401
|
Akagi T, Kato K, Kobayashi M, Kosaka N, Ochiya T, Ichiki T. On-chip immunoelectrophoresis of extracellular vesicles released from human breast cancer cells. PLoS One 2015; 10:e0123603. [PMID: 25928805 PMCID: PMC4415775 DOI: 10.1371/journal.pone.0123603] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 02/20/2015] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) including exosomes and microvesicles have attracted considerable attention in the fields of cell biology and medicine. For a better understanding of EVs and further exploration of their applications, the development of analytical methods for biological nanovesicles has been required. In particular, considering the heterogeneity of EVs, methods capable of measuring individual vesicles are desired. Here, we report that on-chip immunoelectrophoresis can provide a useful method for the differential protein expression profiling of individual EVs. Electrophoresis experiments were performed on EVs collected from the culture supernatant of MDA-MB-231 human breast cancer cells using a measurement platform comprising a microcapillary electrophoresis chip and a laser dark-field microimaging system. The zeta potential distribution of EVs that reacted with an anti-human CD63 (exosome and microvesicle marker) antibody showed a marked positive shift as compared with that for the normal immunoglobulin G (IgG) isotype control. Thus, on-chip immunoelectrophoresis could sensitively detect the over-expression of CD63 glycoproteins on EVs. Moreover, to explore the applicability of on-chip immunoelectrophoresis to cancer diagnosis, EVs collected from the blood of a mouse tumor model were analyzed by this method. By comparing the zeta potential distributions of EVs after their immunochemical reaction with normal IgG, and the anti-human CD63 and anti-human CD44 (cancer stem cell marker) antibodies, EVs of tumor origin circulating in blood were differentially detected in the real sample. The result indicates that the present method is potentially applicable to liquid biopsy, a promising approach to the low-invasive diagnosis of cancer.
Collapse
Affiliation(s)
- Takanori Akagi
- Department of Bioengineering, School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Kei Kato
- Department of Bioengineering, School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Masashi Kobayashi
- Department of Bioengineering, School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, Japan
| | - Nobuyoshi Kosaka
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo Japan
| | - Takanori Ichiki
- Department of Bioengineering, School of Engineering, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
402
|
Tan S, Wu T, Zhang D, Zhang Z. Cell or cell membrane-based drug delivery systems. Theranostics 2015; 5:863-81. [PMID: 26000058 PMCID: PMC4440443 DOI: 10.7150/thno.11852] [Citation(s) in RCA: 338] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 02/18/2015] [Indexed: 01/14/2023] Open
Abstract
Natural cells have been explored as drug carriers for a long period. They have received growing interest as a promising drug delivery system (DDS) until recently along with the development of biology and medical science. The synthetic materials, either organic or inorganic, are found to be with more or less immunogenicity and/or toxicity. The cells and extracellular vesicles (EVs), are endogenous and thought to be much safer and friendlier. Furthermore, in view of their host attributes, they may achieve different biological effects and/or targeting specificity, which can meet the needs of personalized medicine as the next generation of DDS. In this review, we summarized the recent progress in cell or cell membrane-based DDS and their fabrication processes, unique properties and applications, including the whole cells, EVs and cell membrane coated nanoparticles. We expect the continuing development of this cell or cell membrane-based DDS will promote their clinic applications.
Collapse
Affiliation(s)
- Songwei Tan
- 1. Tongji School of Pharmacy
- 2. National Engineering Research Center for Nanomedicine
- 3. Hubei Engineering Research Center for Novel DDS, Huazhong University of Science and Technology, Wuhan 430030, P R China
| | | | | | - Zhiping Zhang
- 1. Tongji School of Pharmacy
- 2. National Engineering Research Center for Nanomedicine
- 3. Hubei Engineering Research Center for Novel DDS, Huazhong University of Science and Technology, Wuhan 430030, P R China
| |
Collapse
|
403
|
Madison MN, Jones PH, Okeoma CM. Exosomes in human semen restrict HIV-1 transmission by vaginal cells and block intravaginal replication of LP-BM5 murine AIDS virus complex. Virology 2015; 482:189-201. [PMID: 25880110 DOI: 10.1016/j.virol.2015.03.040] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/20/2015] [Accepted: 03/23/2015] [Indexed: 11/17/2022]
Abstract
Exosomes are membranous extracellular nanovesicles secreted by diverse cell types. Exosomes from healthy human semen have been shown to inhibit HIV-1 replication and to impair progeny virus infectivity. In this study, we examined the ability of healthy human semen exosomes to restrict HIV-1 and LP-BM5 murine AIDS virus transmission in three different model systems. We show that vaginal cells internalize exosomes with concomitant transfer of functional mRNA. Semen exosomes blocked the spread of HIV-1 from vaginal epithelial cells to target cells in our cell-to-cell infection model and suppressed transmission of HIV-1 across the vaginal epithelial barrier in our trans-well model. Our in vivo model shows that human semen exosomes restrict intravaginal transmission and propagation of murine AIDS virus. Our study highlights an antiretroviral role for semen exosomes that may be harnessed for the development of novel therapeutic strategies to combat HIV-1 transmission.
Collapse
Affiliation(s)
- Marisa N Madison
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Philip H Jones
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | - Chioma M Okeoma
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA; Interdisciplinary Program in Molecular and Cellular Biology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
404
|
Smith JA, Leonardi T, Huang B, Iraci N, Vega B, Pluchino S. Extracellular vesicles and their synthetic analogues in aging and age-associated brain diseases. Biogerontology 2015; 16:147-85. [PMID: 24973266 PMCID: PMC4578234 DOI: 10.1007/s10522-014-9510-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Multicellular organisms rely upon diverse and complex intercellular communications networks for a myriad of physiological processes. Disruption of these processes is implicated in the onset and propagation of disease and disorder, including the mechanisms of senescence at both cellular and organismal levels. In recent years, secreted extracellular vesicles (EVs) have been identified as a particularly novel vector by which cell-to-cell communications are enacted. EVs actively and specifically traffic bioactive proteins, nucleic acids, and metabolites between cells at local and systemic levels, modulating cellular responses in a bidirectional manner under both homeostatic and pathological conditions. EVs are being implicated not only in the generic aging process, but also as vehicles of pathology in a number of age-related diseases, including cancer and neurodegenerative and disease. Thus, circulating EVs-or specific EV cargoes-are being utilised as putative biomarkers of disease. On the other hand, EVs, as targeted intercellular shuttles of multipotent bioactive payloads, have demonstrated promising therapeutic properties, which can potentially be modulated and enhanced through cellular engineering. Furthermore, there is considerable interest in employing nanomedicinal approaches to mimic the putative therapeutic properties of EVs by employing synthetic analogues for targeted drug delivery. Herein we describe what is known about the origin and nature of EVs and subsequently review their putative roles in biology and medicine (including the use of synthetic EV analogues), with a particular focus on their role in aging and age-related brain diseases.
