1
|
Troyer Z, Gololobova O, Koppula A, Liao Z, Horns F, Elowitz MB, Tosar JP, Batish M, Witwer KW. Simultaneous Protein and RNA Analysis in Single Extracellular Vesicles, Including Viruses. ACS NANO 2024; 18:26568-26584. [PMID: 39306763 PMCID: PMC11447916 DOI: 10.1021/acsnano.4c03679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
The individual detection of human immunodeficiency virus (HIV) virions and resolution from extracellular vesicles (EVs) during analysis is a difficult challenge. Infectious enveloped virions and nonviral EVs are released simultaneously by HIV-infected host cells, in addition to hybrid viral EVs containing combinations of HIV and host components but lacking replicative ability. Complicating the issue, EVs and enveloped virions are both delimited by a lipid bilayer and share similar size and density. The feature that distinguishes infectious virions from host and hybrid EVs is the HIV genomic RNA (gRNA), which allows the virus to replicate. Single-particle analysis techniques, which provide snapshots of single biological nanoparticles, could resolve infectious virions from EVs. However, current single-particle analysis techniques focus mainly on protein detection, which fail to resolve hybrid EVs from infectious virions. A method to simultaneously detect viral protein and internal gRNA in the same particle would allow resolution of infectious HIV from EVs and noninfectious virions. Here, we introduce SPIRFISH, a high-throughput method for single-particle protein and RNA analysis, combining single particle interferometric reflectance imaging sensor with single-molecule fluorescence in situ hybridization. Using SPIRFISH, we detect HIV-1 envelope protein gp120 and genomic RNA within single infectious virions, allowing resolution against EV background and noninfectious virions. We further show that SPIRFISH can be used to detect specific RNAs within EVs. This may have major utility for EV therapeutics, which are increasingly focused on EV-mediated RNA delivery. SPIRFISH should enable single particle analysis of a broad class of RNA-containing nanoparticles.
Collapse
Affiliation(s)
- Zach Troyer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Olesia Gololobova
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- EV Core Facility "EXCEL", Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Aakash Koppula
- Department of Medical and Molecular Sciences, and Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
| | - Zhaohao Liao
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Felix Horns
- Howard Hughes Medical Institute and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Michael B Elowitz
- Howard Hughes Medical Institute and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, United States
| | - Juan Pablo Tosar
- Functional Genomics Laboratory, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- School of Science, Universidad de la República, Montevideo 11400, Uruguay
| | - Mona Batish
- Department of Medical and Molecular Sciences, and Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- EV Core Facility "EXCEL", Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
2
|
Persaud AT, Khela J, Fernandes C, Chaphekar D, Burnie J, Tang VA, Colpitts CC, Guzzo C. Virion-incorporated CD14 enables HIV-1 to bind LPS and initiate TLR4 signaling in immune cells. J Virol 2024; 98:e0036324. [PMID: 38661384 PMCID: PMC11092368 DOI: 10.1128/jvi.00363-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024] Open
Abstract
HIV-1 has a broad range of nuanced interactions with the immune system, and the incorporation of cellular proteins by nascent virions continues to redefine our understanding of the virus-host relationship. Proteins located at the sites of viral egress can be selectively incorporated into the HIV-1 envelope, imparting new functions and phenotypes onto virions, and impacting viral spread and disease. Using virion capture assays and western blot, we show that HIV-1 can incorporate the myeloid antigen CD14 into its viral envelope. Virion-incorporated CD14 remained biologically active and able to bind its natural ligand, bacterial lipopolysaccharide (LPS), as demonstrated by flow virometry and immunoprecipitation assays. Using a Toll-like receptor 4 (TLR4) reporter cell line, we also demonstrated that virions with bound LPS can trigger TLR4 signaling to activate transcription factors that regulate inflammatory gene expression. Complementary assays with THP-1 monocytes demonstrated enhanced secretion of inflammatory cytokines like tumor necrosis factor alpha (TNF-α) and the C-C chemokine ligand 5 (CCL5), when exposed to LPS-loaded virus. These data highlight a new type of interplay between HIV-1 and the myeloid cell compartment, a previously well-established cellular contributor to HIV-1 pathogenesis and inflammation. Persistent gut inflammation is a hallmark of chronic HIV-1 infection, and contributing to this effect is the translocation of microbes across the gut epithelium. Our data herein provide proof of principle that virion-incorporated CD14 could be a novel mechanism through which HIV-1 can drive chronic inflammation, facilitated by HIV-1 particles binding bacterial LPS and initiating inflammatory signaling in TLR4-expressing cells.IMPORTANCEHIV-1 establishes a lifelong infection accompanied by numerous immunological changes. Inflammation of the gut epithelia, exacerbated by the loss of mucosal T cells and cytokine dysregulation, persists during HIV-1 infection. Feeding back into this loop of inflammation is the translocation of intestinal microbes across the gut epithelia, resulting in the systemic dissemination of bacterial antigens, like lipopolysaccharide (LPS). Our group previously demonstrated that the LPS receptor, CD14, can be readily incorporated by HIV-1 particles, supporting previous clinical observations of viruses derived from patient plasma. We now show that CD14 can be incorporated by several primary HIV-1 isolates and that this virion-incorporated CD14 can remain functional, enabling HIV-1 to bind to LPS. This subsequently allowed CD14+ virions to transfer LPS to monocytic cells, eliciting pro-inflammatory signaling and cytokine secretion. We posit here that virion-incorporated CD14 is a potential contributor to the dysregulated immune responses present in the setting of HIV-1 infection.
Collapse
Affiliation(s)
- Arvin T. Persaud
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jasmin Khela
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Claire Fernandes
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Deepa Chaphekar
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Burnie
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Vera A. Tang
- Flow Cytometry and Virometry Core Facility, Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Che C. Colpitts
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Christina Guzzo
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Tian MY, Hao DX, Liu Y, He J, Zhao ZH, Guo TY, Li X, Zhang Y. Milk exosomes: an oral drug delivery system with great application potential. Food Funct 2023; 14:1320-1337. [PMID: 36722924 DOI: 10.1039/d2fo02013k] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Exosomes are extracellular vesicles with the smallest diameter, usually divided into cellular sources and body fluid sources. Due to their special properties different from cell-derived exosomes, the application of milk exosomes as an oral drug delivery system has increased greatly. This article introduces the physical and chemical properties of exosomes, separation technology, dyeing and labeling technology, targeted modification technology, and the application of milk exosomes in drug loading and disease therapies.
Collapse
Affiliation(s)
- Meng-Yuan Tian
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Dong-Xia Hao
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Yang Liu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Jin He
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Zhuo-Hua Zhao
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Ting-Yu Guo
- The International Department of the High School Affiliated to Shaanxi Normal University, Xi'an, China
| | - Xing Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Yuan Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education; National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China; College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| |
Collapse
|
4
|
Mekkaoui L, Tejerizo JG, Abreu S, Rubat L, Nikoniuk A, Macmorland W, Horlock C, Matsumoto S, Williams S, Smith K, Price J, Srivastava S, Hussain R, Banani MA, Day W, Stevenson E, Madigan M, Chen J, Khinder R, Miah S, Walker S, Ade-Onojobi M, Domining S, Sillibourne J, Sabatino M, Slepushkin V, Farzaneh F, Pule M. Efficient clinical-grade γ-retroviral vector purification by high-speed centrifugation for CAR T cell manufacturing. Mol Ther Methods Clin Dev 2022; 28:116-128. [PMID: 36620071 PMCID: PMC9808014 DOI: 10.1016/j.omtm.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
γ-Retroviral vectors (γ-RV) are powerful tools for gene therapy applications. Current clinical vectors are produced from stable producer cell lines which require minimal further downstream processing, while purification schemes for γ-RV produced by transient transfection have not been thoroughly investigated. We aimed to develop a method to purify transiently produced γ-RV for early clinical studies. Here, we report a simple one-step purification method by high-speed centrifugation for γ-RV produced by transient transfection for clinical application. High-speed centrifugation enabled the concentration of viral titers in the range of 107-108 TU/mL with >80% overall recovery. Analysis of research-grade concentrated vector revealed sufficient reduction in product- and process-related impurities. Furthermore, product characterization of clinical-grade γ-RV by BioReliance demonstrated two-logs lower impurities per transducing unit compared with regulatory authority-approved stable producer cell line vector for clinical application. In terms of CAR T cell manufacturing, clinical-grade γ-RV produced by transient transfection and purified by high-speed centrifugation was similar to γ-RV produced from a clinical-grade stable producer cell line. This method will be of value for studies using γ-RV to bridge vector supply between early- and late-stage clinical trials.
Collapse
Affiliation(s)
- Leila Mekkaoui
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Jose G. Tejerizo
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Sara Abreu
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Lydie Rubat
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | | | | | - Claire Horlock
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Sofia Matsumoto
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Sarah Williams
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Koval Smith
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Juliet Price
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Saket Srivastava
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Rehan Hussain
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | | | - William Day
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Elena Stevenson
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
| | - Meghan Madigan
- Cell and Gene Therapy, Kings (CGT-K), King’s College London, London SE5 9NU, UK
| | - Jie Chen
- Cell and Gene Therapy, Kings (CGT-K), King’s College London, London SE5 9NU, UK
| | - Ravin Khinder
- Cell and Gene Therapy, Kings (CGT-K), King’s College London, London SE5 9NU, UK
| | - Shahed Miah
- Cell and Gene Therapy, Kings (CGT-K), King’s College London, London SE5 9NU, UK
| | - Simon Walker
- Cell and Gene Therapy, Kings (CGT-K), King’s College London, London SE5 9NU, UK
| | - Michael Ade-Onojobi
- Cell and Gene Therapy, Kings (CGT-K), King’s College London, London SE5 9NU, UK
| | - Sabine Domining
- Cell and Gene Therapy, Kings (CGT-K), King’s College London, London SE5 9NU, UK
| | | | | | | | - Farzin Farzaneh
- Cell and Gene Therapy, Kings (CGT-K), King’s College London, London SE5 9NU, UK
| | - Martin Pule
- Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK
- Deparment of Haematology, Cancer Institute, University College London, London WC1E 6BT, UK
- Corresponding author: Martin Pule, Autolus Limited, The MediaWorks, 191 Wood Lane, London W12 7FP, UK.
| |
Collapse
|
5
|
Coroadinha AS. Cancer Gene Therapy: Development and Production of Lentiviral Vectors for Gene Therapy. Methods Mol Biol 2022; 2521:297-315. [PMID: 35733005 DOI: 10.1007/978-1-0716-2441-8_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Lentiviral vectors are among the most used vectors in gene therapy to treat pathologies of different origins, such as cancers, rare monogenic diseases or neurological disorders. This chapter provides an overview on lentiviral vector developments in terms of vector design and manufacture for gene therapy applications. The state of the art of vector production will be summarized face to the recent developments contributing to improve vector safety, efficacy and manufacturing robustness, focusing on human immunodeficiency virus 1 (HIV-1) based lentiviral vectors. Transient and stable production systems will be discussed highlighting recent advances in producer cell line development. Challenges in lentiviral vector development upstream and downstream will be addressed with a particular focus on the improvements undertaken to increase vector yields and production scalability.
