1
|
Joseph J, Premeaux TA, Pinto DO, Rao A, Guha S, Panfil AR, Carey AJ, Ndhlovu LC, Bergmann‐Leitner ES, Jain P. Retroviral b-Zip protein (HBZ) contributes to the release of soluble and exosomal immune checkpoint molecules in the context of neuroinflammation. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e102. [PMID: 37547182 PMCID: PMC10399615 DOI: 10.1002/jex2.102] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/13/2023] [Accepted: 07/01/2023] [Indexed: 08/08/2023]
Abstract
HTLV-I-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a chronic, progressive, neuroinflammatory demyelinating condition of the spinal cord. We have previously shown that aberrant expression and activity of immune checkpoint (ICP) molecules such as PD-1 and PD-L1/PD-L2, negatively associates with the cytolytic potential of T cells in individuals with HAM/TSP. Interestingly, ICPs can exist in a soluble cell-free form and can be carried on extracellular vesicles (EVs) and exosomes (small EVs, <300nm) while maintaining their immunomodulatory activity. Therefore, we investigated the role of soluble and exosomal ICPs in HTLV-1 associated neuroinflammation. For the very first time, we demonstrate a unique elevated presence of several stimulatory (CD27, CD28, 4-1BB) and inhibitory (BTLA, CTLA-4, LAG-3, PD-1, PD-L2) ICP receptors in HAM/TSP sera, and in purified exosomes from a HAM/TSP-derived HTLV-1-producing (OSP2) cells. These ICPs were found to be co-localized with the endosomal sorting complex required for transport (ESCRT) pathway proteins and exhibited functional binding with their respective ligands. Viral proteins and cytokines (primarily IFNγ) were found to be present in purified exosomes. IFNγ exposure enhanced the release of ICP molecules while antiretroviral drugs (Azidothymidine and Lopinavir) significantly inhibited this process. HTLV-1 b-Zip protein (HBZ) has been linked to factors that enhance EV release and concurrent knockdown here led to the reduced expression of ESCRT associated genes (eg. Hrs, Vsp4, Alix, Tsg101) as well as abrogated the release of ICP molecules, suggesting HBZ involvement in this process. Moreso, exosomes from OSP2 cells adversely affected CD8 T-cell functions by dimishing levels of cytokines and cytotoxic factors. Collectively, these findings highlight exosome-mediated immunmodulation of T-cell functions with HBZ and ESCRT pathways as an underlying mechanism in the context of HTLV-1-induced neuroinflammation.
Collapse
Affiliation(s)
- Julie Joseph
- Department of Microbiology & ImmunologyDrexel University College of MedicinePhiladelphiaPAUSA
| | - Thomas A. Premeaux
- Weill Cornel Medicine Department of MedicineDivision of Infectious DiseasesNew YorkNYUSA
| | - Daniel O. Pinto
- Immunology Core, Biologics Research and DevelopmentWalter Reed Army Institute of ResearchSilver SpringsMDUSA
- Oak Ridge Institute for Science and EducationOak RidgeTNUSA
| | - Abhishek Rao
- Department of Microbiology & ImmunologyDrexel University College of MedicinePhiladelphiaPAUSA
| | - Shrobona Guha
- Department of Neurobiology and AnatomyDrexel University College of MedicinePhiladelphiaPAUSA
| | - Amanda R. Panfil
- The Ohio State University, College of Veterinary Medicine, Center for Retrovirus ResearchColumbusOhioUSA
| | - Alison J. Carey
- Department of Microbiology & ImmunologyDrexel University College of MedicinePhiladelphiaPAUSA
- Department of PediatricsDrexel University College of MedicinePhiladelphiaPAUSA
| | - Lishomwa C. Ndhlovu
- Weill Cornel Medicine Department of MedicineDivision of Infectious DiseasesNew YorkNYUSA
| | - Elke S. Bergmann‐Leitner
- Immunology Core, Biologics Research and DevelopmentWalter Reed Army Institute of ResearchSilver SpringsMDUSA
| | - Pooja Jain
- Department of Microbiology & ImmunologyDrexel University College of MedicinePhiladelphiaPAUSA
- Department of Neurobiology and AnatomyDrexel University College of MedicinePhiladelphiaPAUSA
| |
Collapse
|
2
|
Das U, Banik S, Nadumane SS, Chakrabarti S, Gopal D, Kabekkodu SP, Srisungsitthisunti P, Mazumder N, Biswas R. Isolation, Detection and Analysis of Circulating Tumour Cells: A Nanotechnological Bioscope. Pharmaceutics 2023; 15:280. [PMID: 36678908 PMCID: PMC9864919 DOI: 10.3390/pharmaceutics15010280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/17/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Cancer is one of the dreaded diseases to which a sizeable proportion of the population succumbs every year. Despite the tremendous growth of the health sector, spanning diagnostics to treatment, early diagnosis is still in its infancy. In this regard, circulating tumour cells (CTCs) have of late grabbed the attention of researchers in the detection of metastasis and there has been a huge surge in the surrounding research activities. Acting as a biomarker, CTCs prove beneficial in a variety of aspects. Nanomaterial-based strategies have been devised to have a tremendous impact on the early and rapid examination of tumor cells. This review provides a panoramic overview of the different nanotechnological methodologies employed along with the pharmaceutical purview of cancer. Initiating from fundamentals, the recent nanotechnological developments toward the detection, isolation, and analysis of CTCs are comprehensively delineated. The review also includes state-of-the-art implementations of nanotechnological advances in the enumeration of CTCs, along with future challenges and recommendations thereof.