Collapse
Affiliation(s)
- J A Smith
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, CB2 0PY, UK
| | | | | | | | | | | |
Collapse
|
405
|
Physical characterization and profiling of airway epithelial derived exosomes using light scattering. Methods 2015; 87:59-63. [PMID: 25823850 DOI: 10.1016/j.ymeth.2015.03.013] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/17/2015] [Accepted: 03/22/2015] [Indexed: 01/21/2023] Open
Abstract
Exosomes and other extracellular vesicles have been gaining interest during the last decade due to their emerging role in biology and, disease pathogenesis and their biomarker potential. Almost all published research related to exosomes and other extracellular vesicles include some form of physical characterization. Therefore, these vesicles should be precisely profiled and characterized physically before studying their biological role as intercellular messengers, biomarkers or therapeutic tools. Using a combination of light scattering techniques, including dynamic light scattering (DLS) and multi-angle laser light scattering combined with size exclusion separation (SEC-MALLS), we physically characterized and compared distinct extracellular vesicles derived from the apical secretions of two different cultured airway epithelial cells. The results indicated that epithelial cells release vesicles with distinct physical properties and sizes. Human primary tracheobronchial cell culture (HTBE) derived vesicles have a hydrodynamic radius (Rh) of approximately 340 nm while their radius of gyration (Rg) is approximately 200 nm. Electron microscopy analysis, however, revealed that their spherical component is 40-100 nm in size, and they carry filamentous, entangled membrane mucins on their surface that increases their overall radius. The mucin decoration on the surface defines their size and charge as measured using light scattering techniques. Their surface properties mirror the properties of the cells from which they are derived. This may provide a unique tool for researchers to elucidate the unanswered questions in normal airway biology and innate and adaptive defense, including the remodeling of airways during inflammation, tumorigenesis and metastasis.
Collapse
|
406
|
Joyner C, Barnwell JW, Galinski MR. No more monkeying around: primate malaria model systems are key to understanding Plasmodium vivax liver-stage biology, hypnozoites, and relapses. Front Microbiol 2015; 6:145. [PMID: 25859242 PMCID: PMC4374475 DOI: 10.3389/fmicb.2015.00145] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 02/07/2015] [Indexed: 01/17/2023] Open
Abstract
Plasmodium vivax is a human malaria parasite responsible for significant morbidity worldwide and potentially death. This parasite possesses formidable liver-stage biology that involves the formation of dormant parasites known as hypnozoites. Hypnozoites are capable of activating weeks, months, or years after a primary blood-stage infection causing relapsing bouts of illness. Elimination of this dormant parasitic reservoir will be critical for global malaria eradication. Although hypnozoites were first discovered in 1982, few advancements have been made to understand their composition and biology. Until recently, in vitro models did not exist to study these forms and studying them from human ex vivo samples was virtually impossible. Today, non-human primate (NHP) models and modern systems biology approaches are poised as tools to enable the in-depth study of P. vivax liver-stage biology, including hypnozoites and relapses. NHP liver-stage model systems for P. vivax and the related simian malaria species P. cynomolgi are discussed along with perspectives regarding metabolite biomarker discovery, putative roles of extracellular vesicles, and relapse immunobiology.
Collapse
Affiliation(s)
- Chester Joyner
- Malaria Host–Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory UniversityAtlanta, GA, USA
| | - John W. Barnwell
- Malaria Host–Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory UniversityAtlanta, GA, USA
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and PreventionAtlanta, GA, USA
| | - Mary R. Galinski
- Malaria Host–Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory UniversityAtlanta, GA, USA
- Division of Infectious Diseases, Department of Medicine, Emory UniversityAtlanta, GA, USA
| |
Collapse
|
407
|
Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell 2015; 159:499-513. [PMID: 25417103 DOI: 10.1016/j.cell.2014.09.051] [Citation(s) in RCA: 653] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 06/11/2014] [Accepted: 09/03/2014] [Indexed: 02/08/2023]
Abstract
Stromal communication with cancer cells can influence treatment response. We show that stromal and breast cancer (BrCa) cells utilize paracrine and juxtacrine signaling to drive chemotherapy and radiation resistance. Upon heterotypic interaction, exosomes are transferred from stromal to BrCa cells. RNA within exosomes, which are largely noncoding transcripts and transposable elements, stimulates the pattern recognition receptor RIG-I to activate STAT1-dependent antiviral signaling. In parallel, stromal cells also activate NOTCH3 on BrCa cells. The paracrine antiviral and juxtacrine NOTCH3 pathways converge as STAT1 facilitates transcriptional responses to NOTCH3 and expands therapy-resistant tumor-initiating cells. Primary human and/or mouse BrCa analysis support the role of antiviral/NOTCH3 pathways in NOTCH signaling and stroma-mediated resistance, which is abrogated by combination therapy with gamma secretase inhibitors. Thus, stromal cells orchestrate an intricate crosstalk with BrCa cells by utilizing exosomes to instigate antiviral signaling. This expands BrCa subpopulations adept at resisting therapy and reinitiating tumor growth.
Collapse
|
408
|
Osna NA, Ganesan M, Kharbanda KK. Hepatitis C, innate immunity and alcohol: friends or foes? Biomolecules 2015; 5:76-94. [PMID: 25664450 PMCID: PMC4384112 DOI: 10.3390/biom5010076] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/19/2015] [Accepted: 01/24/2015] [Indexed: 02/05/2023] Open
Abstract
Hepatitis C and alcohol are the most widespread causes of liver disease worldwide. Approximately 80% of patients with a history of hepatitis C and alcohol abuse develop chronic liver injury. Alcohol consumption in hepatitis C virus (HCV)-infected patients exacerbates liver disease leading to rapid progression of fibrosis, cirrhosis and even hepatocellular carcinoma. Hepatocytes are the main sites of HCV-infection and ethanol metabolism, both of which generate oxidative stress. Oxidative stress levels affect HCV replication and innate immunity, resulting in a greater susceptibility for HCV-infection and virus spread in the alcoholic patients. In this review paper, we analyze the effects of ethanol metabolism and other factors on HCV replication. In addition, we illustrate the mechanisms of how HCV hijacks innate immunity and how ethanol exposure regulates this process. We also clarify the effects of HCV and ethanol metabolism on interferon signaling-a crucial point for activation of anti-viral genes to protect cells from virus-and the role that HCV- and ethanol-induced impairments play in adaptive immunity which is necessary for recognition of virally-infected hepatocytes. In conclusion, ethanol exposure potentiates the suppressive effects of HCV on innate immunity, which activates viral spread in the liver and finally, leads to impairments in adaptive immunity. The dysregulation of immune response results in impaired elimination of HCV-infected cells, viral persistence, progressive liver damage and establishment of chronic infection that worsens the outcomes of chronic hepatitis C in alcoholic patients.
Collapse
Affiliation(s)
- Natalia A Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA.
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA.
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, 4101 Woolworth Ave, Omaha, NE 68105, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA.
| |
Collapse
|
409
|
Affiliation(s)
- Paul Klenerman
- NIHR Biomedical Research Centre and Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK.