Collapse
Affiliation(s)
- Ana S Coroadinha
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
6
|
Bazzan E, Tinè M, Casara A, Biondini D, Semenzato U, Cocconcelli E, Balestro E, Damin M, Radu CM, Turato G, Baraldo S, Simioni P, Spagnolo P, Saetta M, Cosio MG. Critical Review of the Evolution of Extracellular Vesicles' Knowledge: From 1946 to Today. Int J Mol Sci 2021; 22:ijms22126417. [PMID: 34203956 PMCID: PMC8232679 DOI: 10.3390/ijms22126417] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) are a family of particles/vesicles present in blood and body fluids, composed of phospholipid bilayers that carry a variety of molecules that can mediate cell communication, modulating crucial cell processes such as homeostasis, induction/dampening of inflammation, and promotion of repair. Their existence, initially suspected in 1946 and confirmed in 1967, spurred a sharp increase in the number of scientific publications. Paradoxically, the increasing interest for EV content and function progressively reduced the relevance for a precise nomenclature in classifying EVs, therefore leading to a confusing scientific production. The aim of this review was to analyze the evolution of the progress in the knowledge and definition of EVs over the years, with an overview of the methodologies used for the identification of the vesicles, their cell of origin, and the detection of their cargo. The MISEV 2018 guidelines for the proper recognition nomenclature and ways to study EVs are summarized. The review finishes with a “more questions than answers” chapter, in which some of the problems we still face to fully understand the EV function and potential as a diagnostic and therapeutic tool are analyzed.
Collapse
Affiliation(s)
- Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
- Correspondence: ; Tel.: +39-0498213449
| | - Mariaenrica Tinè
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Alvise Casara
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Davide Biondini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Umberto Semenzato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Elisabetta Cocconcelli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Elisabetta Balestro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Marco Damin
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Claudia Maria Radu
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
- Department of Medicine, University of Padova, 35128 Padova, Italy;
| | - Graziella Turato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Simonetta Baraldo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Paolo Simioni
- Department of Medicine, University of Padova, 35128 Padova, Italy;
| | - Paolo Spagnolo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
| | - Manuel G. Cosio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (A.C.); (D.B.); (U.S.); (E.C.); (E.B.); (M.D.); (G.T.); (S.B.); (P.S.); (M.S.); (M.G.C.)
- Meakins-Christie Laboratories, Respiratory Division, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
7
|
Perry C, Rayat ACME. Lentiviral Vector Bioprocessing. Viruses 2021; 13:268. [PMID: 33572347 PMCID: PMC7916122 DOI: 10.3390/v13020268] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Lentiviral vectors (LVs) are potent tools for the delivery of genes of interest into mammalian cells and are now commonly utilised within the growing field of cell and gene therapy for the treatment of monogenic diseases and adoptive therapies such as chimeric antigen T-cell (CAR-T) therapy. This is a comprehensive review of the individual bioprocess operations employed in LV production. We highlight the role of envelope proteins in vector design as well as their impact on the bioprocessing of lentiviral vectors. An overview of the current state of these operations provides opportunities for bioprocess discovery and improvement with emphasis on the considerations for optimal and scalable processing of LV during development and clinical production. Upstream culture for LV generation is described with comparisons on the different transfection methods and various bioreactors for suspension and adherent producer cell cultivation. The purification of LV is examined, evaluating different sequences of downstream process operations for both small- and large-scale production requirements. For scalable operations, a key focus is the development in chromatographic purification in addition to an in-depth examination of the application of tangential flow filtration. A summary of vector quantification and characterisation assays is also presented. Finally, the assessment of the whole bioprocess for LV production is discussed to benefit from the broader understanding of potential interactions of the different process options. This review is aimed to assist in the achievement of high quality, high concentration lentiviral vectors from robust and scalable processes.
Collapse
Affiliation(s)
- Christopher Perry
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Gower St, London WC1E 6BT, UK;
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
| | - Andrea C. M. E. Rayat
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Gower St, London WC1E 6BT, UK;
| |
Collapse
|
8
|
Purification Methods and the Presence of RNA in Virus Particles and Extracellular Vesicles. Viruses 2020; 12:v12090917. [PMID: 32825599 PMCID: PMC7552034 DOI: 10.3390/v12090917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/12/2020] [Accepted: 08/18/2020] [Indexed: 12/17/2022] Open
Abstract
The fields of extracellular vesicles (EV) and virus infections are marred in a debate on whether a particular mRNA or non-coding RNA (i.e., miRNA) is packaged into a virus particle or copurifying EV and similarly, whether a particular mRNA or non-coding RNA is contained in meaningful numbers within an EV. Key in settling this debate, is whether the purification methods are adequate to separate virus particles, EV and contaminant soluble RNA and RNA:protein complexes. Differential centrifugation/ultracentrifugation and precipitating agents like polyethylene glycol are widely utilized for both EV and virus purifications. EV are known to co-sediment with virions and other particulates, such as defective interfering particles and protein aggregates. Here, we discuss how encased RNAs from a heterogeneous mixture of particles can be distinguished by different purification methods. This is particularly important for subsequent interpretation of whether the RNA associated phenotype is contributed solely by virus or EV particles or a mixture of both. We also discuss the discrepancy of miRNA abundance in EV from different input material.
Collapse
|
9
|
Abrahem R, Chiang E, Haquang J, Nham A, Ting YS, Venketaraman V. The Role of Dendritic Cells in TB and HIV Infection. J Clin Med 2020; 9:jcm9082661. [PMID: 32824563 PMCID: PMC7465216 DOI: 10.3390/jcm9082661] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells are the principal antigen-presenting cells (APCs) in the host defense mechanism. An altered dendritic cell response increases the risk of susceptibility of infections, such as Mycobacterium tuberculosis (M. tb), and the survival of the human immunodeficiency virus (HIV). The altered response of dendritic cells leads to decreased activity of T-helper-1 (Th1), Th2, Regulatory T cells (Tregs), and Th17 cells in tuberculosis (TB) infections due to a diminishment of cytokine release from these APCs, while HIV infection leads to DC maturation, allowing DCs to migrate to lymph nodes and the sub-mucosa where they then transfer HIV to CD4 T cells, although there is controversy around this topic. Increases in the levels of the antioxidant glutathione (GSH) plays a critical role in maintaining dendritic cell redox homeostasis, leading to an adequate immune response with sufficient cytokine release and a subsequent robust immune response. Thus, an understanding of the intricate pathways involved in the dendritic cell response are needed to prevent co-infections and co-morbidities in individuals with TB and HIV.
Collapse
Affiliation(s)
- Rachel Abrahem
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (R.A.); (E.C.); (J.H.); (A.N.); (Y.-S.T.)
| | - Emerald Chiang
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (R.A.); (E.C.); (J.H.); (A.N.); (Y.-S.T.)
| | - Joseph Haquang
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (R.A.); (E.C.); (J.H.); (A.N.); (Y.-S.T.)
| | - Amy Nham
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (R.A.); (E.C.); (J.H.); (A.N.); (Y.-S.T.)
| | - Yu-Sam Ting
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (R.A.); (E.C.); (J.H.); (A.N.); (Y.-S.T.)
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (R.A.); (E.C.); (J.H.); (A.N.); (Y.-S.T.)
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA
- Correspondence: ; Tel.: +1-909-706-3736; Fax: +1-909-469-5698
| |
Collapse
|
10
|
Transfiguracion J, Tran MY, Lanthier S, Tremblay S, Coulombe N, Acchione M, Kamen AA. Rapid In-Process Monitoring of Lentiviral Vector Particles by High-Performance Liquid Chromatography. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:803-810. [PMID: 32953931 PMCID: PMC7479275 DOI: 10.1016/j.omtm.2020.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/31/2020] [Indexed: 12/28/2022]
Abstract
Lentiviral vectors (LVs) are a popular gene delivery tool in cell and gene therapy and they are a primary tool for ex vivo transduction of T cells for expression of chimeric antigen receptor (CAR) in CAR-T cell therapies. Extensive process and product characterization are required in manufacturing virus-based gene vectors to better control batch-to-batch variability. However, it has been an ongoing challenge to make quantitative assessments of LV product because current analytical tools often are low throughput and lack robustness and standardization is still required. This paper presents a high-throughput and robust physico-chemical characterization method that directly assesses total LV particles. With simple sample preparation and fast elution time (6.24 min) of the LV peak in 440 mM NaCl (in 20 mM Tris-HCl [pH 7.5]), this ion exchange high-performance liquid chromatography (IEX-HPLC) method is ideal for routine in-process monitoring to facilitate the development of scalable and robust LV manufacturing processes. Furthermore, this HPLC method is suitable for the analysis of all in-process samples, from crude samples such as LV supernatants to final purified products. The linearity range of the standard curve is 3.13 × 108 to 1.0 × 1010 total particles/mL, and both the intra- and inter-assay variabilities are less than 5%.