Collapse
Affiliation(s)
- Upama Das
- Applied Optics and Photonics Laboratory, Department of Physics, Tezpur University, Tezpur 784028, Assam, India
| | - Soumyabrata Banik
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sharmila Sajankila Nadumane
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shweta Chakrabarti
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Dharshini Gopal
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Pornsak Srisungsitthisunti
- Department of Production and Robotics Engineering, Faculty of Engineering, King Mongkut’s University of Technology North Bangkok, Bangkok 10800, Thailand
| | - Nirmal Mazumder
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Rajib Biswas
- Applied Optics and Photonics Laboratory, Department of Physics, Tezpur University, Tezpur 784028, Assam, India
| |
Collapse
|
3
|
Mulherkar TH, Gómez DJ, Sandel G, Jain P. Co-Infection and Cancer: Host–Pathogen Interaction between Dendritic Cells and HIV-1, HTLV-1, and Other Oncogenic Viruses. Viruses 2022; 14:v14092037. [PMID: 36146843 PMCID: PMC9503663 DOI: 10.3390/v14092037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Dendritic cells (DCs) function as a link between innate and adaptive immune responses. Retroviruses HIV-1 and HTLV-1 modulate DCs to their advantage and utilize them to propagate infection. Coinfection of HTLV-1 and HIV-1 has implications for cancer malignancies. Both viruses initially infect DCs and propagate the infection to CD4+ T cells through cell-to-cell transmission using mechanisms including the formation of virologic synapses, viral biofilms, and conduits. These retroviruses are both neurotrophic with neurovirulence determinants. The neuropathogenesis of HIV-1 and HTLV-1 results in neurodegenerative diseases such as HIV-associated neurocognitive disorders (HAND) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Infected DCs are known to traffic to the brain (CNS) and periphery (PNS, lymphatics) to induce neurodegeneration in HAND and HAM/TSP patients. Elevated levels of neuroinflammation have been correlated with cognitive decline and impairment of motor control performance. Current vaccinations and therapeutics for HIV-1 and HTLV-1 are assessed and can be applied to patients with HIV-1-associated cancers and adult T cell leukemia/lymphoma (ATL). These diseases caused by co-infections can result in both neurodegeneration and cancer. There are associations with cancer malignancies and HIV-1 and HTLV-1 as well as other human oncogenic viruses (EBV, HBV, HCV, HDV, and HPV). This review contains current knowledge on DC sensing of HIV-1 and HTLV-1 including DC-SIGN, Tat, Tax, and current viral therapies. An overview of DC interaction with oncogenic viruses including EBV, Hepatitis viruses, and HPV is also provided. Vaccines and therapeutics targeting host–pathogen interactions can provide a solution to co-infections, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Tania H. Mulherkar
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Daniel Joseph Gómez
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
- Department of Biological Sciences, California State University, 25800 Carlos Bee Blvd, Hayward, CA 94542, USA
| | - Grace Sandel
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Pooja Jain
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
- Correspondence:
| |
Collapse
|
4
|
Recent Development of Nanomaterials-Based Cytosensors for the Detection of Circulating Tumor Cells. BIOSENSORS-BASEL 2021; 11:bios11080281. [PMID: 34436082 PMCID: PMC8391755 DOI: 10.3390/bios11080281] [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: 07/20/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022]
Abstract
The accurate analysis of circulating tumor cells (CTCs) holds great promise in early diagnosis and prognosis of cancers. However, the extremely low abundance of CTCs in peripheral blood samples limits the practical utility of the traditional methods for CTCs detection. Thus, novel and powerful strategies have been proposed for sensitive detection of CTCs. In particular, nanomaterials with exceptional physical and chemical properties have been used to fabricate cytosensors for amplifying the signal and enhancing the sensitivity. In this review, we summarize the recent development of nanomaterials-based optical and electrochemical analytical techniques for CTCs detection, including fluorescence, colorimetry, surface-enhanced Raman scattering, chemiluminescence, electrochemistry, electrochemiluminescence, photoelectrochemistry and so on.