| | - Narayan Ramamurthy
- NIHR Biomedical Research Centre and Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| |
Collapse
|
410
|
Giugliano S, Kriss M, Golden-Mason L, Dobrinskikh E, Stone AEL, Soto-Gutierrez A, Mitchell A, Khetani SR, Yamane D, Stoddard M, Li H, Shaw GM, Edwards MG, Lemon SM, Gale M, Shah VH, Rosen HR. Hepatitis C virus infection induces autocrine interferon signaling by human liver endothelial cells and release of exosomes, which inhibits viral replication. Gastroenterology 2015; 148:392-402.e13. [PMID: 25447848 PMCID: PMC4765499 DOI: 10.1053/j.gastro.2014.10.040] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 10/21/2014] [Accepted: 10/28/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Liver sinusoidal endothelial cells (LSECs) make up a large proportion of the nonparenchymal cells in the liver. LSECs are involved in induction of immune tolerance, but little is known about their functions during hepatitis C virus (HCV) infection. METHODS Primary human LSECs (HLSECs) and immortalized liver endothelial cells (TMNK-1) were exposed to various forms of HCV, including full-length transmitted/founder virus, sucrose-purified Japanese fulminant hepatitis-1 (JFH-1), a virus encoding a luciferase reporter, and the HCV-specific pathogen-associated molecular pattern molecules. Cells were analyzed by confocal immunofluorescence, immunohistochemical, and polymerase chain reaction assays. RESULTS HLSECs internalized HCV, independent of cell-cell contacts; HCV RNA was translated but not replicated. Through pattern recognition receptors (Toll-like receptor 7 and retinoic acid-inducible gene 1), HCV RNA induced consistent and broad transcription of multiple interferons (IFNs); supernatants from primary HLSECs transfected with HCV-specific pathogen-associated molecular pattern molecules increased induction of IFNs and IFN-stimulated genes in HLSECs. Recombinant type I and type III IFNs strongly up-regulated HLSEC transcription of IFN λ3 (IFNL3) and viperin (RSAD2), which inhibit replication of HCV. Compared with CD8(+) T cells, HLSECs suppressed HCV replication within Huh7.5.1 cells, also inducing IFN-stimulated genes in co-culture. Conditioned media from IFN-stimulated HLSECs induced expression of antiviral genes by uninfected primary human hepatocytes. Exosomes, derived from HLSECs after stimulation with either type I or type III IFNs, controlled HCV replication in a dose-dependent manner. CONCLUSIONS Cultured HLSECs produce factors that mediate immunity against HCV. HLSECs induce self-amplifying IFN-mediated responses and release of exosomes with antiviral activity.
Collapse
Affiliation(s)
- Silvia Giugliano
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Michael Kriss
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| | - Lucy Golden-Mason
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado; Integrated Department in Immunology: University of Colorado Denver and National Jewish Health, Denver, Colorado
| | - Evgenia Dobrinskikh
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Amy E L Stone
- Department of Immunology, University of Washington, School of Medicine, Seattle, Washington
| | - Alejandro Soto-Gutierrez
- Department of Pathology, Center for Innovative Regenerative Therapies, Department of Surgery, Transplantation Section, Children's Hospital of Pittsburgh, McGowan Institute for Regenerative Medicine and the Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Angela Mitchell
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado; Integrated Department in Immunology: University of Colorado Denver and National Jewish Health, Denver, Colorado
| | - Salman R Khetani
- Mechanical and Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| | - Daisuke Yamane
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Mark Stoddard
- Department of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Hui Li
- Department of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - George M Shaw
- Department of Medicine and Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Michael G Edwards
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Stanley M Lemon
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Michael Gale
- Department of Immunology, University of Washington, School of Medicine, Seattle, Washington
| | - Vijay H Shah
- Mayo Clinic, Division of Gastroenterology and Hepatology, Rochester, Minnesota
| | - Hugo R Rosen
- Division of Gastroenterology and Hepatology, Hepatitis C Center, Department of Medicine, University of Colorado, Denver, Aurora, Colorado; Integrated Department in Immunology: University of Colorado Denver and National Jewish Health, Denver, Colorado; Eastern Colorado Veteran's Affairs Medical Center, Denver, Colorado.
| |
Collapse
|
411
|
Rehermann B, Bertoletti A. Immunological aspects of antiviral therapy of chronic hepatitis B virus and hepatitis C virus infections. Hepatology 2015; 61:712-21. [PMID: 25048716 PMCID: PMC4575407 DOI: 10.1002/hep.27323] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 07/12/2014] [Indexed: 12/12/2022]
Abstract
Hepatitis B virus (HBV) and hepatitis C virus (HCV) cause a large proportion of acute and chronic liver disease worldwide. Over the past decades many immunological studies defined host immune responses that mediate spontaneous clearance of acute HBV and HCV infection. However, host immune responses are also relevant in the context of treatment-induced clearance of chronic HBV and HCV infection. First, the pretreatment level of interferon-stimulated genes as well as genetic determinants of innate immune responses, such as single nucleotide polymorphisms near the IFNL3 gene, are strong predictors of the response to interferon-alpha (IFN-α)-based therapy. Second, IFN-α, which has been a mainstay of HBV and HCV therapy over decades, and ribavirin, which has also been included in interferon-free direct antiviral therapy for HCV, modulate host immune responses. Third, both IFN-α-based and IFN-α-free treatment regimens of HBV and HCV infection alter the short-term and long-term adaptive immune response against these viruses. Finally, treatment studies have not just improved the clinical outcomes, but also provided opportunities to study virus-host interaction. This review summarizes our current knowledge on how a patient's immune response affects the treatment outcome of HBV and HCV infection and how innate and adaptive immune responses themselves are altered by the different treatment regimens.
Collapse
Affiliation(s)
- Barbara Rehermann
- Immunology Section, Liver Diseases Branch, NIDDK, National Institutes of Health, DHHSBethesda, MD, USA
| | - Antonio Bertoletti
- Emerging Infectious Diseases, Duke-NUS Graduate Medical School
- Singapore Institute for Clinical Sciences, A* STARSingapore
| |
Collapse
|
412
|
Akagi T, Hanamura N, Ichiki T. Measurement of Individual Nanobioparticles on Microfluidic Chips by Laser Dark-field Imaging. J PHOTOPOLYM SCI TEC 2015. [DOI: 10.2494/photopolymer.28.727] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Takanori Akagi
- Department of Bioengineering, Graduate School of Engineering,University of Tokyo
| | - Nami Hanamura
- Department of Bioengineering, Graduate School of Engineering,University of Tokyo
| | - Takanori Ichiki
- Department of Bioengineering, Graduate School of Engineering,University of Tokyo
- Innovation Center of NanoMedicine (iCONM)
| |
Collapse
|
413
|
Anderson DJ, Politch JA. Role of Seminal Plasma in Human Female Reproductive Failure: Immunomodulation, Inflammation, and Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 868:159-69. [PMID: 26178849 DOI: 10.1007/978-3-319-18881-2_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Human seminal plasma contains factors that can regulate the female immune system and potentially promote reproductive fitness. Adverse effects on fertility and pregnancy may occur when seminal plasma provides insufficient, excessive, or altered signals or when the female partner is incapable of receiving these signals.