Collapse
Affiliation(s)
- Julia Transfiguracion
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Michelle Yen Tran
- Viral Vectors and Vaccine Bioprocessing Group, Department of Bioengineering, McGill University, 817 Sherbrooke West #270, Montreal, QC H3A 0C3, Canada
| | - Stéphane Lanthier
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Sonia Tremblay
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Nathalie Coulombe
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Mauro Acchione
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada
| | - Amine A Kamen
- National Research Council, Human Health Therapeutics Research Center, 6100 Royalmount Avenue, Montreal, QC H4P 2R2, Canada.,Viral Vectors and Vaccine Bioprocessing Group, Department of Bioengineering, McGill University, 817 Sherbrooke West #270, Montreal, QC H3A 0C3, Canada
| |
Collapse
|
11
|
Li Z, Li W, Lu M, Bess J, Chao CW, Gorman J, Terry DS, Zhang B, Zhou T, Blanchard SC, Kwong PD, Lifson JD, Mothes W, Liu J. Subnanometer structures of HIV-1 envelope trimers on aldrithiol-2-inactivated virus particles. Nat Struct Mol Biol 2020; 27:726-734. [PMID: 32601441 PMCID: PMC8138683 DOI: 10.1038/s41594-020-0452-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022]
Abstract
The HIV-1 envelope glycoprotein (Env) trimer, composed of gp120 and gp41 subunits, mediates viral entry into cells. Recombinant Env trimers have been studied structurally, but characterization of Env embedded in intact virus membranes has been limited to low resolution. Here, we deploy cryo-electron tomography and subtomogram averaging to determine the structures of Env trimers on aldrithiol-2 (AT-2)-inactivated virions in ligand-free, antibody-bound and CD4-bound forms at subnanometer resolution. Tomographic reconstructions document molecular features consistent with high-resolution structures of engineered soluble and detergent-solubilized Env trimers. One of three conformational states previously predicted by smFRET was not observed by cryo-ET, potentially owing to AT-2 inactivation. We did observe Env trimers to open in situ in response to CD4 binding, with an outward movement of gp120-variable loops and an extension of a critical gp41 helix. Overall features of Env trimer embedded in AT-2-treated virions appear well-represented by current engineered trimers.
Collapse
Affiliation(s)
- Ze Li
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
- Microbial Sciences Institute, Yale University, West Haven, CT, USA
- Fudan University Shanghai Cancer Center, Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenwei Li
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
- Microbial Sciences Institute, Yale University, West Haven, CT, USA
| | - Maolin Lu
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Julian Bess
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Cara W Chao
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S Terry
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Scott C Blanchard
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Walther Mothes
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA.
| | - Jun Liu
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA.
- Microbial Sciences Institute, Yale University, West Haven, CT, USA.
| |
Collapse
|
12
|
Selectively-Packaged Proteins in Breast Cancer Extracellular Vesicles Involved in Metastasis. Int J Mol Sci 2020; 21:ijms21144990. [PMID: 32679759 PMCID: PMC7403963 DOI: 10.3390/ijms21144990] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 01/02/2023] Open
Abstract
Cancer-derived extracellular vesicles are known to play a role in the progression of the disease. In this rapidly-growing field, there are many reports of phenotypic changes in cells following exposure to cancer-derived extracellular vesicles. This study examines the protein contents of vesicles derived from three well-known breast cancer cell lines, MCF-7, MDA-MB-231 and T47D, using peptide-centric LC-MS/MS and cytokine multiplex immunoassay analysis to understand the molecular basis of these changes. Through these techniques a large number of proteins within these vesicles were identified. A large proportion of these proteins are known to be important in cancer formation and progression and associated with cancer signaling, angiogenesis, metastasis and invasion and immune regulation. This highlights the importance of extracellular vesicles (EVs) in cancer communications and shows some of the mechanisms the vesicles use to assist in cancer progression.
Collapse
|
13
|
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: 46] [Impact Index Per Article: 9.2] [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
|
14
|
The duck EB66® cell substrate reveals a novel retrotransposon. Biologicals 2019; 61:22-31. [DOI: 10.1016/j.biologicals.2019.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 11/18/2022] Open
|
15
|
Ressel S, Rosca A, Gordon K, Buck AH. Extracellular RNA in viral-host interactions: Thinking outside the cell. WILEY INTERDISCIPLINARY REVIEWS. RNA 2019; 10:e1535. [PMID: 30963709 PMCID: PMC6617787 DOI: 10.1002/wrna.1535] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 12/15/2022]
Abstract
Small RNAs and their associated RNA interference (RNAi) pathways underpin diverse mechanisms of gene regulation and genome defense across all three kingdoms of life and are integral to virus-host interactions. In plants, fungi and many animals, an ancestral RNAi pathway exists as a host defense mechanism whereby viral double-stranded RNA is processed to small RNAs that enable recognition and degradation of the virus. While this antiviral RNAi pathway is not generally thought to be present in mammals, other RNAi mechanisms can influence infection through both viral- and host-derived small RNAs. Furthermore, a burgeoning body of data suggests that small RNAs in mammals can function in a non-cell autonomous manner to play various roles in cell-to-cell communication and disease through their transport in extracellular vesicles. While vesicular small RNAs have not been proposed as an antiviral defense pathway per se, there is increasing evidence that the export of host- or viral-derived RNAs from infected cells can influence various aspects of the infection process. This review discusses the current knowledge of extracellular RNA functions in viral infection and the technical challenges surrounding this field of research. This article is categorized under: Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs RNA in Disease and Development > RNA in Disease Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.
Collapse
Affiliation(s)
- Sarah Ressel
- Institute of Immunology and Infection Research, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Adelina Rosca
- Department of VirologyCarol Davila University of Medicine and PharmacyBucharestRomania
| | - Katrina Gordon
- Institute of Immunology and Infection Research, School of Biological SciencesUniversity of EdinburghEdinburghUK
| | - Amy H. Buck
- Institute of Immunology and Infection Research, School of Biological SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
16
|
Circulating Noncoding RNAs Have a Promising Future Acting as Novel Biomarkers for Colorectal Cancer. DISEASE MARKERS 2019; 2019:2587109. [PMID: 31275444 PMCID: PMC6589288 DOI: 10.1155/2019/2587109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/23/2019] [Accepted: 05/21/2019] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors worldwide, causing a large number of cancer-related deaths each year. Patients are usually diagnosed at advanced and incurable stages due to the lack of suitable screening methods for early detection. Noncoding RNAs (ncRNAs), including small and long noncoding RNAs (lncRNA), are known to have significant regulatory functions, and accumulating evidence suggests that circulating ncRNAs have potential applications as noninvasive biomarkers for diagnosing CRC, evaluating its prognosis, or predicting chemosensitivity in the general population. In this review, we summarize the origins of circulating ncRNAs and provide details of single and multiple circulating ncRNAs that might have roles as diagnostic and prognostic biomarkers in CRC. We end by discussing circulating ncRNAs that may distinguish patients with resistance to chemotherapy.
Collapse
|
17
|
Activated dendritic cells and monocytes in HIV immunological nonresponders: HIV-induced interferon-inducible protein-10 correlates with low future CD4+ recovery. AIDS 2019; 33:1117-1129. [PMID: 30789356 PMCID: PMC6511429 DOI: 10.1097/qad.0000000000002173] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text Objective: To explore monocyte and dendritic cell immune responses, and their association with future CD4+ gain in treated HIV patients with suboptimal CD4+ recovery. Design: A cross-sectional study of HIV-infected, virally suppressed individuals on antiretroviral therapy for at least 24 months; 41 immunological nonresponders (INRs) (CD4+ cell count <400 cells/μl) and 26 immunological responders (CD4+ cell count >600 cells/μl). Ten HIV-infected antiretroviral therapy-naive and 10 HIV-negative healthy persons served as controls. CD4+ cell counts were registered after median 2.4 and 4.7 years. Methods: Monocyte, dendritic-cell and T-cell activation and regulatory T cells (Tregs) were analyzed by flow cytometry. In INR and immunological responder subgroups matched on age and nadir CD4+ cell count, upregulation of interferon-inducible protein-10 (IP-10) and indoleamine 2,3-dioxygenase in monocytes and dendritic cells and cytokines in cell supernatants were measured in vitro in peripheral blood mononuclear cells stimulated with aldrithiol-2-inactivated HIV-1. Results: The INR group displayed higher spontaneous activation of both monocytes (HLA-DR+) and myeloid and plasmacytoid dendritic cells (HLA-DR+, CD83+ and CD86+) compared with immunological responders, and this was associated with increased T-cell activation (CD38+HLA-DR+), an effector memory T-cell phenotype and activated Tregs. The IP-10 response in monocytes after in-vitro HIV stimulation was negatively associated with prospective CD4+ gain. IP-10, indoleamine 2,3-dioxygenase and cytokines levels were comparable between the groups, but inversely correlated with activated Tregs in INRs. Conclusion: HIV-infected individuals with suboptimal immune recovery demonstrated more activated monocytes and in particular dendritic cells, compared with patients with acceptable CD4+ gain. A high level of HIV-specific IP-10 expression in monocytes may be predictive of future CD4+ recovery.
Collapse
|
18
|
Reiter K, Aguilar PP, Wetter V, Steppert P, Tover A, Jungbauer A. Separation of virus-like particles and extracellular vesicles by flow-through and heparin affinity chromatography. J Chromatogr A 2019; 1588:77-84. [DOI: 10.1016/j.chroma.2018.12.035] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 12/31/2022]
|
19
|
Burnie J, Guzzo C. The Incorporation of Host Proteins into the External HIV-1 Envelope. Viruses 2019; 11:v11010085. [PMID: 30669528 PMCID: PMC6356245 DOI: 10.3390/v11010085] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023] Open
Abstract
The incorporation of biologically active host proteins into HIV-1 is a well-established phenomenon, particularly due to the budding mechanism of viral egress in which viruses acquire their external lipid membrane directly from the host cell. While this mechanism might seemingly imply that host protein incorporation is a passive uptake of all cellular antigens associated with the plasma membrane at the site of budding, this is not the case. Herein, we review the evidence indicating that host protein incorporation can be a selective and conserved process. We discuss how HIV-1 virions displaying host proteins on their surface can exhibit a myriad of altered phenotypes, with notable impacts on infectivity, homing, neutralization, and pathogenesis. This review describes the canonical and emerging methods to detect host protein incorporation, highlights the well-established host proteins that have been identified on HIV-1 virions, and reflects on the role of these incorporated proteins in viral pathogenesis and therapeutic targeting. Despite many advances in HIV treatment and prevention, there remains a global effort to develop increasingly effective anti-HIV therapies. Given the broad range of biologically active host proteins acquired on the surface of HIV-1, additional studies on the mechanisms and impacts of these incorporated host proteins may inform the development of novel treatments and vaccine designs.
Collapse
Affiliation(s)
- Jonathan Burnie
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada.
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| | - Christina Guzzo
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada.