Collapse
|
5
|
Li Q, Yin W, Li W, Zhang Z, Zhang X, Zhang XE, Cui Z. Encapsulating Quantum Dots within HIV-1 Virions through Site-Specific Decoration of the Matrix Protein Enables Single Virus Tracking in Live Primary Macrophages. NANO LETTERS 2018; 18:7457-7468. [PMID: 30398057 DOI: 10.1021/acs.nanolett.8b02800] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Labeling and imaging with quantum dots (QDs) provides powerful tools to visualize viral infection in living cells. Encapsulating QDs within virions represents a novel strategy for virus labeling. Here, we developed infectious HIV-1 virions encapsulating QDs through site-specific decoration of the viral matrix protein (MA) and used them to visualize early infection events in human primary macrophages by single-particle imaging. The MA protein was fused to a biotin acceptor peptide (BAP) tag, biotinylated, complexed with streptavidin-conjugated QDs in live cells, and incorporated into virions during virus assembly. The QD-encapsulated virions were tracked during infection of macrophages at a single particle level. The dynamic dissociation of MA and Vpr was also tracked in real time, and the results demonstrated that MA has multiple dynamic behaviors and functions during virus entry. More importantly, we tracked the dynamic interplay of QD-encapsulated virions with cellular mitochondria in live primary macrophages. We also found that HIV-1 can induce fission of mitochondria during the early phases of infection. In summary, we have constructed a type of QD-encapsulated virus particle and used this technology to further our understanding of the early events of HIV-1 infection.
Collapse
Affiliation(s)
- Qin Li
- State Key Laboratory of Virology, Wuhan Institute of Virology , Chinese Academy of Sciences , Wuhan , People's Republic of China
- Engineering Research Center of Industrial Microbiology, Ministry of Education, College of Life Sciences , Fujian Normal University , Fujian 350007 , People's Republic of China
| | - Wen Yin
- State Key Laboratory of Virology, Wuhan Institute of Virology , Chinese Academy of Sciences , Wuhan , People's Republic of China
| | - Wei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology , Chinese Academy of Sciences , Wuhan , People's Republic of China
| | - Zhiping Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology , Chinese Academy of Sciences , Wuhan , People's Republic of China
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology , Chinese Academy of Sciences , Wuhan , People's Republic of China
| | - Xian-En Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology , Chinese Academy of Sciences , Wuhan , People's Republic of China
| |
Collapse
|
6
|
Norouzi M, Zarei Ghobadi M, Golmimi M, Mozhgani SH, Ghourchian H, Rezaee SA. Quantum Dot-Based Biosensor for the Detection of Human T-Lymphotropic Virus-1. ANAL LETT 2017. [DOI: 10.1080/00032719.2017.1287714] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Mehdi Norouzi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Golmimi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Sayed-Hamidreza Mozhgani
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Seyed Abdolrahim Rezaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Herod MR, Pineda RG, Mautner V, Onion D. Quantum dot labelling of adenovirus allows highly sensitive single cell flow and imaging cytometry. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:797-803. [PMID: 25285963 DOI: 10.1002/smll.201401885] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 08/29/2014] [Indexed: 06/03/2023]
Abstract
A quantum dot method for highly efficient labelling of single adenoviral particles is developed. The technique has no impact on viral fitness and allows the imaging and tracking of virus binding and internalisation events using a variety of techniques including imaging cytometry and confocal microscopy. The method is applied to characterise the tropism of different adenoviral vectors.
Collapse
Affiliation(s)
- Morgan R Herod
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | | | | | | |
Collapse
|
8
|
Kim G, Moon JH, Moh CY, Lim JG. A microfluidic nano-biosensor for the detection of pathogenic Salmonella. Biosens Bioelectron 2014; 67:243-7. [PMID: 25172028 DOI: 10.1016/j.bios.2014.08.023] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/28/2014] [Accepted: 08/15/2014] [Indexed: 12/01/2022]
Abstract
Rapid detection of pathogenic Salmonella in food products is extremely important for protecting the public from salmonellosis. The objective of the present study was to explore the feasibility of using a microfluidic nano-biosensor to rapidly detect pathogenic Salmonella. Quantum dot nanoparticles were used to detect Salmonella cells. For selective detection of Salmonella, anti-Salmonella polyclonal antibodies were covalently immobilized onto the quantum dot surface. To separate and concentrate the cells from the sample, superparamagnetic particles and a microfluidic chip were used. A portable fluorometer was developed to measure the fluorescence signal from the quantum dot nanoparticles attached to Salmonella in the samples. The sensitivity for detection of pathogenic Salmonella was evaluated using serially diluted Salmonella Typhimurium in borate buffer and chicken extract. The fluorescence response of the nano-biosensor increased with increasing cell concentration. The detection limit of the sensor was 10(3) CFU/mL Salmonella in both borate buffer and food extract.
Collapse
Affiliation(s)
- Giyoung Kim
- National Academy of Agricultural Science, Department of Agricultural Engineering, RDA, 150 Suin-Ro, Kweonseonku, Suwon 441-100, Republic of Korea.