Collapse
Affiliation(s)
- Deborah J Anderson
- Department of Obstetrics and Gynecology, Boston University School of Medicine, 670 Albany Street, Suite 516, Boston, MA, 02118, USA,
| | | |
Collapse
|
414
|
Hantaan virus can infect human keratinocytes and activate an interferon response through the nuclear translocation of IRF-3. INFECTION GENETICS AND EVOLUTION 2015; 29:146-55. [DOI: 10.1016/j.meegid.2014.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 11/09/2014] [Accepted: 11/11/2014] [Indexed: 12/11/2022]
|
415
|
Mueller N, Avota E, Collenburg L, Grassmé H, Schneider-Schaulies S. Neutral sphingomyelinase in physiological and measles virus induced T cell suppression. PLoS Pathog 2014; 10:e1004574. [PMID: 25521388 PMCID: PMC4270778 DOI: 10.1371/journal.ppat.1004574] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 11/12/2014] [Indexed: 11/20/2022] Open
Abstract
T cell paralysis is a main feature of measles virus (MV) induced immunosuppression. MV contact mediated activation of sphingomyelinases was found to contribute to MV interference with T cell actin reorganization. The role of these enzymes in MV-induced inhibition of T cell activation remained equally undefined as their general role in regulating immune synapse (IS) activity which relies on spatiotemporal membrane patterning. Our study for the first time reveals that transient activation of the neutral sphingomyelinase 2 (NSM2) occurs in physiological co-stimulation of primary T cells where ceramide accumulation is confined to the lamellum (where also NSM2 can be detected) and excluded from IS areas of high actin turnover. Genetic ablation of the enzyme is associated with T cell hyper-responsiveness as revealed by actin dynamics, tyrosine phosphorylation, Ca2+-mobilization and expansion indicating that NSM2 acts to suppress overshooting T cell responses. In line with its suppressive activity, exaggerated, prolonged NSM2 activation as occurring in co-stimulated T cells following MV exposure was associated with aberrant compartmentalization of ceramides, loss of spreading responses, interference with accumulation of tyrosine phosphorylated protein species and expansion. Altogether, this study for the first time reveals a role of NSM2 in physiological T cell stimulation which is dampening and can be abused by a virus, which promotes enhanced and prolonged NSM2 activation to cause pathological T cell suppression. Though the ability of measles virus (MV) to impair T cell activation has long been known, it is mechanistically not well understood. We have shown earlier that MV can contact dependently trigger activation of sphingomyelinases which is known to affect compartmentalization of membrane lipids and proteins. Because these are particularly important in the activity of the immune synapse (IS), we investigated whether MV-induced sphingomyelinase activity would interfere at that level with T cell activation. Our study for the first time revealed that the neutral sphingomyelinase 2 (NSM2) is transiently activated in primary T cells by co-stimulation through CD3 and CD28, and that this does occur to dampen early T cell responses. The virus appears to exploit this inhibitory activity of the enzyme to suppress T cell activation by promoting an enhanced and prolonged NSM2 activation. These findings do not only assign a hitherto novel role of the NSM2 in regulating T cell responses, but also reveal a novel strategy for viral T cell suppression.
Collapse
Affiliation(s)
- Nora Mueller
- University of Würzburg, Institute for Virology and Immunobiology, Wuerzburg, Germany
| | - Elita Avota
- University of Würzburg, Institute for Virology and Immunobiology, Wuerzburg, Germany
| | - Lena Collenburg
- University of Würzburg, Institute for Virology and Immunobiology, Wuerzburg, Germany
| | | | | |
Collapse
|
416
|
Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep 2014; 16:24-43. [PMID: 25488940 DOI: 10.15252/embr.201439363] [Citation(s) in RCA: 547] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An effective immune response requires the engagement of host receptors by pathogen-derived molecules and the stimulation of an appropriate cellular response. Therefore, a crucial factor in our ability to control an infection is the accessibility of our immune cells to the foreign material. Exosomes-which are extracellular vesicles that function in intercellular communication-may play a key role in the dissemination of pathogen- as well as host-derived molecules during infection. In this review, we highlight the composition and function of exosomes and other extracellular vesicles produced during viral, parasitic, fungal and bacterial infections and describe how these vesicles could function to either promote or inhibit host immunity.
Collapse
Affiliation(s)
- Jeffrey S Schorey
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| | - Yong Cheng
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| | - Prachi P Singh
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| | - Victoria L Smith
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
417
|
Lin Y, Li D, Liang Q, Liu S, Zuo X, Li L, Sun X, Li W, Guo M, Huang Z. miR-638 regulates differentiation and proliferation in leukemic cells by targeting cyclin-dependent kinase 2. J Biol Chem 2014; 290:1818-28. [PMID: 25451924 DOI: 10.1074/jbc.m114.599191] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs have been extensively studied as regulators of hematopoiesis and leukemogenesis. We identified miR-638 as a novel regulator in myeloid differentiation and proliferation of leukemic cells. We found that miR-638 was developmentally up-regulated in cells of myeloid but not lymphoid lineage. Furthermore, significant miR-638 down-regulation was observed in primary acute myeloid leukemia (AML) blasts, whereas miR-638 expression was dramatically up-regulated in primary AML blasts and leukemic cell lines undergoing forced myeloid differentiation. These observations suggest that miR-638 might play a role in myeloid differentiation, and its dysregulation may contribute to leukemogenesis. Indeed, ectopic expression of miR-638 promoted phorbol 12-myristate 13-acetate- or all-trans-retinoic acid-induced differentiation of leukemic cell lines and primary AML blasts, whereas miR-638 inhibition caused an opposite phenotype. Consistently, miR-638 overexpression induced G1 cell cycle arrest and reduced colony formation in soft agar. Cyclin-dependent kinase 2 (CDK2) was found to be a target gene of miR-638. CDK2 inhibition phenotypically mimicked the overexpression of miR-638. Moreover, forced expression of CDK2 restored the proliferation and the colony-forming ability inhibited by miR-638. Our data suggest that miR-638 regulates proliferation and myeloid differentiation by targeting CDK2 and may serve as a novel target for leukemia therapy or marker for AML diagnosis and prognosis.
Collapse
Affiliation(s)
- Yi Lin
- From the College of Life Sciences, Wuhan University, Wuhan, Hubei, China, 430072
| | - Dengju Li
- the Department of Hematology, Tongji Hospital of Huazhong Technology University, Wuhan, Hubei, China, 430030
| | - Qing Liang
- the Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China, 430071, and
| | - Shangqing Liu
- the Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China, 430071, and
| | - Xuelan Zuo
- the Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China, 430071, and
| | - Lin Li
- the Department of Hematology, Jiangsu Province Hospital of Traditional Chinese Medicine (TCM), Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China, 210029
| | - Xuemei Sun
- the Department of Hematology, Jiangsu Province Hospital of Traditional Chinese Medicine (TCM), Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China, 210029
| | - Wenxin Li
- From the College of Life Sciences, Wuhan University, Wuhan, Hubei, China, 430072
| | - Mingxiong Guo
- From the College of Life Sciences, Wuhan University, Wuhan, Hubei, China, 430072,
| | - Zan Huang
- From the College of Life Sciences, Wuhan University, Wuhan, Hubei, China, 430072,
| |
Collapse
|
418
|
Madison MN, Roller RJ, Okeoma CM. Human semen contains exosomes with potent anti-HIV-1 activity. Retrovirology 2014; 11:102. [PMID: 25407601 PMCID: PMC4245725 DOI: 10.1186/s12977-014-0102-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 10/31/2014] [Indexed: 11/16/2022] Open
Abstract
Background Exosomes are membranous nanovesicles secreted into the extracellular milieu by diverse cell types. Exosomes facilitate intercellular communication, modulate cellular pheno/genotype, and regulate microbial pathogenesis. Although human semen contains exosomes, their role in regulating infection with viruses that are sexually transmitted remains unknown. In this study, we used semen exosomes purified from healthy human donors to evaluate the role of exosomes on the infectivity of different strains of HIV-1 in a variety of cell lines. Results We show that human semen contains a heterologous population of exosomes, enriched in mRNA encoding tetraspanin exosomal markers and various antiviral factors. Semen exosomes are internalized by recipient cells and upon internalization, inhibit replication of a broad array of HIV-1 strains. Remarkably, the anti-HIV-1 activity of semen exosomes is specific to retroviruses because semen exosomes blocked replication of the murine AIDS (mAIDS) virus complex (LP-BM5). However, exosomes from blood had no effect on HIV-1 or LP-BM5 replication. Additionally, semen and blood exosomes had no effect on replication of herpes simplex virus; types 1 and 2 (HSV1 and HSV2). Mechanistic studies indicate that semen exosomes exert a post-entry block on HIV-1 replication by orchestrating deleterious effects on particle-associated reverse transcriptase activity and infectivity. Conclusions These illuminating findings i) improve our knowledge of the cargo of semen exosomes, ii) reveal that semen exosomes possess anti-retroviral activity, and iii) suggest that semen exosome-mediated inhibition of HIV-1 replication may provide novel opportunities for the development of new therapeutics for HIV-1.