- Department of Biological Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| |
Collapse
|
20
|
Sviben D, Forcic D, Halassy B, Allmaier G, Marchetti-Deschmann M, Brgles M. Mass spectrometry-based investigation of measles and mumps virus proteome. Virol J 2018; 15:160. [PMID: 30326905 PMCID: PMC6192076 DOI: 10.1186/s12985-018-1073-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/02/2018] [Indexed: 02/08/2023] Open
Abstract
Background Measles (MEV) and mumps virus (MUV) are enveloped, non-segmented, negative single stranded RNA viruses of the family Paramyxoviridae, and are the cause of measles and mumps, respectively, both preventable by vaccination. Aside from proteins coded by the viral genome, viruses are considered to contain host cell proteins (HCPs). The presence of extracellular vesicles (ECVs), which are often co-purified with viruses due to their similarity in size, density and composition, also contributes to HCPs detected in virus preparations, and this has often been neglected. The aim was to identify which virus-coded proteins are present in MEV and MUV virions, and to try to detect which HCPs, if any, are incorporated inside the virions or adsorbed on their outer surface, and which are more likely to be a contamination from co-purified ECVs. Methods MUV, MEV and ECVs were purified by ultracentrifugation, hydrophobic interaction chromatography and immunoaffinity chromatography, proteins in the samples were resolved by SDS-PAGE and subjected to identification by MALDI-TOF/TOF-MS. A comparative analysis of HCPs present in all samples was carried out. Results By proteomics approach, it was verified that almost all virus-coded proteins are present in MEV and MUV particles. Protein C in MEV which was until now considered to be non-structural viral protein, was found to be present inside the MeV virions. Results on the presence of HCPs in differently purified virus preparations imply that actin, annexins, cyclophilin A, moesin and integrin β1 are part of the virions. Conclusions All HCPs detected in the viruses are present in ECVs as well, indicating their possible function in vesicle formation, or that most of them are only present in ECVs. Only five HCPs were constantly present in purified virus preparations, regardless of the purification method used, implying they are likely the integral part of the virions. The approach described here is helpful for further investigation of HCPs in other virus preparations. Electronic supplementary material The online version of this article (10.1186/s12985-018-1073-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dora Sviben
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia. .,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia.
| | - Dubravko Forcic
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia.,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| | - Beata Halassy
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia.,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| | - Günter Allmaier
- Institute of Chemical Technologies and Analytics, TU Wien, Getreidemarkt 9, AT-1060, Vienna, Austria
| | | | - Marija Brgles
- Centre for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, HR-10 000, Zagreb, Croatia.,Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Zagreb, Croatia
| |
Collapse
|
21
|
Mallery DL, Márquez CL, McEwan WA, Dickson CF, Jacques DA, Anandapadamanaban M, Bichel K, Towers GJ, Saiardi A, Böcking T, James LC. IP6 is an HIV pocket factor that prevents capsid collapse and promotes DNA synthesis. eLife 2018; 7:e35335. [PMID: 29848441 PMCID: PMC6039178 DOI: 10.7554/elife.35335] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022] Open
Abstract
The HIV capsid is semipermeable and covered in electropositive pores that are essential for viral DNA synthesis and infection. Here, we show that these pores bind the abundant cellular polyanion IP6, transforming viral stability from minutes to hours and allowing newly synthesised DNA to accumulate inside the capsid. An arginine ring within the pore coordinates IP6, which strengthens capsid hexamers by almost 10°C. Single molecule measurements demonstrate that this renders native HIV capsids highly stable and protected from spontaneous collapse. Moreover, encapsidated reverse transcription assays reveal that, once stabilised by IP6, the accumulation of new viral DNA inside the capsid increases >100 fold. Remarkably, isotopic labelling of inositol in virus-producing cells reveals that HIV selectively packages over 300 IP6 molecules per infectious virion. We propose that HIV recruits IP6 to regulate capsid stability and uncoating, analogous to picornavirus pocket factors. HIV-1/IP6/capsid/co-factor/reverse transcription.
Collapse
Affiliation(s)
- Donna L Mallery
- Medical Research Council Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Chantal L Márquez
- EMBL Australia Node, Single Molecule Science, School of Medical SciencesUniversity of New South WalesSydneyAustralia
- ARC Centre of Excellence in Advanced Molecular Imaging, School of Medical SciencesUniversity of New South WalesSydneyAustralia
| | - William A McEwan
- Medical Research Council Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Claire F Dickson
- Medical Research Council Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - David A Jacques
- EMBL Australia Node, Single Molecule Science, School of Medical SciencesUniversity of New South WalesSydneyAustralia
- ARC Centre of Excellence in Advanced Molecular Imaging, School of Medical SciencesUniversity of New South WalesSydneyAustralia
| | | | - Katsiaryna Bichel
- Division of Infection and ImmunityUniversity College LondonLondonUnited Kingdom
| | - Gregory J Towers
- Division of Infection and ImmunityUniversity College LondonLondonUnited Kingdom
| | - Adolfo Saiardi
- Medical Research Council Laboratory for Molecular Cell BiologyUniversity College LondonLondonUnited Kingdom
| | - Till Böcking
- EMBL Australia Node, Single Molecule Science, School of Medical SciencesUniversity of New South WalesSydneyAustralia
- ARC Centre of Excellence in Advanced Molecular Imaging, School of Medical SciencesUniversity of New South WalesSydneyAustralia
| | - Leo C James
- Medical Research Council Laboratory of Molecular BiologyCambridgeUnited Kingdom
| |
Collapse
|
22
|
Barklis E, Staubus AO, Mack A, Harper L, Barklis RL, Alfadhli A. Lipid biosensor interactions with wild type and matrix deletion HIV-1 Gag proteins. Virology 2018; 518:264-271. [PMID: 29549788 DOI: 10.1016/j.virol.2018.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/03/2018] [Accepted: 03/06/2018] [Indexed: 11/19/2022]
Abstract
The matrix (MA) domain of the HIV-1 precursor Gag protein (PrGag) has been shown interact with the HIV-1 envelope (Env) protein, and to direct PrGag proteins to plasma membrane (PM) assembly sites by virtue of its affinity to phosphatidylinositol-4,5-bisphosphate (PI[4,5]P2). Unexpectedly, HIV-1 viruses with large MA deletions (ΔMA) have been shown to be conditionally infectious as long as they are matched with Env truncation mutant proteins or alternative viral glycoproteins. To characterize the interactions of wild type (WT) and ΔMA Gag proteins with PI(4,5)P2 and other acidic phospholipids, we have employed a set of lipid biosensors as probes. Our investigations showed marked differences in WT and ΔMA Gag colocalization with biosensors, effects on biosensor release, and association of biosensors with virus-like particles. These results demonstrate an alternative approach to the analysis of viral protein-lipid associations, and provide new data as to the lipid compositions of HIV-1 assembly sites.
Collapse
Affiliation(s)
- Eric Barklis
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States.
| | - August O Staubus
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| | - Andrew Mack
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| | - Logan Harper
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| | - Robin Lid Barklis
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| | - Ayna Alfadhli
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97035, United States
| |
Collapse
|
23
|
Halvaei S, Daryani S, Eslami-S Z, Samadi T, Jafarbeik-Iravani N, Bakhshayesh TO, Majidzadeh-A K, Esmaeili R. Exosomes in Cancer Liquid Biopsy: A Focus on Breast Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 10:131-141. [PMID: 29499928 PMCID: PMC5862028 DOI: 10.1016/j.omtn.2017.11.014] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 11/04/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023]
Abstract
The important challenge about cancer is diagnosis in primary stages and proper treatment. Although classical clinico-pathological features of the tumor have major prognostic value, the advances in diagnosis and treatment are indebted to discovery of molecular biomarkers and control of cancer in the pre-invasive state. Moreover, the efficiency of available therapeutic options is highly diminished, and chemotherapy is still the main treatment due to lack of enough specific targets. Accordingly, finding the new noninvasive biomarkers for cancer is still an important clinical challenge that is not achieved yet. There are current technologies to screen, diagnose, prognose, and treat cancer, but the limitations of these implements and procedures are undeniable. Liquid biopsy as a noninvasive method has a promising future in the field of cancer, and exosomes as one of the recent areas have drawn much attention. In this review, the potential capability of exosomes is summarized in cancer with the special focus on breast cancer as the second cause of cancer mortality in women all around the world. It discusses reasons to choose exosomes for liquid biopsy and the studies related to different potential biomarkers found in the exosomes. Moreover, exosome studies on milk as a specific biofluid are also discussed. At last, because choosing the method for exosome studies is very challenging, a summary of different techniques is provided.
Collapse
Affiliation(s)
- Sina Halvaei
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Shiva Daryani
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Zahra Eslami-S
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Tannaz Samadi
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Narges Jafarbeik-Iravani
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | | | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Rezvan Esmaeili
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
24
|
Exosomes Mediate Intercellular Transmission of Porcine Reproductive and Respiratory Syndrome Virus. J Virol 2018; 92:JVI.01734-17. [PMID: 29187541 DOI: 10.1128/jvi.01734-17] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 11/22/2017] [Indexed: 12/13/2022] Open
Abstract
Exosomes are small membrane-enclosed vesicles produced by various cells and actively released into the extracellular space. They participate in intercellular communication and transfer of biologically active proteins, lipids, and nucleic acids. Accumulating evidence suggests that exosomes derived from cells infected by some viruses selectively encapsulate viral proteins, genetic materials, or even virions to mediate cell-to-cell communication and/or virus transmission. Porcine reproductive and respiratory syndrome virus (PRRSV) is an Arterivirus that has been devastating the global swine industry since the late 1980s. Recent studies have shown that major proteins secreted from PRRSV-infected cells are exosomal proteins and that the serum-derived exosomes from PRRSV-infected pigs contain viral proteins. However, the role of exosomes in PRRSV infection remains unclear. In this study, purified exosomes isolated from PRRSV-infected cells were shown with reverse transcription-PCR and mass spectrometry to contain viral genomic RNA and partial viral proteins. Furthermore, exosomes from PRRSV-infected cells established productive infection in both PRRSV-susceptible and -nonsusceptible cells. More importantly, exosome-mediated infection was not completely blocked by PRRSV-specific neutralizing antibodies. In summary, this study demonstrated that exosomes can mediate PRRSV transmission and are even resistant to antibody neutralization, identifying a potential immune evasion mechanism utilized by PRRSV.IMPORTANCE Exosomes have recently been characterized as bioactive vesicles that function to promote intercellular communication. The exosomes from virally infected cells containing altered compositions confer numerous novel functionalities. A study of the secretome of cells infected with PRRSV indicated that the exosomal pathway is strongly activated by PRRSV infection. Here, we demonstrate that PRRSV can utilize host exosomes to infect naive healthy cells. Furthermore, exosome-mediated viral transmission is largely resistant to PRRSV-specific neutralizing antibodies. Our study provides novel insights into an alternative mechanism of PRRSV transmission that can compromise the host's anti-PRRSV immune response.
