-5] [Citation(s) in RCA: 302] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/22/2019] [Indexed: 02/08/2023] Open
Abstract
Exosomes are small extracellular vesicles that contain genetic material, proteins, and lipids. They function as potent signaling molecules between cancer cells and the surrounding cells that comprise the tumor microenvironment (TME). Exosomes derived from both tumor and stromal cells have been implicated in all stages of cancer progression and play an important role in therapy resistance. Moreover, due to their nature as mediators of cell-cell communication, they are integral to TME-dependent therapy resistance. In this review, we discuss current exosome isolation and profiling techniques and their role in TME interactions and therapy resistance. We also explore emerging clinical applications of both exosomes as biomarkers, direct therapeutic targets, and engineered nanocarriers. In order to fully understand the TME, careful interrogation of exosomes and their cargo is critical. This understanding is a promising avenue for the development of effective clinical applications.
Collapse
Affiliation(s)
- Irene Li
- Stanford Cancer Biology Program, Stanford University, 318 Campus Drive, Stanford, CA 94305 USA
- Department of Radiology, Stanford University, 318 Campus Drive, Stanford, CA 94305 USA
| | - Barzin Y. Nabet
- Department of Radiation Oncology, Stanford University, 265 Campus Drive, Stanford, CA 94305 USA
- Stanford Cancer Institute, Stanford University, 265 Campus Drive, Stanford, CA 94305 USA
| |
Collapse
|
244
|
Welch JL, Stapleton JT, Okeoma CM. Vehicles of intercellular communication: exosomes and HIV-1. J Gen Virol 2019; 100:350-366. [PMID: 30702421 PMCID: PMC7011712 DOI: 10.1099/jgv.0.001193] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/15/2018] [Indexed: 12/20/2022] Open
Abstract
The terms extracellular vesicles, microvesicles, oncosomes, or exosomes are often used interchangeably as descriptors of particles that are released from cells and comprise a lipid membrane that encapsulates nucleic acids and proteins. Although these entities are defined based on a specific size range and/or mechanism of release, the terminology is often ambiguous. Nevertheless, these vesicles are increasingly recognized as important modulators of intercellular communication. The generic characterization of extracellular vesicles could also be used as a descriptor of enveloped viruses, highlighting the fact that extracellular vesicles and enveloped viruses are similar in both composition and function. Their high degree of similarity makes differentiating between vesicles and enveloped viruses in biological specimens particularly difficult. Because viral particles and extracellular vesicles are produced simultaneously in infected cells, it is necessary to separate these populations to understand their independent functions. We summarize current understanding of the similarities and differences of extracellular vesicles, which henceforth we will refer to as exosomes, and the enveloped retrovirus, HIV-1. Here, we focus on the presence of these particles in semen, as these are of particular importance during HIV-1 sexual transmission. While there is overlap in the terminology and physical qualities between HIV-1 virions and exosomes, these two types of intercellular vehicles may differ depending on the bio-fluid source. Recent data have demonstrated that exosomes from human semen serve as regulators of HIV-1 infection that may contribute to the remarkably low risk of infection per sexual exposure.
Collapse
Affiliation(s)
- Jennifer L. Welch
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242-1109, USA
- Medical Service, Iowa City Veterans Affairs Medical Center, University of Iowa, 604 Highway 6, Iowa City, IA 52246-2208, USA
| | - Jack T. Stapleton
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242-1109, USA
- Medical Service, Iowa City Veterans Affairs Medical Center, University of Iowa, 604 Highway 6, Iowa City, IA 52246-2208, USA
| | - Chioma M. Okeoma
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
- Department of Pharmacologic Sciences, Basic Sciences Tower, Rm 8-142, Stony Brook, University School of Medicine, Stony Brook, NY 11794-8651, USA
| |
Collapse
|
245
|
Picornavirus infection induces temporal release of multiple extracellular vesicle subsets that differ in molecular composition and infectious potential. PLoS Pathog 2019; 15:e1007594. [PMID: 30779790 PMCID: PMC6396942 DOI: 10.1371/journal.ppat.1007594] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 03/01/2019] [Accepted: 01/24/2019] [Indexed: 02/04/2023] Open
Abstract
Several naked virus species, including members of the Picornaviridae family, have recently been described to escape their host cells and spread infection via enclosure in extracellular vesicles (EV). EV are 50-300 nm sized lipid membrane-enclosed particles produced by all cells that are broadly recognized for playing regulatory roles in numerous (patho)physiological processes, including viral infection. Both pro- and antiviral functions have been ascribed to EV released by virus-infected cells. It is currently not known whether this reported functional diversity is a result of the release of multiple virus-containing and non-virus containing EV subpopulations that differ in composition and function. Using encephalomyocarditis virus infection (EMCV, Picornaviridae family), we here provide evidence that EV populations released by infected cells are highly heterogeneous. Virus was contained in two distinct EV populations that differed in physical characteristics, such as sedimentation properties, and in enrichment for proteins indicative of different EV biogenesis pathways, such as the plasma membrane resident proteins Flotillin-1 and CD9, and the autophagy regulatory protein LC3. Additional levels of EV heterogeneity were identified using high-resolution flow cytometric analysis of single EV. Importantly, we demonstrate that EV subsets released during EMCV infection varied largely in potency of transferring virus infection and in their kinetics of release from infected cells. These data support the notion that heterogeneous EV populations released by virus-infected cells can exert diverse functions at distinct time points during infection. Unraveling the compositional, temporal and functional heterogeneity of these EV populations using single EV analysis technologies, as employed in this study, is vital to understanding the role of EV in virus dissemination and antiviral host responses.