Collapse
Affiliation(s)
- Marisa N Madison
- Department of Microbiology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA 52242-1109, USA.
| | - Richard J Roller
- Department of Microbiology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA 52242-1109, USA.
| | - Chioma M Okeoma
- Department of Microbiology, Carver College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA 52242-1109, USA. .,Interdisciplinary Program in Molecular and Cellular Biology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
419
|
Akamatsu S, Hayes CN, Tsuge M, Miki D, Akiyama R, Abe H, Ochi H, Hiraga N, Imamura M, Takahashi S, Aikata H, Kawaoka T, Kawakami Y, Ohishi W, Chayama K. Differences in serum microRNA profiles in hepatitis B and C virus infection. J Infect 2014; 70:273-87. [PMID: 25452043 DOI: 10.1016/j.jinf.2014.10.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 09/18/2014] [Accepted: 10/17/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Patients infected with chronic hepatitis B virus (HBV) or hepatitis C virus (HCV) are at greater risk of cirrhosis and hepatocellular carcinoma. The objective of this study was to identify virus-specific serum microRNA profiles associated with liver function and disease progression. Microarray analysis of serum microRNAs was performed using the Toray 3D array system in 22 healthy subjects, 42 HBV patients, and 30 HCV patients. Selected microRNAs were then validated by qRT-PCR in 186 HBV patients, 107 HCV patients, and 22 healthy subjects. RESULTS Microarray analysis showed up-regulation of a number of microRNAs in serum of both HBV and HCV patients. In qRT-PCR analysis, miR-122, miR-99a, miR-125b, miR-720, miR-22, and miR-1275 were up-regulated both in HBV patients relative to healthy subjects, and all except miR-1275 were up-regulated in HBeAg-positive patients relative to HBeAg-negative patients. Specific microRNAs were independently associated with different aspects of HBV infection. MiR-122 was independently associated with HBV DNA level, whereas miR-125b was independently associated with levels of HBV DNA, HBsAg, and HBeAg. MiR-22 and miR-1275 were independently associated with serum γ-glutamyl transpeptidase levels. CONCLUSIONS Serum microRNA levels reflect differences in the etiology and stage of viral hepatitis.
Collapse
Affiliation(s)
- Sakura Akamatsu
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - C Nelson Hayes
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Masataka Tsuge
- Liver Research Project Center, Hiroshima University, Hiroshima, Japan; Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Daiki Miki
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Rie Akiyama
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Hiromi Abe
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Hidenori Ochi
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Nobuhiko Hiraga
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Michio Imamura
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | | | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Tomokazu Kawaoka
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Yoshiiku Kawakami
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan
| | - Waka Ohishi
- Department of Clinical Studies, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Laboratory for Digestive Diseases, Center for Genomic Medicine, RIKEN, Hiroshima, Japan; Liver Research Project Center, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
420
|
Liu Z, Zhang X, Yu Q, He JJ. Exosome-associated hepatitis C virus in cell cultures and patient plasma. Biochem Biophys Res Commun 2014; 455:218-22. [PMID: 25449270 DOI: 10.1016/j.bbrc.2014.10.146] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 10/29/2014] [Indexed: 01/09/2023]
Abstract
Hepatitis C virus (HCV) infects its target cells in the form of cell-free viruses and through cell-cell contact. Here we report that HCV is associated with exosomes. Using highly purified exosomes and transmission electron microscopic imaging, we demonstrated that HCV occurred in both exosome-free and exosome-associated forms. Exosome-associated HCV was infectious and resistant to neutralization by an anti-HCV neutralizing antibody. There were more exosome-associated HCV than exosome-free HCV detected in the plasma of HCV-infected patients. These results suggest exosome-associated HCV as an alternative form for HCV infection and transmission.
Collapse
Affiliation(s)
- Ziqing Liu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Xiugen Zhang
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, United States
| | - Qigui Yu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Johnny J He
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, United States.
| |
Collapse
|
421
|
Cossetti C, Iraci N, Mercer TR, Leonardi T, Alpi E, Drago D, Alfaro-Cervello C, Saini HK, Davis MP, Schaeffer J, Vega B, Stefanini M, Zhao C, Muller W, Garcia-Verdugo JM, Mathivanan S, Bachi A, Enright AJ, Mattick JS, Pluchino S. Extracellular vesicles from neural stem cells transfer IFN-γ via Ifngr1 to activate Stat1 signaling in target cells. Mol Cell 2014; 56:193-204. [PMID: 25242146 PMCID: PMC4578249 DOI: 10.1016/j.molcel.2014.08.020] [Citation(s) in RCA: 236] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 07/22/2014] [Accepted: 08/15/2014] [Indexed: 12/20/2022]
Abstract
The idea that stem cell therapies work only via cell replacement is challenged by the observation of consistent intercellular molecule exchange between the graft and the host. Here we defined a mechanism of cellular signaling by which neural stem/precursor cells (NPCs) communicate with the microenvironment via extracellular vesicles (EVs), and we elucidated its molecular signature and function. We observed cytokine-regulated pathways that sort proteins and mRNAs into EVs. We described induction of interferon gamma (IFN-γ) pathway in NPCs exposed to proinflammatory cytokines that is mirrored in EVs. We showed that IFN-γ bound to EVs through Ifngr1 activates Stat1 in target cells. Finally, we demonstrated that endogenous Stat1 and Ifngr1 in target cells are indispensable to sustain the activation of Stat1 signaling by EV-associated IFN-γ/Ifngr1 complexes. Our study identifies a mechanism of cellular signaling regulated by EV-associated IFN-γ/Ifngr1 complexes, which grafted stem cells may use to communicate with the host immune system.
Collapse
Affiliation(s)
- Chiara Cossetti
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY Cambridge, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge, UK
| | - Nunzio Iraci
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY Cambridge, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge, UK
| | - Tim R Mercer
- Institute for Molecular Bioscience, University of Queensland, St Lucia QLD 4072, Australia
| | - Tommaso Leonardi
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY Cambridge, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge, UK; The EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Emanuele Alpi
- Biomolecular Mass Spectrometry Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milano, Italy
| | - Denise Drago
- Biomolecular Mass Spectrometry Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milano, Italy
| | - Clara Alfaro-Cervello
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY Cambridge, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge, UK
| | - Harpreet K Saini
- The EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Matthew P Davis
- The EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Julia Schaeffer
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY Cambridge, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge, UK
| | - Beatriz Vega
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY Cambridge, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge, UK
| | - Matilde Stefanini
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY Cambridge, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge, UK
| | - CongJian Zhao
- Southwest Hospital, Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| | - Werner Muller
- Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Jose Manuel Garcia-Verdugo
- Departamento de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, 46980 Valencia, Spain
| | - Suresh Mathivanan
- Department of Biochemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Bundoora, Victoria 3086, Australia
| | - Angela Bachi
- Biomolecular Mass Spectrometry Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milano, Italy
| | - Anton J Enright
- The EMBL-European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | | | - Stefano Pluchino
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, and NIHR Biomedical Research Centre, University of Cambridge, CB2 0PY Cambridge, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, Cambridge, UK.