Collapse
|
25
|
Identification of HIV-1-Based Virus-like Particles by Multifrequency Atomic Force Microscopy. Biophys J 2017; 111:1173-1179. [PMID: 27653476 DOI: 10.1016/j.bpj.2016.07.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/05/2016] [Accepted: 07/11/2016] [Indexed: 11/21/2022] Open
Abstract
Virus-like particles (VLPs) have become a promising platform for vaccine production. VLPs are formed by structural viral proteins that inherently self-assemble when expressed in a host cell. They represent a highly immunogenic and safe vaccine platform, due to the absence of the viral genome and its high protein density. One of the most important parameters in vaccine production is the quality of the product. A related bottleneck in VLP-based products is the presence of cellular vesicles as a major contaminant in the preparations, which will require the set up of techniques allowing for specific discrimination of VLPs from host vesicular bodies. In this work novel, to our knowledge, multifrequency (MF) atomic force microscopy (AFM) has permitted full structural nanophysical characterization by its access to the virus capsid of the HIV-based VLPs. The assessment of these particles by advanced amplitude modulation-frequency modulation (AM-FM) viscoelastic mapping mode has enhanced the imaging resolution of their nanomechanical properties, opening a new window for the study of the biophysical attributes of VLPs. Finally, the identification and differentiation of HIV-based VLPs from cellular vesicles has been performed under ambient conditions, providing, to our knowledge, novel methodology for the monitoring and quality control of VLPs.
Collapse
|
26
|
Sviben D, Forcic D, Ivancic-Jelecki J, Halassy B, Brgles M. Recovery of infective virus particles in ion-exchange and hydrophobic interaction monolith chromatography is influenced by particle charge and total-to-infective particle ratio. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1054:10-19. [PMID: 28415019 DOI: 10.1016/j.jchromb.2017.04.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/10/2017] [Accepted: 04/06/2017] [Indexed: 10/19/2022]
Abstract
Viral particles are used in medical applications as vaccines or gene therapy vectors. In order to obtain product of high purity, potency and safety for medical use purification of virus particles is a prerequisite, and chromatography is gaining increased attention to meet this aim. Here, we report on the use of ion-exchange and hydrophobic interaction chromatography on monolithic columns for purification of mumps virus (MuV) and measles virus (MeV). Efficiency of the process was monitored by quantification of infective virus particles (by 50% cell culture infective dose assay) and total virus particles, and monitoring of their size (by Nanoparticle Tracking Analysis). Ion-exchange chromatography was shown to be inefficient for MuV and best results for MeV were obtained on QA column with recovery around 17%. Purification of MuV and MeV by hydrophobic interaction chromatography resulted in recoveries around 60%. Results showed that columns with small channels (d=1.4μm) are not suitable for MuV and MeV, although their size is below 400nm, whereas columns with large channels (6μm) showed to be efficient and recoveries independent on the flow rate up to 10mL/min. Heterogeneity of the virus suspension and its interday variability mostly regarding total-to-infective particle ratio was observed. Interestingly, a trend in recovery depending on the day of the harvest was also observed for both viruses, and it correlated with the total-to-infective particle ratio, indicating influence of the virus sample composition on the chromatography results.
Collapse
Affiliation(s)
- Dora Sviben
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia
| | - Dubravko Forcic
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia
| | - Jelena Ivancic-Jelecki
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia
| | - Beata Halassy
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia
| | - Marija Brgles
- University of Zagreb, Centre for Research and Knowledge Transfer in Biotechnology, Rockefellerova 10, HR-10000 Zagreb, Croatia; Centre of Excellence for Viral Immunology and Vaccines, CERVirVac, Croatia.
| |
Collapse
|
27
|
Hu G, Witwer KW, Bond VC, Haughey N, Kashanchi F, Pulliam L, Buch S. Proceedings of the ISEV symposium on "HIV, NeuroAIDS, drug abuse & EVs". J Extracell Vesicles 2017; 6:1294360. [PMID: 28800366 PMCID: PMC5373676 DOI: 10.1080/20013078.2017.1294360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/06/2017] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) are globular, membrane bound nanovesicles (30-100 nm range) that are shed both during normal cellular functioning and under pathological conditions by most cell types. In recent years, there has been significant interest in the study of these vesicles as conduits for the delivery of information between cells from both analogous and disparate tissues. Their ability to carry specialised cargo including signalling mediators, proteins, messenger RNA and miRNAs characterises these vesicles as primary facilitators of cell-to-cell communication and regulation. EVs have also been demonstrated to play important roles in the field of cancer biology and metastasis. However, significant knowledge gaps exist in the role these vesicles play in the context of HIV infection and drug abuse. To foster discussion in this area a satellite symposium on "HIV, NeuroAIDS, Drug Abuse & EVs", was held in conjunction with the annual meeting of the International Society for Extracellular Vesicles (ISEV) in Bethesda, in April 2015. Experts in HIV and drug abuse fields were invited to share their findings on the role of EVs in HIV-1 infection and drug addiction. Additional discussion included current areas of research in EV biology in HIV infection and drug abuse.
Collapse
Affiliation(s)
- Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vincent C Bond
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Norman Haughey
- Department of Neurology, the Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Psychiatry, the Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Lynn Pulliam
- Departments of Laboratory Medicine and Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
28
|
Mateescu B, Kowal EJK, van Balkom BWM, Bartel S, Bhattacharyya SN, Buzás EI, Buck AH, de Candia P, Chow FWN, Das S, Driedonks TAP, Fernández-Messina L, Haderk F, Hill AF, Jones JC, Van Keuren-Jensen KR, Lai CP, Lässer C, Liegro ID, Lunavat TR, Lorenowicz MJ, Maas SLN, Mäger I, Mittelbrunn M, Momma S, Mukherjee K, Nawaz M, Pegtel DM, Pfaffl MW, Schiffelers RM, Tahara H, Théry C, Tosar JP, Wauben MHM, Witwer KW, Nolte-'t Hoen ENM. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - an ISEV position paper. J Extracell Vesicles 2017; 6:1286095. [PMID: 28326170 PMCID: PMC5345583 DOI: 10.1080/20013078.2017.1286095] [Citation(s) in RCA: 525] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/25/2016] [Indexed: 02/07/2023] Open
Abstract
The release of RNA-containing extracellular vesicles (EV) into the extracellular milieu has been demonstrated in a multitude of different in vitro cell systems and in a variety of body fluids. RNA-containing EV are in the limelight for their capacity to communicate genetically encoded messages to other cells, their suitability as candidate biomarkers for diseases, and their use as therapeutic agents. Although EV-RNA has attracted enormous interest from basic researchers, clinicians, and industry, we currently have limited knowledge on which mechanisms drive and regulate RNA incorporation into EV and on how RNA-encoded messages affect signalling processes in EV-targeted cells. Moreover, EV-RNA research faces various technical challenges, such as standardisation of EV isolation methods, optimisation of methodologies to isolate and characterise minute quantities of RNA found in EV, and development of approaches to demonstrate functional transfer of EV-RNA in vivo. These topics were discussed at the 2015 EV-RNA workshop of the International Society for Extracellular Vesicles. This position paper was written by the participants of the workshop not only to give an overview of the current state of knowledge in the field, but also to clarify that our incomplete knowledge – of the nature of EV(-RNA)s and of how to effectively and reliably study them – currently prohibits the implementation of gold standards in EV-RNA research. In addition, this paper creates awareness of possibilities and limitations of currently used strategies to investigate EV-RNA and calls for caution in interpretation of the obtained data.
Collapse
Affiliation(s)
- Bogdan Mateescu
- Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zürich) , Zurich , Switzerland
| | - Emma J K Kowal
- Department of Biology, Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Bas W M van Balkom
- Department of Nephrology and Hypertension, UMC Utrecht , Utrecht , the Netherlands
| | - Sabine Bartel
- Experimental Asthma Research, Priority Area Asthma & Allergy, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL) , Borstel , Germany
| | - Suvendra N Bhattacharyya
- Department of Science and Technology, CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University , Budapest , Hungary
| | - Amy H Buck
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh , Edinburgh , UK
| | | | - Franklin W N Chow
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and Evolution, School of Biological Sciences, University of Edinburgh , Edinburgh , UK
| | - Saumya Das
- Cardiovascular Research Institute, Massachusetts General Hospital , Boston , MA , USA
| | - Tom A P Driedonks
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University , Utrecht , the Netherlands
| | | | - Franziska Haderk
- Department of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Medicine, Helen Diller Family Comprehensive Cancer Center, UC San Francisco, San Francisco, CA, USA
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University , Bundoora , Australia
| | - Jennifer C Jones
- Molecular Immunogenetics & Vaccine Research Section, Vaccine Branch, CCR, NCI , Bethesda , MD , USA
| | | | - Charles P Lai
- Institute of Biomedical Engineering, National Tsing Hua University , Hsinchu , Taiwan
| | - Cecilia Lässer
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, USA; Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Italia di Liegro
- Department of Experimental Biomedicine and Clinical Neurosciences (BIONEC), University of Palermo , Palermo , Italy
| | - Taral R Lunavat
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, USA; Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magdalena J Lorenowicz
- Center for Molecular Medicine, University Medical Center Utrecht & Regenerative Medicine Center , Utrecht , the Netherlands
| | - Sybren L N Maas
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School , Boston , MA , USA
| | - Imre Mäger
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Institute of Technology, University of Tartu, Tartu, Estonia
| | - Maria Mittelbrunn
- Instituto de Investigación del Hospital 12 de Octubre , Madrid , Spain
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School , Frankfurt am Main , Germany
| | - Kamalika Mukherjee
- Department of Science and Technology, CSIR-Indian Institute of Chemical Biology , Kolkata , India
| | - Muhammed Nawaz
- Department of Pathology and Forensic Medicine, Ribeirão Preto School of Medicine, University of Sao Paulo , Sao Paulo , Brazil
| | - D Michiel Pegtel
- Department of Pathology, Exosomes Research Group, VU University Medical Center , Amsterdam , the Netherlands
| | - Michael W Pfaffl
- Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich (TUM) Weihenstephan , Freising , Germany
| | - Raymond M Schiffelers
- Laboratory Clinical Chemistry & Haematology, University Medical Center Utrecht , Utrecht , the Netherlands
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Institute of Biomedical & Health Sciences, Hiroshima University , Hiroshima , Japan
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERM U932 , Paris , France
| | - Juan Pablo Tosar
- Functional Genomics Unit, Institut Pasteur de Montevideo, Nuclear Research Center, Faculty of Science, Universidad de la República , Montevideo , Uruguay
| | - Marca H M Wauben
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University , Utrecht , the Netherlands
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology and Department of Neurology, The Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Esther N M Nolte-'t Hoen
- Department of Biochemistry & Cell Biology, Faculty of Veterinary Medicine, Utrecht University , Utrecht , the Netherlands
| |
Collapse
|
29
|
Chen C, Luo F, Liu X, Lu L, Xu H, Yang Q, Xue J, Shi L, Li J, Zhang A, Liu Q. NF-kB-regulated exosomal miR-155 promotes the inflammation associated with arsenite carcinogenesis. Cancer Lett 2017; 388:21-33. [DOI: 10.1016/j.canlet.2016.11.027] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/03/2016] [Accepted: 11/24/2016] [Indexed: 01/08/2023]
|
30
|
Bonar MM, Tilton JC. High sensitivity detection and sorting of infectious human immunodeficiency virus (HIV-1) particles by flow virometry. Virology 2017; 505:80-90. [PMID: 28235684 DOI: 10.1016/j.virol.2017.02.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/10/2017] [Accepted: 02/16/2017] [Indexed: 12/21/2022]
Abstract
Detection of viruses by flow cytometry is complicated by their small size. Here, we characterized the ability of a standard (FACSAria II) and a sub-micron flow cytometer (A50 Micro) to resolve HIV-1 viruses. The A50 was superior at resolving small particles but did not reliably distinguish HIV-1, extracellular vesicles, and laser noise by light scatter properties alone. However, single fluorescent HIV-1 particles could readily be detected by both cytometers. Fluorescent particles were sorted and retained infectivity, permitting further exploration of the functional consequences of HIV-1 heterogeneity. Finally, flow cytometry had a limit of detection of 80 viruses/ml, nearly equal to PCR assays. These studies demonstrate the power of flow cytometry to detect and sort viral particles and provide a critical toolkit to validate methods to label wild-type HIV-1; quantitatively assess integrity and aggregation of viruses and virus-based therapeutics; and efficiently screen drugs inhibiting viral assembly and release.