| | - Ji-Hea Moon
- National Academy of Agricultural Science, Department of Agricultural Engineering, RDA, 150 Suin-Ro, Kweonseonku, Suwon 441-100, Republic of Korea
| | - Chang-Yeon Moh
- National Academy of Agricultural Science, Department of Agricultural Engineering, RDA, 150 Suin-Ro, Kweonseonku, Suwon 441-100, Republic of Korea
| | - Jong-guk Lim
- National Academy of Agricultural Science, Department of Agricultural Engineering, RDA, 150 Suin-Ro, Kweonseonku, Suwon 441-100, Republic of Korea
| |
Collapse
|
9
|
|
10
|
Pustylnikov S, Sagar D, Jain P, Khan ZK. Targeting the C-type lectins-mediated host-pathogen interactions with dextran. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2014; 17:371-92. [PMID: 25224349 PMCID: PMC5553543 DOI: 10.18433/j3n590] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Dextran, the α-1,6-linked glucose polymer widely used in biology and medicine, promises new applications. Linear dextran applied as a blood plasma substitute demonstrates a high rate of biocompatibility. Dextran is present in foods, drugs, and vaccines and in most cases is applied as a biologically inert substance. In this review we analyze dextran's cellular uptake principles, receptor specificity and, therefore, its ability to interfere with pathogen-lectin interactions: a promising basis for new antimicrobial strategies. Dextran-binding receptors in humans include the DC-SIGN (dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin) family receptors: DC-SIGN (CD209) and L-SIGN (the liver and lymphatic endothelium homologue of DC-SIGN), the mannose receptor (CD206), and langerin. These receptors take part in the uptake of pathogens by dendritic cells and macrophages and may also participate in the modulation of immune responses, mostly shown to be beneficial for pathogens per se rather than host(s). It is logical to predict that owing to receptor-specific interactions, dextran or its derivatives can interfere with these immune responses and improve infection outcome. Recent data support this hypothesis. We consider dextran a promising molecule for the development of lectin-glycan interaction-blocking molecules (such as DC-SIGN inhibitors) that could be applied in the treatment of diseases including tuberculosis, influenza, hepatitis B and C, human immunodeficiency virus infection and AIDS, etc. Dextran derivatives indeed change the pathology of infections dependent on DC-SIGN and mannose receptors. Complete knowledge of specific dextran-lectin interactions may also be important for development of future dextran applications in biological research and medicine.
Collapse
Affiliation(s)
- Sergey Pustylnikov
- Group of Molecular Biology Research, Novosibirsk Tuberculosis Research Institute, Novosibirsk, Russia. Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | |
Collapse
|
11
|
Fluorosomes: fluorescent virus-like nanoparticles that represent a convenient tool to visualize receptor-ligand interactions. SENSORS 2013; 13:8722-49. [PMID: 23881135 PMCID: PMC3758619 DOI: 10.3390/s130708722] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 06/28/2013] [Accepted: 07/05/2013] [Indexed: 01/03/2023]
Abstract
Viruses are the smallest life forms and parasitize on many eukaryotic organisms, including humans. Consequently, the study of viruses and viral diseases has had an enormous impact on diverse fields of biology and medicine. Due to their often pathogenic properties, viruses have not only had a strong impact on the development of immune cells but also on shaping entire immune mechanisms in their hosts. In order to better characterize virus-specific surface receptors, pathways of virus entry and the mechanisms of virus assembly, diverse methods to visualize virus particles themselves have been developed in the past decades. Apart from characterization of virus-specific mechanisms, fluorescent virus particles also serve as valuable platforms to study receptor-ligand interactions. Along those lines the authors have developed non-infectious virus-like nanoparticles (VNP), which can be decorated with immune receptors of choice and used for probing receptor-ligand interactions, an especially interesting application in the field of basic but also applied immunology research. To be able to better trace receptor-decorated VNP the authors have developed technology to introduce fluorescent proteins into such particles and henceforth termed them fluorosomes (FS). Since VNP are assembled in a simple expression system relying on HEK-293 cells, gene-products of interest can be assembled in a simple and straightforward fashion—one of the reasons why the authors like to call fluorosomes ‘the poor-man's staining tool’. Within this review article an overview on virus particle assembly, chemical and recombinant methods of virus particle labeling and examples on how FS can be applied as sensors to monitor receptor-ligand interactions on leukocytes are given.
Collapse
|
12
|
Huang BH, Lin Y, Zhang ZL, Zhuan F, Liu AA, Xie M, Tian ZQ, Zhang Z, Wang H, Pang DW. Surface labeling of enveloped viruses assisted by host cells. ACS Chem Biol 2012; 7:683-8. [PMID: 22248430 DOI: 10.1021/cb2001878] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Labeling of virus opens new pathways for the understanding of viruses themselves and facilitates the utilization of viruses in modern biology, medicine, and materials. Based on the characteristic that viruses hijack their host cellular machineries to survive and reproduce themselves, a host-cell-assisted strategy is proposed to label enveloped viruses. By simply feeding Vero cells with commercial 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-(cap biotinyl) (sodium salt) (Biotin-Cap-PE), we obtained biotinylated Vero cells whose membrane systems were modified with biotin. Subsequently, pseudorabies viruses (PrV) were cultivated in the biotinylated Vero cells, and the PrV progenies were spontaneously labeled with Biotin-Cap-PE during viral natural assembly process. Since the viral natural assembly process was employed for the labeling, potential threats of genetic engineering and difficulties in keeping viral natural bioactivity were avoided. Importantly, this labeling strategy for enveloped virus greatly reduces the technical complexity and allows researchers from different backgrounds to apply it for their specified demands.