| |
Collapse
|
422
|
Zhao X, Wu Y, Duan J, Ma Y, Shen Z, Wei L, Cui X, Zhang J, Xie Y, Liu J. Quantitative Proteomic Analysis of Exosome Protein Content Changes Induced by Hepatitis B Virus in Huh-7 Cells Using SILAC Labeling and LC–MS/MS. J Proteome Res 2014; 13:5391-402. [PMID: 25265333 DOI: 10.1021/pr5008703] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Xue Zhao
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
- Microbiology
Laboratory, Shanghai Municipal Center for Disease Control and Prevention, No. 1380 West Zhongshan Road, Shanghai 200336, People’s Republic of China
| | - Yanxin Wu
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Jinlin Duan
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
- Department
of Pathology, Tongren Hospital Affiliated to Shanghai Jiaotong University School of Medicine, 786 YuYuan Road, Shanghai 200336, People’s Republic of China
| | - Yanchun Ma
- Lab
Center, Putuo District Center Hospital, Shanghai University of Traditional Chinese Medicine, 164 Lanxi Road, Shanghai 200062, People’s Republic of China
| | - Zhongliang Shen
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Lili Wei
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Xiaoxian Cui
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Junqi Zhang
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Youhua Xie
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| | - Jing Liu
- Key
Laboratory of Medical Molecular Virology (MOE/MOH) and Institutes
of Biomedical Sciences, Shanghai Medical College, Fudan University, 138
YiXueYuan Road, Shanghai 200032, People’s Republic of China
| |
Collapse
|
423
|
Ding S, Robek MD. Peroxisomal MAVS activates IRF1-mediated IFN-λ production. Nat Immunol 2014; 15:700-1. [PMID: 25045870 DOI: 10.1038/ni.2924] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Siyuan Ding
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael D Robek
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
424
|
György B, Hung ME, Breakefield XO, Leonard JN. Therapeutic applications of extracellular vesicles: clinical promise and open questions. Annu Rev Pharmacol Toxicol 2014; 55:439-464. [PMID: 25292428 DOI: 10.1146/annurev-pharmtox-010814-124630] [Citation(s) in RCA: 404] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review provides an updated perspective on rapidly proliferating efforts to harness extracellular vesicles (EVs) for therapeutic applications. We summarize current knowledge, emerging strategies, and open questions pertaining to clinical potential and translation. Potentially useful EVs comprise diverse products of various cell types and species. EV components may also be combined with liposomes and nanoparticles to facilitate manufacturing as well as product safety and evaluation. Potential therapeutic cargoes include RNA, proteins, and drugs. Strategic issues considered herein include choice of therapeutic agent, means of loading cargoes into EVs, promotion of EV stability, tissue targeting, and functional delivery of cargo to recipient cells. Some applications may harness natural EV properties, such as immune modulation, regeneration promotion, and pathogen suppression. These properties can be enhanced or customized to enable a wide range of therapeutic applications, including vaccination, improvement of pregnancy outcome, and treatment of autoimmune disease, cancer, and tissue injury.
Collapse
Affiliation(s)
- Bence György
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02114.,Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115
| | - Michelle E Hung
- Interdepartmental Biological Sciences Graduate Program, Northwestern University, Evanston, Illinois 60208
| | - Xandra O Breakefield
- Molecular Neurogenetics Unit, Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02114
| | - Joshua N Leonard
- Department of Chemical and Biological Engineering, Robert H. Lurie Comprehensive Cancer Center, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
425
|
Zhu X, He Z, Yuan J, Wen W, Huang X, Hu Y, Lin C, Pan J, Li R, Deng H, Liao S, Zhou R, Wu J, Li J, Li M. IFITM3-containing exosome as a novel mediator for anti-viral response in dengue virus infection. Cell Microbiol 2014; 17:105-18. [PMID: 25131332 PMCID: PMC7162390 DOI: 10.1111/cmi.12339] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 07/29/2014] [Accepted: 07/31/2014] [Indexed: 01/11/2023]
Abstract
Interferon-inducible transmembrane proteins 1, 2 and 3 (IFITM1, IFITM2 and IFITM3) have recently been identified as potent antiviral effectors that function to suppress the entry of a broad range of enveloped viruses and modulate cellular tropism independent of viral receptor expression. However, the antiviral effect and mechanisms of IFITMs in response to viral infections remain incompletely understood and characterized. In this work, we focused our investigation on the function of the extracellular IFITM3 protein. In cell models of DENV-2 infection, we found that IFITM3 contributed to both the baseline and interferon-induced inhibition of DENV entry. Most importantly, our study for the first time demonstrated the presence of IFITM-containing exosome in the extracellular environment, and identified an ability of cellular exosome to intercellularly deliver IFITM3 and thus transmit its antiviral effect from infected to non-infected cells. Thus, our findings provide new insights in the basic mechanisms underlying the actions of IFITM3, which might lead to future development of exosome-mediated anti-viral strategies using IFITM3 as a therapeutic agent. Conceivably, variations in the basal and inducible levels of IFITMs, as well as in intracellular and extracellular levels of IFITMs, might predict the severity of dengue virus infections among individuals or across species.
Collapse
Affiliation(s)
- Xun Zhu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China; Guangdong Province Key Laboratory of Functional Molecules in Oceanic Microorganism (Sun Yat-sen University), Bureau of Education, Guangzhou, China; Department of Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
426
|
Frenz T, Graalmann L, Detje CN, Döring M, Grabski E, Scheu S, Kalinke U. Independent of Plasmacytoid Dendritic Cell (pDC) infection, pDC Triggered by Virus-Infected Cells Mount Enhanced Type I IFN Responses of Different Composition as Opposed to pDC Stimulated with Free Virus. THE JOURNAL OF IMMUNOLOGY 2014; 193:2496-503. [DOI: 10.4049/jimmunol.1400215] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
427
|
Ramakrishnaiah V, van der Laan LJW. Hepatitis virus hijacks shuttle: exosome-like vesicles provide protection against neutralizing antibodies. Hepatology 2014; 59:2416-8. [PMID: 24273053 DOI: 10.1002/hep.26943] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/08/2013] [Accepted: 11/17/2013] [Indexed: 01/21/2023]
|
428
|
Altevogt P, Bretz NP, Ridinger J, Utikal J, Umansky V. Novel insights into exosome-induced, tumor-associated inflammation and immunomodulation. Semin Cancer Biol 2014; 28:51-7. [PMID: 24769223 DOI: 10.1016/j.semcancer.2014.04.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/09/2014] [Indexed: 12/13/2022]
Abstract
The immune system of cancer patients is often suppressed. Accumulating evidence suggests that exosomes released from tumor cells may play an essential role in this process but the mechanisms are not fully understood. Here we review recent papers showing that exosomes trigger the release of cytokines/chemokines from immune cells. We suggest that this process will either result in the stimulation of anti-tumor immune reactions or in a systemic immunosuppression. The direction appears to be largely dependent on the duration of interactions between immune cells and exosomes leading to the accumulation of inflammatory factors, i.e. on the length of the exposure to these factors. We propose that a long-term interaction of the immune system with elevated levels of tumor exosomes contributes to the development of immunosuppression in cancer patients.