Collapse
Affiliation(s)
- Michał M Bonar
- Center for Proteomics and Bioinformatics, Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland OH 44106, USA
| | - John C Tilton
- Center for Proteomics and Bioinformatics, Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland OH 44106, USA.
| |
Collapse
|
31
|
Nasimuzzaman M, Lynn D, van der Loo JC, Malik P. Purification of baculovirus vectors using heparin affinity chromatography. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16071. [PMID: 27933303 PMCID: PMC5142510 DOI: 10.1038/mtm.2016.71] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/13/2016] [Accepted: 09/19/2016] [Indexed: 12/21/2022]
Abstract
Baculoviruses are commonly used for recombinant protein and vaccine production. Baculoviruses are nonpathogenic to vertebrates, have a large packaging capacity, display broad host and cell type tropism, infect both dividing and nondividing cells, and do not elicit strong immune or allergic responses in vivo. Hence, their use as gene delivery vehicles has become increasingly popular in recent years. Moreover, baculovirus vectors carrying mammalian regulatory elements can efficiently transduce and express transgenes in mammalian cells. Based on the finding that heparan sulfate, which is structurally similar to heparin, is an attachment receptor for baculovirus, we developed a novel scalable baculovirus purification method using heparin-affinity chromatography. Baculovirus supernatants were loaded onto a POROS heparin column, washed to remove unbound materials, and eluted with 1.5 mol/l NaCl, which yielded a recovery of purified baculovirus of 85%. After ultracentrifugation, baculovirus titers increased from 200- to 700-fold with overall yields of 26–29%. We further show that baculovirus particles were infectious, normal in morphology and size, despite high-salt elution and shear forces used during purification and concentration. Our chromatography-based purification method is scalable and, together with ultracentrifugation and/or tangential flow filtration, will be suitable for large-scale manufacturing of baculovirus stocks for protein and vaccine production and in gene therapy applications.
Collapse
Affiliation(s)
- Md Nasimuzzaman
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Danielle Lynn
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center , Cincinnati, Ohio, USA
| | - Johannes Cm van der Loo
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Punam Malik
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
32
|
Sosale NG, Ivanovska II, Tsai RK, Swift J, Hsu JW, Alvey CM, Zoltick PW, Discher DE. "Marker of Self" CD47 on lentiviral vectors decreases macrophage-mediated clearance and increases delivery to SIRPA-expressing lung carcinoma tumors. Mol Ther Methods Clin Dev 2016; 3:16080. [PMID: 28053997 PMCID: PMC5148596 DOI: 10.1038/mtm.2016.80] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 10/05/2016] [Accepted: 10/06/2016] [Indexed: 02/08/2023]
Abstract
Lentiviruses infect many cell types and are now widely used for gene delivery in vitro, but in vivo uptake of these foreign vectors by macrophages is a limitation. Lentivectors are produced here from packaging cells that overexpress "Marker of Self" CD47, which inhibits macrophage uptake of cells when prophagocytic factors are also displayed. Single particle analyses show "hCD47-Lenti" display properly oriented human-CD47 for interactions with the macrophage's inhibitory receptor SIRPA. Macrophages derived from human and NOD/SCID/Il2rg-/- (NSG) mice show a SIRPA-dependent decrease in transduction, i.e., transgene expression, by hCD47-Lenti compared to control Lenti. Consistent with known "Self" signaling pathways, macrophage transduction by control Lenti is decreased by drug inhibition of Myosin-II to the same levels as hCD47-Lenti. In contrast, human lung carcinoma cells express SIRPA and use it to enhance transduction by hCD47-Lenti- as illustrated by more efficient gene deletion using CRISPR/Cas9. Intravenous injection of hCD47-Lenti into NSG mice shows hCD47 prolongs circulation, unless a blocking anti-SIRPA is preinjected. In vivo transduction of spleen and liver macrophages also decreases for hCD47-Lenti while transduction of lung carcinoma xenografts increases. hCD47 could be useful when macrophage uptake is limiting on other viral vectors that are emerging in cancer treatments (e.g., Measles glycoprotein-pseudotyped lentivectors) and also in targeting various SIRPA-expressing tumors such as glioblastomas.
Collapse
Affiliation(s)
- Nisha G Sosale
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Irena I Ivanovska
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Richard K Tsai
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joe Swift
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jake W Hsu
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cory M Alvey
- Pharmacological Sciences Graduate Group, University of Pennsylvania, Pennsylvania, USA
| | - Philip W Zoltick
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Dennis E Discher
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Pharmacological Sciences Graduate Group, University of Pennsylvania, Pennsylvania, USA
| |
Collapse
|
33
|
Kolegraff K, Bostik P, Ansari AA. Characterization and Role of Lentivirus-Associated Host Proteins. Exp Biol Med (Maywood) 2016; 231:252-63. [PMID: 16514170 DOI: 10.1177/153537020623100303] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Enveloped viruses obtain their envelopes during the process of budding from infected cells. During this process, however, these viruses acquire parts of the host cell membranes and host cell-derived proteins as integral parts of their mature envelopes. These host-derived components of viral envelopes may subsequently exhibit various effects on the life cycle of the virus; virus cell interactions, especially host response to virus-incorporated self-proteins; and the pathogenesis of the disease induced by these viruses. Although it was known for some time that various viruses incorporate host cell-derived proteins, the issue of the role of these proteins has received increased attention, specifically in connection with human immunodeficiency virus (HIV) infection and development of acquired immunodeficiency syndrome (AIDS) in humans. The aim of this review is to summarize our current knowledge of the analysis and role of host-derived proteins associated with enveloped viruses, with emphasis on the potential role of these proteins in the pathogenesis of AIDS. Clearly, differences in the clinical outcome of those nonhuman primates infected with simian immunodeficiency virus (SIV) that are disease resistant compared with SIV-infected species that are disease susceptible provide a unique opportunity to determine whether differences in the incorporation of distinct sets of host proteins play a role with distinct clinical outcomes.
Collapse
Affiliation(s)
- Keli Kolegraff
- Department of Pathology and Laboratory Medicine, Emory University, WMB Room 2309, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
34
|
Steppert P, Burgstaller D, Klausberger M, Berger E, Aguilar PP, Schneider TA, Kramberger P, Tover A, Nöbauer K, Razzazi-Fazeli E, Jungbauer A. Purification of HIV-1 gag virus-like particles and separation of other extracellular particles. J Chromatogr A 2016; 1455:93-101. [DOI: 10.1016/j.chroma.2016.05.053] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 05/04/2016] [Accepted: 05/13/2016] [Indexed: 12/31/2022]
|
35
|
Li H, Wang S, Kong R, Ding W, Lee FH, Parker Z, Kim E, Learn GH, Hahn P, Policicchio B, Brocca-Cofano E, Deleage C, Hao X, Chuang GY, Gorman J, Gardner M, Lewis MG, Hatziioannou T, Santra S, Apetrei C, Pandrea I, Alam SM, Liao HX, Shen X, Tomaras GD, Farzan M, Chertova E, Keele BF, Estes JD, Lifson JD, Doms RW, Montefiori DC, Haynes BF, Sodroski JG, Kwong PD, Hahn BH, Shaw GM. Envelope residue 375 substitutions in simian-human immunodeficiency viruses enhance CD4 binding and replication in rhesus macaques. Proc Natl Acad Sci U S A 2016; 113:E3413-22. [PMID: 27247400 PMCID: PMC4914158 DOI: 10.1073/pnas.1606636113] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Most simian-human immunodeficiency viruses (SHIVs) bearing envelope (Env) glycoproteins from primary HIV-1 strains fail to infect rhesus macaques (RMs). We hypothesized that inefficient Env binding to rhesus CD4 (rhCD4) limits virus entry and replication and could be enhanced by substituting naturally occurring simian immunodeficiency virus Env residues at position 375, which resides at a critical location in the CD4-binding pocket and is under strong positive evolutionary pressure across the broad spectrum of primate lentiviruses. SHIVs containing primary or transmitted/founder HIV-1 subtype A, B, C, or D Envs with genotypic variants at residue 375 were constructed and analyzed in vitro and in vivo. Bulky hydrophobic or basic amino acids substituted for serine-375 enhanced Env affinity for rhCD4, virus entry into cells bearing rhCD4, and virus replication in primary rhCD4 T cells without appreciably affecting antigenicity or antibody-mediated neutralization sensitivity. Twenty-four RMs inoculated with subtype A, B, C, or D SHIVs all became productively infected with different Env375 variants-S, M, Y, H, W, or F-that were differentially selected in different Env backbones. Notably, SHIVs replicated persistently at titers comparable to HIV-1 in humans and elicited autologous neutralizing antibody responses typical of HIV-1. Seven animals succumbed to AIDS. These findings identify Env-rhCD4 binding as a critical determinant for productive SHIV infection in RMs and validate a novel and generalizable strategy for constructing SHIVs with Env glycoproteins of interest, including those that in humans elicit broadly neutralizing antibodies or bind particular Ig germ-line B-cell receptors.