Collapse
Affiliation(s)
- Bi-Hai Huang
- Key Laboratory of Analytical
Chemistry for Biology and
Medicine (Ministry of Education), College of Chemistry and Molecular
Sciences, Research Center for Nanobiology and Nanomedicine (MOE 985
Innovative Platform), State Key Laboratory of Virology, and Wuhan
Institute of Biotechnology, Wuhan University, Wuhan 430072, P. R. China
| | - Yi Lin
- Key Laboratory of Analytical
Chemistry for Biology and
Medicine (Ministry of Education), College of Chemistry and Molecular
Sciences, Research Center for Nanobiology and Nanomedicine (MOE 985
Innovative Platform), State Key Laboratory of Virology, and Wuhan
Institute of Biotechnology, Wuhan University, Wuhan 430072, P. R. China
| | - Zhi-Ling Zhang
- Key Laboratory of Analytical
Chemistry for Biology and
Medicine (Ministry of Education), College of Chemistry and Molecular
Sciences, Research Center for Nanobiology and Nanomedicine (MOE 985
Innovative Platform), State Key Laboratory of Virology, and Wuhan
Institute of Biotechnology, Wuhan University, Wuhan 430072, P. R. China
| | - Fangfang Zhuan
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan No. 44, Wuhan 430071, P. R. China
| | - An-An Liu
- Key Laboratory of Analytical
Chemistry for Biology and
Medicine (Ministry of Education), College of Chemistry and Molecular
Sciences, Research Center for Nanobiology and Nanomedicine (MOE 985
Innovative Platform), State Key Laboratory of Virology, and Wuhan
Institute of Biotechnology, Wuhan University, Wuhan 430072, P. R. China
| | - Min Xie
- Key Laboratory of Analytical
Chemistry for Biology and
Medicine (Ministry of Education), College of Chemistry and Molecular
Sciences, Research Center for Nanobiology and Nanomedicine (MOE 985
Innovative Platform), State Key Laboratory of Virology, and Wuhan
Institute of Biotechnology, Wuhan University, Wuhan 430072, P. R. China
| | - Zhi-Quan Tian
- Key Laboratory of Analytical
Chemistry for Biology and
Medicine (Ministry of Education), College of Chemistry and Molecular
Sciences, Research Center for Nanobiology and Nanomedicine (MOE 985
Innovative Platform), State Key Laboratory of Virology, and Wuhan
Institute of Biotechnology, Wuhan University, Wuhan 430072, P. R. China
| | - Zhenfeng Zhang
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan No. 44, Wuhan 430071, P. R. China
| | - Hanzhong Wang
- State Key Laboratory of Virology,
Wuhan Institute of Virology, Chinese Academy of Sciences, Xiaohongshan No. 44, Wuhan 430071, P. R. China
| | - Dai-Wen Pang
- Key Laboratory of Analytical
Chemistry for Biology and
Medicine (Ministry of Education), College of Chemistry and Molecular
Sciences, Research Center for Nanobiology and Nanomedicine (MOE 985
Innovative Platform), State Key Laboratory of Virology, and Wuhan
Institute of Biotechnology, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
13
|
Tran TH, El Baz R, Cuconati A, Arthos J, Jain P, Khan ZK. A Novel High-Throughput Screening Assay to Identify Inhibitors of HIV-1 gp120 Protein Interaction with DC-SIGN. JOURNAL OF ANTIVIRALS & ANTIRETROVIRALS 2011; 3:49-54. [PMID: 22102941 PMCID: PMC3217269 DOI: 10.4172/jaa.1000035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The 2010 UNAIDS report states that approximately 34 million people are living with human immunodeficiency virus type 1 (HIV-1), despite highly active antiretroviral therapy (HAART). Despite being effective, ARV therapy has many disadvantages including a cost trajectory unsustainable for economically challenged countries, serious side effects, and the development of drug-resistant strains. Several measures are under way to develop alternatives for ARV therapy, particularly for the control of early HIV-1 infection, but lack of efficient drug targets and assays hinders the search of potential ARV molecules. The dendritic cells present in the mucosal tissue, together with CD4(+) T lymphocytes and macrophages, are among the first cells to encounter HIV-1. The dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) molecule plays a crucial role in binding HIV-1 through high affinity interaction with viral envelope glycoprotein gp120. DC-SIGN, a mannose-binding C-type lectin expressed on cells in the mucosal tissue of the rectum, uterus and cervix, facilitates early HIV-1 infection after sexual transmission. In this study we report a novel target-specific high-throughput screening (HTS) assay capable of quantifying the binding as well as the inhibition of DC-SIGN and gp120. The specificity of the assay was determined through competitive inhibition while optimization occurred for DMSO tolerance (0.5%), Z' factor (0.51), signal-to-noise ratio (3.26), and coefficient of variation (5.1%). For assay validation previously recognized antagonists of DC-SIGN/gp120 binding were tested to detect inhibition demonstrating the suitability of the assay for future HTS screen of potential inhibitors that block the binding between DC-SIGN and gp120 which may prevent early HIV-1 infection.