Collapse
Affiliation(s)
- Peter Altevogt
- Translational Immunology, D015, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany.
| | - Niko P Bretz
- Translational Immunology, D015, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Johannes Ridinger
- Translational Immunology, D015, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany; Skin Cancer Unit, German Cancer Research Center DKFZ, Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, 69120 Heidelberg, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center DKFZ, Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, 69120 Heidelberg, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center DKFZ, Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
429
|
Villarroya-Beltri C, Baixauli F, Gutiérrez-Vázquez C, Sánchez-Madrid F, Mittelbrunn M. Sorting it out: regulation of exosome loading. Semin Cancer Biol 2014; 28:3-13. [PMID: 24769058 DOI: 10.1016/j.semcancer.2014.04.009] [Citation(s) in RCA: 588] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 04/16/2014] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles (EVs), a term that includes both exosomes of endocytic origin and vesicles derived from plasma membranes, are continuously secreted by cells to the extracellular environment, and represent a novel vehicle for cell-cell communication. Exosomes contain specific repertoires of proteins and RNAs, indicating the existence of mechanisms that control the sorting of molecules into them. Although the molecular mechanisms that regulate the loading of proteins into exosomes have been studied for years, the sorting of RNA has been elusive until recently. Here we review the molecular mechanisms that control the sorting of molecules into exosomes, with special attention to the sorting of RNA. We also discuss how the cellular context affects the composition of exosomes, and thus the outcome of the communication between the exosome-producer and recipient cells, with particular focus on the communication between tumor cells and with cells of the tumor microenvironment.
Collapse
Affiliation(s)
- Carolina Villarroya-Beltri
- Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Servicio de Inmunología, Hospital de la Princesa, Madrid, Spain
| | - Francesc Baixauli
- Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Servicio de Inmunología, Hospital de la Princesa, Madrid, Spain
| | - Cristina Gutiérrez-Vázquez
- Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Servicio de Inmunología, Hospital de la Princesa, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Servicio de Inmunología, Hospital de la Princesa, Madrid, Spain.
| | - María Mittelbrunn
- Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
430
|
Allweiss L, Volz T, Lütgehetmann M, Giersch K, Bornscheuer T, Lohse AW, Petersen J, Ma H, Klumpp K, Fletcher SP, Dandri M. Immune cell responses are not required to induce substantial hepatitis B virus antigen decline during pegylated interferon-alpha administration. J Hepatol 2014; 60:500-7. [PMID: 24398036 DOI: 10.1016/j.jhep.2013.10.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 10/15/2013] [Accepted: 10/21/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Pegylated interferon-alpha (PegIFNα) remains an attractive treatment option for chronic hepatitis B virus (HBV) infection because it induces higher rates of antigen loss and seroconversion than treatment with polymerase inhibitors. Although early HBsAg decline is recognised as the best predictor of sustained response to IFN-based therapy, it is unclear whether immune cell functions are required to induce significant antigenemia reduction in the first weeks of treatment. Aim of the study was to investigate whether PegIFNα can induce sustained human hepatocyte responsiveness and substantial loss of circulating and intrahepatic viral antigen loads in a system lacking immune cell functions. METHODS HBV-infected humanized uPA/SCID mice received either PegIFNα, entecavir (ETV), or both agents in combination. Serological and intrahepatic changes were determined by qRT-PCR and immunohistochemistry and compared to untreated mice. RESULTS After 4 weeks of treatment, median viremia reduction was greater in mice treated with ETV (either with or without PegIFNα) than with PegIFNα. In contrast, levels of circulating HBeAg, HBsAg, and intrahepatic HBcAg were significantly reduced (p = 0.03) only in mice receiving PegIFNα alone or in combination, as compared to mice receiving ETV monotherapy. Progressive antigen reduction was also demonstrated in mice receiving PegIFNα for 12 weeks (HBeAg = Δ1log; HBsAg = Δ1.4log; p < 0.0001). Notably, repeated administrations of the longer-active PegIFNα could breach the impairment of HBV-infected hepatocyte responsiveness and induce sustained enhancement of human interferon stimulated genes (ISG). CONCLUSIONS The antiviral effects of PegIFNα exerted on the human hepatocytes can induce sustained responsiveness and trigger substantial HBV antigen decline without claiming the involvement of immune cell responses.
Collapse
Affiliation(s)
- Lena Allweiss
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Tassilo Volz
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Marc Lütgehetmann
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Germany; Department of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katja Giersch
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Till Bornscheuer
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Ansgar W Lohse
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Germany; German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, Germany
| | - Joerg Petersen
- IFI Institute for Interdisciplinary Medicine at Asklepios Clinic St. Georg, Hamburg, Germany
| | - Han Ma
- Hoffmann-La Roche, Inc., Nutley, NJ, USA
| | | | | | - Maura Dandri
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Germany; German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, Germany.
| |
Collapse
|
431
|
Abstract
Interferon-stimulated gene (ISG) products take on a number of diverse roles. Collectively, they are highly effective at resisting and controlling pathogens. In this review, we begin by introducing interferon (IFN) and the JAK-STAT signaling pathway to highlight features that impact ISG production. Next, we describe ways in which ISGs both enhance innate pathogen-sensing capabilities and negatively regulate signaling through the JAK-STAT pathway. Several ISGs that directly inhibit virus infection are described with an emphasis on those that impact early and late stages of the virus life cycle. Finally, we describe ongoing efforts to identify and characterize antiviral ISGs, and we provide a forward-looking perspective on the ISG landscape.
Collapse
Affiliation(s)
- William M. Schneider
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | | | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| |
Collapse
|
432
|
Interplay between hepatitis B virus and the innate immune responses: implications for new therapeutic strategies. Virol Sin 2014; 29:17-24. [PMID: 24452540 DOI: 10.1007/s12250-014-3412-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/06/2014] [Indexed: 01/03/2023] Open
Abstract
Hepatitis B virus (HBV) infection is still a worldwide health problem; however, the current antiviral therapies for chronic hepatitis B are limited in efficacy. The outcome of HBV infection is thought to be the result of complex interactions between the HBV and the host immune system. While the role of the adaptive immune responses in the resolution of HBV infection has been well characterized, the contribution of innate immune mechanisms remains elusive until recent evidence implicates that HBV appears to activate the innate immune response and this response is important for controlling HBV infection. Here, we review our current understanding of innate immune responses to HBV infection and the multifaceted evasion by the virus and discuss the potential strategies to combat chronic HBV infection via induction and restoration of host innate antiviral responses.
Collapse
|
433
|
Abstract
UNLABELLED Serious permanent neurological or psychiatric dysfunction may result from virus infections in the central nervous system (CNS). Olfactory sensory neurons are in direct contact with the external environment, making them susceptible to infection by viruses that can enter the brain via the olfactory nerve. The rarity of full brain viral infections raises the important question of whether unique immune defense mechanisms protect the brain. Here we show that both RNA (vesicular stomatitis virus [VSV]) and DNA (cytomegalovirus [CMV]) virus inoculations of the nasal mucosa leading to olfactory bulb (OB) infection activate long-distance signaling that upregulates antiviral interferon (IFN)-stimulated gene (ISG) expression in uninfected remote regions of the brain. This signaling mechanism is dependent on IFN-α/β receptors deep within the brain, leading to the activation of a distant antiviral state that prevents infection of the caudal brain. In normal mice, VSV replication is limited to the OB, and these animals typically survive the infection. In contrast, mice lacking the IFN-α/β receptor succumbed to the infection, with VSV spreading throughout the brain. Chemical destruction of the olfactory sensory neurons blocked both virus trafficking into the OB and the IFN response in the caudal brain, indicating a direct signaling within the brain after intranasal infection. Most signaling within the brain occurs across the 20-nm synaptic cleft. The unique long-distance IFN signaling described here occurs across many millimeters within the brain and is critical for survival and normal brain function. IMPORTANCE The olfactory mucosa can serve as a conduit for a number of viruses to enter the brain. Yet infections in the CNS rarely occur. The mechanism responsible for protecting the brain from viruses that successfully invade the OB, the first site of infection subsequent to infection of the nasal mucosa, remains elusive. Here we demonstrate that the protection is mediated by a long-distance interferon signaling, particularly IFN-β released by infected neurons in the OB. Strikingly, in the absence of neurotropic virus infection, ISGs are induced in the posterior regions of the brain, activating an antiviral state and preventing further virus invasion.