Collapse
Affiliation(s)
- Hui Li
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Shuyi Wang
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Rui Kong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Wenge Ding
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Fang-Hua Lee
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Zahra Parker
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Eunlim Kim
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Gerald H Learn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Paul Hahn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Ben Policicchio
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
| | | | - Claire Deleage
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Xingpei Hao
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Gwo-Yu Chuang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Matthew Gardner
- Department of Infectious Disease, Scripps Research Institute, Jupiter, FL 33458
| | | | | | - Sampa Santra
- Center of Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
| | - Ivona Pandrea
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261
| | - S Munir Alam
- Department of Medicine, Duke University, Durham, NC 27710
| | - Hua-Xin Liao
- Department of Medicine, Duke University, Durham, NC 27710
| | - Xiaoying Shen
- Department of Medicine, Duke University, Durham, NC 27710
| | | | - Michael Farzan
- Department of Infectious Disease, Scripps Research Institute, Jupiter, FL 33458
| | - Elena Chertova
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Robert W Doms
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | | | | | - Joseph G Sodroski
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215
| | - Peter D Kwong
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, MD 21702
| | - Beatrice H Hahn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| | - George M Shaw
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
36
|
Pang Y, Song H, Cheng W. Using optical trap to measure the refractive index of a single animal virus in culture fluid with high precision. BIOMEDICAL OPTICS EXPRESS 2016; 7:1672-89. [PMID: 27231613 PMCID: PMC4871073 DOI: 10.1364/boe.7.001672] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/20/2016] [Accepted: 03/29/2016] [Indexed: 05/22/2023]
Abstract
The refractive index (RI) is a fundamental parameter of materials that can be used to distinguish and sort materials of different nature. Although the RI of a virus is required for many optics-based biosensing applications, RIs of animal viruses have never been measured. Here we have developed a technique that can measure the RI of individual viruses in aqueous media with high precision. This technique is based on optical trapping of single virions and works by relating the size and RI of a single virus to the stiffness of an optical trap. We have derived an analytic expression to quantitatively describe the optical trapping of these particles. We have validated this equation using nanoparticles of known RI, and measured the RI of individual human immunodeficiency viruses type-1, which yielded a value of 1.42 at 830 nm with less than 2% coefficient of variation. This value is much lower than the RI typically assumed for viruses, but very close to that of 2.0 M sucrose solution in water. To the best of our knowledge, this is the first report on the experimental measurement of the RI for a single animal virus in aqueous media. This technique does not require prior knowledge on the diameter of the nanoparticles, and can be applied to other viruses or nanoparticles for accurate measurement of RI that is critical for the label-free detection of these particles in various settings.
Collapse
Affiliation(s)
- Yuanjie Pang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, USA
| | - Hanna Song
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, USA
| | - Wei Cheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, USA
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
- Department of Biophysics, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
37
|
Nasimuzzaman M, Lynn D, Ernst R, Beuerlein M, Smith RH, Shrestha A, Cross S, Link K, Lutzko C, Nordling D, Russell DW, Larochelle A, Malik P, Van der Loo JC. Production and purification of high-titer foamy virus vector for the treatment of leukocyte adhesion deficiency. Mol Ther Methods Clin Dev 2016; 3:16004. [PMID: 27722179 PMCID: PMC5052019 DOI: 10.1038/mtm.2016.4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/16/2015] [Accepted: 12/17/2015] [Indexed: 01/22/2023]
Abstract
Compared to other integrating viral vectors, foamy virus (FV) vectors have distinct advantages as a gene transfer tool, including their nonpathogenicity, the ability to carry larger transgene cassettes, and increased stability of virus particles due to DNA genome formation within the virions. Proof of principle of its therapeutic utility was provided with the correction of canine leukocyte adhesion deficiency using autologous CD34+ cells transduced with FV vector carrying the canine CD18 gene, demonstrating its long-term safety and efficacy. However, infectious titers of FV-human(h)CD18 were low and not suitable for manufacturing of clinical-grade product. Herein, we developed a scalable production and purification process that resulted in 60-fold higher FV-hCD18 titers from ~1.7 × 104 to 1.0 × 106 infectious units (IU)/ml. Process development improvements included use of polyethylenimine-based transfection, use of a codon-optimized gag, heparin affinity chromatography, tangential flow filtration, and ultracentrifugation, which reproducibly resulted in 5,000-fold concentrated and purified virus, an overall yield of 19 ± 3%, and final titers of 1-2 × 109 IU/ml. Highly concentrated vector allowed reduction of final dimethyl sulfoxide (DMSO) concentration, thereby avoiding DMSO-induced toxicity to CD34+ cells while maintaining high transduction efficiencies. This process development results in clinically relevant, high titer FV which can be scaled up for clinical grade production.
Collapse
Affiliation(s)
- Md Nasimuzzaman
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
- University of Cincinnati College of
Medicine, Cincinnati, Ohio, USA
| | - Danielle Lynn
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
| | - Rebecca Ernst
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
| | - Michele Beuerlein
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
| | - Richard H. Smith
- Hematology Branch, National Heart, Lung, and
Blood Institute, National Institutes of Health, Bethesda,
Maryland, USA
| | - Archana Shrestha
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
- University of Cincinnati College of
Medicine, Cincinnati, Ohio, USA
| | - Scott Cross
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
| | - Kevin Link
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
| | - Carolyn Lutzko
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
- University of Cincinnati College of
Medicine, Cincinnati, Ohio, USA
- Division of Regenerative Medicine and
Cellular Therapies, Hoxworth Blood Center, University of Cincinnati,
Cincinnati, Ohio, USA
| | - Diana Nordling
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
| | - David W. Russell
- Division of Hematology, University of
Washington, Seattle, Washington, USA
| | - Andre Larochelle
- Hematology Branch, National Heart, Lung, and
Blood Institute, National Institutes of Health, Bethesda,
Maryland, USA
| | - Punam Malik
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
- University of Cincinnati College of
Medicine, Cincinnati, Ohio, USA
| | - Johannes C.M. Van der Loo
- Division of Experimental Hematology and
Cancer Biology, Cincinnati Children’s Hospital Medical Center,
Cincinnati, Ohio, USA
- University of Cincinnati College of
Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
38
|
Abstract
Extracellular vesicles (EVs), spherical bilayered proteolipids, behave as paracrine effectors since they are released from cells to deliver signals to other cells. They control a diverse range of biological processes by transferring proteins, lipids, and nucleic acids between cells and are secreted by a wide spectrum of cell types and are found in various biological fluids. EVs are formed at the plasma membrane or in endosomes and are heterogeneous in size and composition. Increasing understanding of the working mechanisms is promising for therapeutic and diagnostic opportunities. In this review, we will focus on the recent developments in this emerging field with special emphasis on the role of EVs in the bone microenvironment, with a central role for the osteoblasts in the communication with a diversity of cells, including bone metastases.
Collapse
|
39
|
Alexander MR, Sanders RW, Moore JP, Klasse PJ. Short Communication: Virion Aggregation by Neutralizing and Nonneutralizing Antibodies to the HIV-1 Envelope Glycoprotein. AIDS Res Hum Retroviruses 2015; 31:1160-5. [PMID: 26086186 DOI: 10.1089/aid.2015.0050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We used dynamic light scattering to detect aggregation of HIV-1 virions by antibodies (IgG) to the viral envelope glycoprotein (Env). Virions of different strains were inactivated by 2,2'-dithiodipyridine (AT-2), a procedure that abrogates infectivity but preserves the native antigenic structure of Env. Neutralizing antibodies directed to a V3-base- and glycan-dependent epitope on gp120 and to the apex of the Env trimer, as well as nonneutralizing antibodies to the epitope cluster I on the gp41-ectodomain, aggregated virions, but in markedly narrow concentration ranges. In contrast, the neutralizing antibody 2G12, which is specific for a composite glycan-dependent epitope on gp120 and functionally monovalent because of its unusual domain-swap structure, was nonaggregating. These results have potentially complex implications for vaccine development.
Collapse
Affiliation(s)
- Marina R. Alexander
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
| | - Rogier W. Sanders
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - John P. Moore
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
| | - Per Johan Klasse
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York
| |
Collapse
|
40
|
Exosomes and Their Role in the Life Cycle and Pathogenesis of RNA Viruses. Viruses 2015; 7:3204-25. [PMID: 26102580 PMCID: PMC4488737 DOI: 10.3390/v7062770] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/03/2015] [Accepted: 06/05/2015] [Indexed: 12/21/2022] Open
Abstract
Exosomes are membrane-enclosed vesicles actively released into the extracellular space, whose content reflect the physiological/pathological state of the cells they originate from. These vesicles participate in cell-to-cell communication and transfer of biologically active proteins, lipids, and RNAs. Their role in viral infections is just beginning to be appreciated. RNA viruses are an important class of pathogens and affect millions of people worldwide. Recent studies on Human Immunodeficiency Virus (HIV), Hepatitis C Virus (HCV), human T-cell lymphotropic virus (HTLV), and Dengue Virus (DENV) have demonstrated that exosomes released from infected cells harbor and deliver many regulatory factors including viral RNA and proteins, viral and cellular miRNA, and other host functional genetic elements to neighboring cells, helping to establish productive infections and modulating cellular responses. Exosomes can either spread or limit an infection depending on the type of pathogen and target cells, and can be exploited as candidates for development of antiviral or vaccine treatments. This review summarizes recent progress made in understanding the role of exosomes in RNA virus infections with an emphasis on their potential contribution to pathogenesis.