Collapse
Affiliation(s)
- Thuong H. Tran
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Rasha El Baz
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Andrea Cuconati
- Institute for Hepatitis and Virus Research, The Hepatitis B Foundation, Pennsylvania Biotechnology Center, Doylestown, PA 18902, USA
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergies and Infectious Disease, Rockville Pike, Bethesda MD 20892, USA
| | - Pooja Jain
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Zafar K. Khan
- Drexel Institute for Biotechnology and Virology Research, and Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| |
Collapse
|
14
|
Expression of an RSV-gag virus-like particle in insect cell lines and silkworm larvae. J Virol Methods 2011; 177:147-52. [PMID: 21816175 DOI: 10.1016/j.jviromet.2011.07.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 07/17/2011] [Accepted: 07/20/2011] [Indexed: 10/17/2022]
Abstract
Rous sarcoma virus group antigen protein-based virus-like particles (VLPs) are well known for their structural integrity and ease of handling. VLPs play an important role in drug delivery systems because they can be manipulated with ease. In this study, a new method was established for expressing Rous sarcoma virus group antigen protein based VLPs in silkworm larvae and establishing stably expressing insect cell lines. These VLPs have been isolated by ultracentrifugation using a sucrose step gradient of 10-60% (v/v), and their spherical structure has been confirmed using transmission electron microscopy (TEM). The spherical morphology is similar in both the silkworm larvae and in stably expressing cell lines. Silkworm larvae are better suited for producing Rous sarcoma virus group antigen protein-based VLPs on a large scale; yields from silkworm larvae were approximately 8.2-fold higher than yields from stable cell lines. These VLPs provide a new method for large-scale application in vaccine development and drug delivery systems.
Collapse
|
15
|
Clathrin-mediated endocytosis in living host cells visualized through quantum dot labeling of infectious hematopoietic necrosis virus. J Virol 2011; 85:6252-62. [PMID: 21525360 DOI: 10.1128/jvi.00109-11] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infectious hematopoietic necrosis virus (IHNV) is an important fish pathogen that infects both wild and cultured salmonids. As a species of the genus Novirhabdovirus, IHNV is a valuable model system for exploring the host entry mechanisms of rhabdoviruses. In this study, quantum dots (QDs) were used as fluorescent labels for sensitive, long-term tracking of IHNV entry. Using live-cell fluorescence microscopy, we found that IHNV is internalized through clathrin-coated pits after the virus binds to host cell membranes. Pretreatment of host cells with chlorpromazine, a drug that blocks clathrin-mediated endocytosis, and clathrin light chain (LCa) depletion using RNA interference both resulted in a marked reduction in viral entry. We also visualized transport of the virus via the cytoskeleton (i.e., actin filaments and microtubules) in real time. Actin polymerization is involved in the transport of endocytic vesicles into the cytosol, whereas microtubules are required for the trafficking of clathrin-coated vesicles to early endosomes, late endosomes, and lysosomes. Disrupting the host cell cytoskeleton with cytochalasin D or nocodazole significantly impaired IHNV infectivity. Furthermore, infection was significantly affected by pretreating the host cells with bafilomycin A1, a compound that inhibits the acidification of endosomes and lysosomes. Strong colocalizations of IHNV with endosomes indicated that the virus is internalized into these membrane-bound compartments. This is the first report in which QD labeling is used to visualize the dynamic interactions between viruses and endocytic structures; the results presented demonstrate that IHNV enters host cells via clathrin-mediated endocytic, cytoskeleton-dependent, and low-pH-dependent pathways.
Collapse
|
16
|
Coelho-Dos-Reis JGA, Gomes OA, Bortolini DE, Martins ML, Almeida MR, Martins CS, Carvalho LD, Souza JG, Vilela JMC, Andrade MS, Barbosa-Stancioli EF. Evaluation of the effects of Quercetin and Kaempherol on the surface of MT-2 cells visualized by atomic force microscopy. J Virol Methods 2011; 174:47-52. [PMID: 21507333 DOI: 10.1016/j.jviromet.2011.03.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 03/10/2011] [Accepted: 03/15/2011] [Indexed: 02/08/2023]
Abstract
This study investigated the anti-viral effects of the polyphenolic compounds Quercetin and Kaempherol on the release of HTLV-1 from the surface of MT-2 cells. Atomic force microscopy (AFM) was used to scan the surface of the MT-2 cells. MT-2 cells were fixed with 100% methanol on round glass lamina or cleaved mica and dried under UV light and laminar flow. The images were captured on a Multimode equipment monitored by a NanoScope IIId controller from Veeco Instruments Inc operated in tapping mode and equipped with phase-imaging hardware. The images demonstrated viral budding structures 131 ± 57 nm in size, indicating profuse viral budding. Interestingly, cell-free viruses and budding structures visualized on the surface of cells were less common when MT-2 was incubated with Quercetin, and no particles were seen on the surface of cells incubated with Kaempherol. In summary, these data indicate that HTLV-1 is budding constantly from the MT-2 cell surface and that polyphenolic compounds were able to reduce this viral release. Biological samples were analyzed with crude cell preparations just after cultivation in the presence of Quercetin and Kaempherol, showing that the AFM technique is a rapid and powerful tool for analysis of antiviral activity of new biological compounds.