Collapse
|
434
|
Abstract
Since the discovery of hepatitis C virus (HCV) by molecular cloning almost a quarter of a century ago, unprecedented at the time because the virus had never been grown in cell culture or detected serologically, there have been impressive strides in many facets of our understanding of the natural history of the disease, the viral life cycle, the pathogenesis, and antiviral therapy. It is apparent that the virus has developed multiple strategies to evade immune surveillance and eradication. This Review covers what we currently understand of the temporal and spatial immunological changes within the human innate and adaptive host immune responses that ultimately determine the outcomes of HCV infection.
Collapse
|
435
|
Jiang S, Liu Y, Wang L, Duan C, Liu M. A meta-analysis and systematic review: adjuvant interferon therapy for patients with viral hepatitis-related hepatocellular carcinoma. World J Surg Oncol 2013; 11:240. [PMID: 24060218 PMCID: PMC3851844 DOI: 10.1186/1477-7819-11-240] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 09/05/2013] [Indexed: 12/15/2022] Open
Abstract
Objective To evaluate the efficacy and safety of adjuvant IFN therapy for viral hepatitis-related hepatocellular carcinoma (HCC) after treatment with surgical resection or transarterial chemoembolization (TACE). Methods Controlled trials of adjuvant treatment with IFN for patients with HCC published between 2000 and 2012 were searched electronically in MEDLINE, PubMed, Cochrane Library, and EMBASE databases. According to the heterogeneity of the studies, two different models - the fixed-effect model and the random-effect model - were applied to analyze the results. Results Ten trials were screened according to inclusion and exclusion standards. Eight randomized, controlled trials and two non-randomized, controlled trials were included. These ten trials with a total of 1,029 subjects were eventually involved in the meta-analysis; 528 HCC patients were treated with adjuvant treatment with IFN and 501 patients with placebo. Compared to the control group, the recurrence rates of HCC in IFN group were significantly lower (odds ratio (OR) = 0.66; 95% confidence interval (CI) = 0.50 to 0.86; P = 0.02), especially after TACE treatment according to subgroup analysis (OR = 0.73; 95% CI = 0.52 to 1.01; P = 0.06 for surgical resection; and OR = 0.54; 95% CI = 0.33 to 0.86, P = 0.01 for TACE). The death rates in the IFN group also significantly decreased according to not only total events analysis (OR = 0.42; 95% CI = 0.32 to 0.56; P < 0.00001) but also subgroup analysis (OR = 0.51; 95% CI = 0.36 to 0.72; P = 0.0002 for surgical resection; and OR = 0.33; 95% CI = 0.21 to 0.50; P < 0.00001 for TACE). Conclusions Adjuvant IFN therapy may significantly reduce the recurrence rates of patients with viral hepatitis-related HCC and improve the survival of patients after surgical resection or TACE. The ideal dose mostly selected is 3 MIU/ml, three times per week, which can make patients tolerate the adverse reactions of IFN better and maintain effective concentrations for a long time.
Collapse
Affiliation(s)
- Shaojie Jiang
- Department of Hepatobiliary Surgery, Shenyang Military Region General Hospital, Shenhe District in Shenyang culture road no, 83, Shenyang 110015, China.
| | | | | | | | | |
Collapse
|
436
|
Chen J, Zhang W, Lin J, Wang F, Wu M, Chen C, Zheng Y, Peng X, Li J, Yuan Z. An efficient antiviral strategy for targeting hepatitis B virus genome using transcription activator-like effector nucleases. Mol Ther 2013; 22:303-311. [PMID: 24025750 DOI: 10.1038/mt.2013.212] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 08/26/2013] [Indexed: 02/07/2023] Open
Abstract
The hepatitis B virus (HBV) is a DNA virus that can cause chronic hepatitis B (CHB) in humans. Current therapies for CHB infection are limited in efficacy and do not target the pre-existing viral genomic DNA, which are present in the nucleus as a covalently closed circular DNA (cccDNA) form. The transcription activator-like (TAL) effector nucleases (TALENs) are newly developed enzymes that can cleave sequence-specific DNA targets. Here, TALENs targeting the conserved regions of the viral genomic DNA among different HBV genotypes were constructed. The expression of TALENs in Huh7 cells transfected with monomeric linear full-length HBV DNA significantly reduced the viral production of HBeAg, HBsAg, HBcAg, and pgRNA, resulted in a decreased cccDNA level and misrepaired cccDNAs without apparent cytotoxic effects. The anti-HBV effect of TALENs was further demonstrated in a hydrodynamic injection-based mouse model. In addition, an enhanced antiviral effect with combinations of TALENs and interferon-α (IFN-α) treatment was observed and expression of TALENs restored HBV suppressed IFN-stimulated response element-directed transcription. Taken together, these data indicate that TALENs can specifically target and successfully inactivate the HBV genome and are potently synergistic with IFN-α, thus providing a potential therapeutic strategy for treating CHB infection.
Collapse
Affiliation(s)
- Jieliang Chen
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China; Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wen Zhang
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China; Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Junyu Lin
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Fan Wang
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China; Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Min Wu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Cuncun Chen
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China; Institutes of Medical Microbiology and Biomedical Sciences, Fudan University, Shanghai, China
| | - Ye Zheng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiuhua Peng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jianhua Li
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China; Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institutes of Medical Microbiology and Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
437
|
|
438
|
Seya T, Oshiumi H, Matsumoto M. [Immunobiological response against RNA virus infection]. Uirusu 2013; 63:135-42. [PMID: 25366048 DOI: 10.2222/jsv.63.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Viruses infect host circumventing the host immune system; a variety of strategies for establishment of viral infection have been found in a virus-specific fashion. Infection with RNA viruses allows host dendritic cells to present antigens and a typical pattern (PAMP) of virus products, including the RNA genomes and replication intermediates such as double-stranded RNA (dsRNA), which induce antiviral effectors: type I interferons (IFN), cytokines, NK cell activation, Th1 polarization, CD8 T cell proliferation, etc. These findings revealed that RNA-sensing innate system closely links to a trigger of cellular immunity. This process unequivocally involves the maturation of antigen-presenting dendritic cell (mDC), and virus products frequently block this step. According to these findings, mDC have to sense non-self RNA to establish antiviral immunity without spoiling their functions via infection, except several exceptional cases. The notion infers that the RNA recognition in cytosol of infected cells (intrinsic sensing) functions as virocidal whereas that in mDC (extrinsic sensing) differentially converges on another antiviral strategy, activation of the immune system. In this review, we focus on the potential role of hepatitis C virus (HCV) RNA in modulating the inflammatory milieu around mDCs and evoking antiviral immunity to drive specific cellular effectors against the virus.
Collapse
Affiliation(s)
- Tsukasa Seya
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University
| | | | | |
Collapse
|