Collapse
|
246
|
Exosomes Released from Rabies Virus-Infected Cells May be Involved in the Infection Process. Virol Sin 2019; 34:59-65. [PMID: 30725320 DOI: 10.1007/s12250-019-00087-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 12/10/2018] [Indexed: 12/25/2022] Open
Abstract
Exosomes are cell-derived vesicles that are secreted by many eukaryotic cells. It has recently attracted attention as vehicles of intercellular communication. Virus-infected cells release exosomes, which contain viral proteins, RNA, and pathogenic molecules. However, the role of exosomes in virus infection process remains unclear and needs to be further investigated. In this study, we aimed to evaluate the effects of exosomes on rabies virus infection. OptiPrep™ density gradient centrifugation was used to isolate exosomes from rabies virus-infected cell culture supernatants. A rabies virus G protein enzyme-linked immunosorbent assay and acetylcholinesterase activity assays were performed to verify the centrifugation fractions. Exosomes were then characterized using transmission electron microscopy and Western blotting. Our results showed that rabies virus infection increased the release of exosomes. Treatment with GW4869 and si-Rab27a, two exosomal secretion inhibitors, inhibited exosome release. Furthermore, the inhibitors reduced the levels of extracellular and intracellular viral RNA. These data indicated that exosomes may participate in the viral infection process. Moreover, our results establish a basis for future research into the roles of exosomes in rabies virus infection and as potential targets for developing new antiviral strategies.
Collapse
|
247
|
Kim JH, Lee CH, Lee SW. Exosomal Transmission of MicroRNA from HCV Replicating Cells Stimulates Transdifferentiation in Hepatic Stellate Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 14:483-497. [PMID: 30753992 PMCID: PMC6369229 DOI: 10.1016/j.omtn.2019.01.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 12/12/2022]
Abstract
The mechanism by which hepatitis C virus (HCV) causes fibrosis and other chronic liver diseases remains poorly understood. Previously, we observed that HCV infection induces microRNA-192 (miR-192) expression, which in turn upregulates transforming growth factor β1 (TGF-β1) in hepatocytes. In this study, we aimed to determine the roles and mechanisms of HCV-induced miR-192 expression during chronic liver injury and fibrosis and to identify potential target of the liver disease. Noticeably, miR-192 is secreted and transmitted through exosomes from HCV-replicating hepatocytes into hepatic stellate cells (HSCs). Exosomal transferred miR-192 upregulated fibrogenic markers in HSCs through TGF-β1 upregulation, resulting in the activation and transdifferentiation of HSCs into myofibroblasts. Anti-miR-192 treatment of HCV-replicating hepatocytes efficiently reduced miR-192 levels in exosomes, downregulated miR-192 and fibrogenic marker levels in HSCs, and impeded transdifferentiation of the cells. In contrast, miR-192 mimic RNA treatment significantly increased miR-192 levels in exosomes from naive hepatocytes, increased miR-192 and fibrogenic marker expression in HSCs, and induced transdifferentiation of the cells. Notably, transdifferentiation of exosome-exposed HSCs was reversed following treatment with anti-miR-192 into the HSCs. This study revealed a novel mechanism of HCV-induced liver fibrosis and identified exosomal miR-192 as a major regulator and potential treatment target for HCV-mediated hepatic fibrosis.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Integrated Life Sciences, Research Institute of Advanced Omics, Dankook University, 152, Jukjeon-ro, Suji-gu, Yongin 16890, Republic of Korea; Department of Molecular Biology, Dankook University, 119, Dandae-ro, Dongnam-gu, Cheonan 31116, Republic of Korea
| | - Chang Ho Lee
- Department of Integrated Life Sciences, Research Institute of Advanced Omics, Dankook University, 152, Jukjeon-ro, Suji-gu, Yongin 16890, Republic of Korea
| | - Seong-Wook Lee
- Department of Integrated Life Sciences, Research Institute of Advanced Omics, Dankook University, 152, Jukjeon-ro, Suji-gu, Yongin 16890, Republic of Korea.