Collapse
|
17
|
Rahman S, Khan ZK, Wigdahl B, Jennings SR, Tangy F, Jain P. Murine FLT3 ligand-derived dendritic cell-mediated early immune responses are critical to controlling cell-free human T cell leukemia virus type 1 infection. THE JOURNAL OF IMMUNOLOGY 2010; 186:390-402. [PMID: 21115731 DOI: 10.4049/jimmunol.1002570] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human T cell leukemia virus type 1 (HTLV-1) is associated with two immunologically distinct diseases: HTLV-1-associated myelopathy/tropical spastic paraparesis and adult T cell leukemia. We observed previously that depletion of dendritic cells (DCs) in CD11c-diphtheria toxin receptor transgenic mice followed by infection with cell-free virus led to greater proviral and Tax mRNA loads and diminished cellular immune response compared with mice infected with cell-associated virus. To understand the significance of these in vivo results and explore the host-pathogen interaction between DCs and cell-free HTLV-1, we used FLT3 ligand-cultured mouse bone marrow-derived DCs (FL-DCs) and chimeric HTLV-1. Phenotypically, the FL-DCs upregulated expression of surface markers (CD80, CD86, and MHC class II) on infection; however, the level of MHC class I remained unchanged. We performed kinetic studies to understand viral entry, proviral integration, and expression of the viral protein Tax. Multiplex cytokine profiling revealed production of an array of proinflammatory cytokines and type 1 IFN (IFN-α) by FL-DCs treated with virus. Virus-matured FL-DCs stimulated proliferation of autologous CD3(+) T cells as shown by intracellular nuclear Ki67 staining and produced IFN-γ when cultured with infected FL-DCs. Gene expression studies using type 1 IFN-specific and DC-specific arrays revealed upregulation of IFN-stimulated genes, most cytokines, and transcription factors, but a distinct downregulation of many chemokines. Overall, these results highlight the critical early responses generated by FL-DCs on challenge with cell-free chimeric HTLV-1.
Collapse
Affiliation(s)
- Saifur Rahman
- Drexel Institute for Biotechnology and Virology Research, Doylestown, PA 18902, USA
| | | | | | | | | | | |
Collapse
|
18
|
Clinical potential of quantum dots. J Biomed Biotechnol 2010; 2007:76087. [PMID: 18317518 PMCID: PMC2254660 DOI: 10.1155/2007/76087] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Revised: 07/27/2007] [Accepted: 12/13/2007] [Indexed: 11/27/2022] Open
Abstract
Advances in nanotechnology have led to the development of novel fluorescent probes called quantum dots. Quantum dots have revolutionalized the processes of tagging molecules within research settings and are improving sentinel lymph node mapping and identification in vivo studies. As the unique physical and chemical properties of these fluorescent probes are being unraveled, new potential methods of early cancer detection, rapid spread and therapeutic management, that is, photodynamic therapy are being explored. Encouraging results of optical and real time identification of sentinel lymph nodes and lymph flow using quantum dots in vivo models are emerging. Quantum dots have also superseded many of the limitations of organic fluorophores and are a promising alternative as a research tool. In this review, we examine the promising clinical potential of quantum dots, their hindrances for clinical use and the current progress in abrogating their inherent toxicity.
Collapse
|
19
|
Rahman S, Manuel SL, Khan ZK, Wigdahl B, Acheampong E, Tangy F, Jain P. Depletion of dendritic cells enhances susceptibility to cell-free infection of human T cell leukemia virus type 1 in CD11c-diphtheria toxin receptor transgenic mice. THE JOURNAL OF IMMUNOLOGY 2010; 184:5553-61. [PMID: 20382884 DOI: 10.4049/jimmunol.0903226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Human T cell leukemia virus type 1 (HTLV-1) is associated with two immunologically distinct diseases: HTLV-1-associated myelopathy/tropical spastic paraparesis and adult T cell leukemia. The genesis of these diseases is believed to be associated with the route (mucosa versus blood) and mode (cell-free versus cell-associated) of primary infection as well as the modulation of dendritic cell (DC) functions. To explore the role of DCs during early HTLV-1 infection in vivo, we used a chimeric HTLV-1 with a replaced envelope gene from Moloney murine leukemia virus to allow HTLV-1 to fuse with murine cells, which are generally not susceptible to infection with human retroviruses. We also used a CD11c-diphtheria toxin receptor transgenic mouse model system that permits conditional transient depletion of CD11c(+) DCs. We infected these transgenic mice with HTLV-1 using both cell-free and cell-associated infection routes in the absence and presence of DCs. The ablation of DCs led to an enhanced susceptibility to infection with cell-free but not cell-associated HTLV-1 in both CD4 and non-CD4 fractions, as measured by the proviral load. Infection with cell-free virus in the absence of DCs was also found to have increased levels of Tax mRNA in the non-CD4 fraction. Moreover, depletion of DCs significantly dampened the cellular immune response (IFN-gamma(+)CD8(+) T cells) against both cell-free and cell-associated virus. These results uniquely differentiate the involvement of DCs in early cell-free versus late cell-associated infection of HTLV-1 and highlight a significant aspect of viral immunopathogenesis related to the progression of adult T cell leukemia and HTLV-1-associated myelopathy/tropical spastic paraparesis after the initial infection.