Collapse
|
41
|
Morhayim J, Baroncelli M, van Leeuwen JP. Extracellular vesicles: Specialized bone messengers. Arch Biochem Biophys 2014; 561:38-45. [DOI: 10.1016/j.abb.2014.05.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 04/16/2014] [Accepted: 05/08/2014] [Indexed: 12/22/2022]
|
42
|
Goodson P. Questioning the HIV-AIDS Hypothesis: 30 Years of Dissent. Front Public Health 2014; 2:154. [PMID: 25695040 PMCID: PMC4172096 DOI: 10.3389/fpubh.2014.00154] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/07/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
- Patricia Goodson
- Department of Health & Kinesiology, Texas A&M University, College Station, TX, USA
| |
Collapse
|
43
|
Natural mannosylation of HIV-1 gp120 imposes no immunoregulatory effects in primary human plasmacytoid dendritic cells. Mol Immunol 2014; 59:180-7. [DOI: 10.1016/j.molimm.2014.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 02/09/2014] [Accepted: 02/11/2014] [Indexed: 01/15/2023]
|
44
|
Mercier SK, Donaghy H, Botting RA, Turville SG, Harman AN, Nasr N, Ji H, Kusebauch U, Mendoza L, Shteynberg D, Sandgren K, Simpson RJ, Moritz RL, Cunningham AL. The microvesicle component of HIV-1 inocula modulates dendritic cell infection and maturation and enhances adhesion to and activation of T lymphocytes. PLoS Pathog 2013; 9:e1003700. [PMID: 24204260 PMCID: PMC3798598 DOI: 10.1371/journal.ppat.1003700] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 08/26/2013] [Indexed: 01/05/2023] Open
Abstract
HIV-1 is taken up by immature monocyte derived dendritic cells (iMDDCs) into tetraspanin rich caves from which the virus can either be transferred to T lymphocytes or enter into endosomes resulting in degradation. HIV-1 binding and fusion with the DC membrane results in low level de novo infection that can also be transferred to T lymphocytes at a later stage. We have previously reported that HIV-1 can induce partial maturation of iMDDCs at both stages of trafficking. Here we show that CD45⁺ microvesicles (MV) which contaminate purified HIV-1 inocula due to similar size and density, affect DC maturation, de novo HIV-1 infection and transfer to T lymphocytes. Comparing iMDDCs infected with CD45-depleted HIV-1BaL or matched non-depleted preparations, the presence of CD45⁺ MVs was shown to enhance DC maturation and ICAM-1 (CD54) expression, which is involved in DC∶T lymphocyte interactions, while restricting HIV-1 infection of MDDCs. Furthermore, in the DC culture HIV-1 infected (p24⁺) MDDCs were more mature than bystander cells. Depletion of MVs from the HIV-1 inoculum markedly inhibited DC∶T lymphocyte clustering and the induction of alloproliferation as well as limiting HIV-1 transfer from DCs to T lymphocytes. The effects of MV depletion on these functions were reversed by the re-addition of purified MVs from activated but not non-activated SUPT1.CCR5-CL.30 or primary T cells. Analysis of the protein complement of these MVs and of these HIV-1 inocula before and after MV depletion showed that Heat Shock Proteins (HSPs) and nef were the likely DC maturation candidates. Recombinant HSP90α and β and nef all induced DC maturation and ICAM-1 expression, greater when combined. These results suggest that MVs contaminating HIV-1 released from infected T lymphocytes may be biologically important, especially in enhancing T cell activation, during uptake by DCs in vitro and in vivo, particularly as MVs have been detected in the circulation of HIV-1 infected subjects.
Collapse
Affiliation(s)
- Sarah K. Mercier
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Heather Donaghy
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- * E-mail: (HD); (ALC)
| | - Rachel A. Botting
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Stuart G. Turville
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Andrew N. Harman
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Najla Nasr
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Hong Ji
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Ulrike Kusebauch
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Luis Mendoza
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - David Shteynberg
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Kerrie Sandgren
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Richard J. Simpson
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Robert L. Moritz
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Anthony L. Cunningham
- Centre for Virus Research, Westmead Millennium Institute, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
- * E-mail: (HD); (ALC)
| |
Collapse
|
45
|
Linde ME, Colquhoun DR, Ubaida Mohien C, Kole T, Aquino V, Cotter R, Edwards N, Hildreth JEK, Graham DR. The conserved set of host proteins incorporated into HIV-1 virions suggests a common egress pathway in multiple cell types. J Proteome Res 2013; 12:2045-54. [PMID: 23432411 DOI: 10.1021/pr300918r] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
HIV-1 incorporates a large array of host proteins into virions. Determining the host protein composition in HIV virions has technical difficulties, including copurification of microvesicles. We developed an alternative purification technique using cholesterol that differentially modulates the density of virions and microvesicles (density modification, DM) allowing for high-yield virion purification that is essential for tandem mass spectrometric and quantitative proteomic (iTRAQ) analysis. DM purified virions were analyzed using iTRAQ and validated against Optiprep (60% iodixanol) purified virions. We were able to characterize host protein incorporation in DM-purified HIV particles derived from CD4+ T-cell lines; we compared this data set to a reprocessed data set of monocyte-derived macrophages (MDM) derived HIV-1 using the same bioinformatics pipeline. Seventy-nine clustered proteins were shared between the MDM derived and T-cell derived data set. These clusters included an extensive collection of actin isoforms, HLA proteins, chaperones, and a handful of other proteins, many of which have previously been documented to interact with viral proteins. Other proteins of note were ERM proteins, the dynamin domain containing protein EH4, a phosphodiesterase, and cyclophilin A. As these proteins are incorporated in virions produced in both cell types, we hypothesize that these proteins may have direct interactions with viral proteins or may be important in the viral life cycle. Additionally, identified common set proteins are predicted to interact with >1000 related human proteins. Many of these secondary interacting proteins are reported to be incorporated into virions, including ERM proteins and adhesion molecules. Thus, only a few direct interactions between host and viral proteins may dictate the host protein composition in virions. Ultimately, interaction and expression differences in host proteins between cell types may drive virion phenotypic diversity, despite conserved viral protein-host protein interactions between cell types.
Collapse
Affiliation(s)
- Michael E Linde
- Graduate Program in Immunology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gould SJ, Raposo G. As we wait: coping with an imperfect nomenclature for extracellular vesicles. J Extracell Vesicles 2013; 2:20389. [PMID: 24009890 PMCID: PMC3760635 DOI: 10.3402/jev.v2i0.20389] [Citation(s) in RCA: 635] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Stephen J Gould
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
47
|
Coleman BM, Hanssen E, Lawson VA, Hill AF. Prion‐infected cells regulate the release of exosomes with distinct ultrastructural features. FASEB J 2012; 26:4160-73. [DOI: 10.1096/fj.11-202077] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Bradley M. Coleman
- Department of Biochemistry and Molecular BiologyThe University of MelbourneParkvilleVictoriaAustralia
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
- Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Eric Hanssen
- Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
- Bio21 Electron Microscopy UnitThe University of MelbourneParkvilleVictoriaAustralia
| | - Victoria A. Lawson
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
- The Mental Health Research InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Andrew F. Hill
- Department of Biochemistry and Molecular BiologyThe University of MelbourneParkvilleVictoriaAustralia
- Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
- The Mental Health Research InstituteThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
48
|
Ducloux C, Mougel M, Goldschmidt V, Didierlaurent L, Marquet R, Isel C. A pyrophosphatase activity associated with purified HIV-1 particles. Biochimie 2012; 94:2498-507. [PMID: 22766015 DOI: 10.1016/j.biochi.2012.06.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 06/22/2012] [Indexed: 01/17/2023]
Abstract
Treatment of HIV-1 with nucleoside reverse transcription inhibitors leads to the emergence of resistance mutations in the reverse transcriptase (RT) gene. Resistance to 3'-azido-3'-deoxythymidine (AZT) and to a lesser extent to 2'-3'-didehydro-2'-3'-dideoxythymidine is mediated by phosphorolytic excision of the chain terminator. Wild-type RT excises AZT by pyrophosphorolysis, while thymidine-associated resistance mutations in RT (TAMs) favour ATP as the donor substrate. However, in vitro, resistant RT still uses pyrophosphate more efficiently than ATP. We performed in vitro (-) strong-stop DNA synthesis experiments, with wild-type and AZT-resistant HIV-1 RTs, in the presence of physiologically relevant pyrophosphate and/or ATP concentrations and found that in the presence of pyrophosphate, ATP and AZTTP, TAMs do not enhance in vitro (-) strong-stop DNA synthesis. We hypothesized that utilisation of ATP in vivo is driven by intrinsic low pyrophosphate concentrations within the reverse transcription complex, which could be explained by the packaging of a cellular pyrophosphatase. We showed that over-expressed flagged-pyrophosphatase was associated with HIV-1 viral-like particles. In addition, we demonstrated that when HIV-1 particles were purified in order to avoid cellular microvesicle contamination, a pyrophosphatase activity was specifically associated to them. The presence of a pyrophosphatase activity in close proximity to the reverse transcription complex is most likely advantageous to the virus, even in the absence of any drug pressure.
Collapse
Affiliation(s)
- Céline Ducloux
- Architecture et Réactivité de l'ARN, Université de Strasbourg, CNRS, IBMC, 15 Rue René Descartes, 67084 Strasbourg, France.
| | | | | | | | | | | |
Collapse
|
49
|
Ectopic ATP synthase facilitates transfer of HIV-1 from antigen-presenting cells to CD4(+) target cells. Blood 2012; 120:1246-53. [PMID: 22753871 DOI: 10.1182/blood-2011-12-399063] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Antigen-presenting cells (APCs) act as vehicles that transfer HIV to their target CD4(+) cells through an intercellular junction, termed the virologic synapse. The molecules that are involved in this process remain largely unidentified. In this study, we used photoaffinity labeling and a proteomic approach to identify new proteins that facilitate HIV-1 transfer. We identified ectopic mitochondrial ATP synthase as a factor that mediates HIV-1 transfer between APCs and CD4(+) target cells. Monoclonal antibodies against the β-subunit of ATP synthase inhibited APC-mediated transfer of multiple strains HIV-1 to CD4(+) target cells. Likewise, the specific inhibitors of ATPase, citreoviridin and IF1, completely blocked APC-mediated transfer of HIV-1 at the APC-target cell interaction step. Confocal fluorescent microscopy showed localization of extracellular ATP synthase at junctions between APC and CD4(+) target cells. We conclude that ectopic ATP synthase could be an accessible molecular target for inhibiting HIV-1 proliferation in vivo.
Collapse
|
50
|
van der Pol E, Böing AN, Harrison P, Sturk A, Nieuwland R. Classification, Functions, and Clinical Relevance of Extracellular Vesicles. Pharmacol Rev 2012; 64:676-705. [PMID: 22722893 DOI: 10.1124/pr.112.005983] [Citation(s) in RCA: 1284] [Impact Index Per Article: 107.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Edwin van der Pol
- Department of Clinical Chemistry, Academic Medical Centre of the University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|