| |
Collapse
|
248
|
Yang B, Chen Y, Shi J. Exosome Biochemistry and Advanced Nanotechnology for Next-Generation Theranostic Platforms. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1802896. [PMID: 30126052 DOI: 10.1002/adma.201802896] [Citation(s) in RCA: 255] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 06/07/2018] [Indexed: 05/26/2023]
Abstract
Recent marked technological advances in the field of exosome nanotechnology have provided unprecedented opportunities to bloom the developments of exosome-related biology, chemistry, pathology, and therapeutics, which have laid a solid basis for scientific community to design exosome-based nanotheranostic platforms. The unique structural/compositional/morphological characteristics of exosomes as natural nanocarriers, as well as their fascinating physicochemical/biochemical properties, which underpin their special physiopathological roles, have triggered the concept that these cell-derived nanovesicles with intrinsic biological functions can be highly competent for the establishment of next-generation nanomedicine. Herein, efforts are made to give a comprehensive overview on the recent advances of exosome nanotechnology based on the representative examples of the current state of the art of exosome-based research, ranging from their formation, biological function, preparation, and characterization to their extensive nanomedical applications. It is highly expected that the better and clearer elucidation of the fundamental principles for advanced nanotechnology in constructing exosome-based theranostic nanoplatforms, as well as integrating the intrinsic advantages of exosomes as endogenous cell-derived nanocarriers with the advanced design methodology of traditional nanomedicine, will help to unlock the innate powers of exosomes for the establishment of next-generation theranostic nanoplatforms.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
249
|
Cancer-derived exosomal miR-25-3p promotes pre-metastatic niche formation by inducing vascular permeability and angiogenesis. Nat Commun 2018; 9:5395. [PMID: 30568162 PMCID: PMC6300604 DOI: 10.1038/s41467-018-07810-w] [Citation(s) in RCA: 695] [Impact Index Per Article: 99.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 11/22/2018] [Indexed: 12/11/2022] Open
Abstract
Cancer-derived exosomes are considered a major driver of cancer-induced pre-metastatic niche formation at foreign sites, but the mechanisms remain unclear. Here, we show that miR-25-3p, a metastasis-promoting miRNA of colorectal cancer (CRC), can be transferred from CRC cells to endothelial cells via exosomes. Exosomal miR-25-3p regulates the expression of VEGFR2, ZO-1, occludin and Claudin5 in endothelial cells by targeting KLF2 and KLF4, consequently promotes vascular permeability and angiogenesis. In addition, exosomal miR-25-3p from CRC cells dramatically induces vascular leakiness and enhances CRC metastasis in liver and lung of mice. Moreover, the expression level of miR-25-3p from circulating exosomes is significantly higher in CRC patients with metastasis than those without metastasis. Our work suggests that exosomal miR-25-3p is involved in pre-metastatic niche formation and may be used as a blood-based biomarker for CRC metastasis.
Collapse
|
250
|
Yao Z, Jia X, Megger DA, Chen J, Liu Y, Li J, Sitek B, Yuan Z. Label-Free Proteomic Analysis of Exosomes Secreted from THP-1-Derived Macrophages Treated with IFN-α Identifies Antiviral Proteins Enriched in Exosomes. J Proteome Res 2018; 18:855-864. [DOI: 10.1021/acs.jproteome.8b00514] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Zhenlan Yao
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Xiaofang Jia
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Dominik A. Megger
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 50 Virchowstraße 179, 45147 Essen, Germany
| | - Jieliang Chen
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yuyi Liu
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jianhua Li
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Barbara Sitek
- Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| |
Collapse
|