Collapse
Affiliation(s)
- Saifur Rahman
- Drexel Institute for Biotechnology and Virology Research, Drexel University College of Medicine, Philadelphia, PA 19127, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Kaittanis C, Santra S, Perez JM. Emerging nanotechnology-based strategies for the identification of microbial pathogenesis. Adv Drug Deliv Rev 2010; 62:408-23. [PMID: 19914316 DOI: 10.1016/j.addr.2009.11.013] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 09/14/2009] [Indexed: 01/04/2023]
Abstract
Infectious diseases are still a major healthcare problem. From food intoxication and contaminated water, to hospital-acquired diseases and pandemics, infectious agents cause disease throughout the world. Despite advancements in pathogens' identification, some of the gold-standard diagnostic methods have limitations, including laborious sample preparation, bulky instrumentation and slow data readout. In addition, new field-deployable diagnostic modalities are urgently needed in first responder and point-of-care applications. Apart from compact, these sensors must be sensitive, specific, robust and fast, in order to facilitate detection of the pathogen even in remote rural areas. Considering these characteristics, researchers have utilized innovative approaches by employing the unique properties of nanomaterials in order to achieve detection of infectious agents, even in complex media like blood. From gold nanoparticles and their plasmonic shifts to iron oxide nanoparticles and changes in magnetic properties, detection of pathogens, toxins, antigens and nucleic acids has been achieved with impressive detection thresholds. Additionally, as bacteria become resistant to antibiotics, nanotechnology has achieved the rapid determination of bacterial drug susceptibility and resistance using novel methods, such as amperometry and magnetic relaxation. Overall, these promising results hint to the adoption of nanotechnology-based diagnostics for the diagnosis of infectious diseases in diverse settings throughout the globe, preventing epidemics and safeguarding human and economic wellness.
Collapse
|
21
|
Hsieh YH, Liu SJ, Chen HW, Lin YK, Liang KS, Lai LJ. Highly sensitive rare cell detection based on quantum dot probe fluorescence analysis. Anal Bioanal Chem 2009; 396:1135-41. [PMID: 19998027 DOI: 10.1007/s00216-009-3323-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 11/08/2009] [Accepted: 11/16/2009] [Indexed: 11/25/2022]
Abstract
This study presents an efficient and sensitive method for detecting rare cells without cell culture, in which cells are analyzed quantitatively using quantum dots (QDs) as a fluorescent probe. By the conjugation of QDs with cells, the biotin-streptavidin reaction functions as a bridge to connect QDs and cells. The cells can be quantified based on the correlation of the QD fluorescence intensity with the cell population. Non-specific adsorption and cross-reaction of QD625-streptavidin on T cell membrane are neglected by reacting with biotin anti-human CD3 and mixing with red blood cell, respectively. Additionally, the photo-activation period and pH can be controlled to enhance the fluorescence of cell populations, which increases linearly with the number of T cells from 40 to 100,000, not only in a single T cell line but also in mixing with a total of 10(6) red blood cells. Moreover, the specific T cells can be detected in less than 15 min, even though rare specific cells may number only 40 cells. Among the advantages, the proposed system for detecting rare cells include simplicity of preparation, low cost, rapid detection, and high sensitivity, all of which can facilitate the detection of circulating tumor cells in early stages of diagnosis or prognosis.
Collapse
Affiliation(s)
- Yi-Heui Hsieh
- National Synchrotron Radiation Research Center, 101 Hsin-Ann Road, Hsinchu Science Park, Hsinchu, 30076, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
22
|
Development of a fluorescent in situ method for visualization of enteric viruses. Appl Environ Microbiol 2009; 75:7822-7. [PMID: 19854924 DOI: 10.1128/aem.01986-09] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Studying the interactions between enteric pathogens and their environment is important to improving our understanding of their persistence and transmission. However, this remains challenging in large part because of difficulties associated with tracking pathogens in their natural environment(s). In this study, we report a fluorescent labeling strategy which was applied to murine norovirus (MNV-1), a human norovirus surrogate, and hepatitis A virus (HAV). Specifically, streptavidin-labeled Quantum dots (Q-Dots) were bound to biotinylated capsids of MNV-1 and HAV (bio-MNV-1 and bio-HAV); the process was confirmed by using a sandwich-type approach in which streptavidin-bound plates were reacted with biotinylated virus followed by a secondary binding to Q-Dots with an emission range of 635 to 675 nm (Q-Dots 655). The assay demonstrated a relative fluorescence of 528 +/- 48.1 and 112 +/- 8.6 for bio-MNV-1 and control MNV-1, respectively. The biotinylation process did not impact virus infectivity, nor did it interfere with the interactions between the virus and host cells or model produce items. Using fluorescent microscopy, it was possible to visualize both bio-HAV and bio-MNV-1 attached to the surfaces of permissive mammalian cells and green onion tissue. The method provides a powerful tool for the labeling and detection of enteric viruses (and their surrogates) which can be used to track virus behavior in situ.
Collapse
|
23
|
DC-SIGN mediates cell-free infection and transmission of human T-cell lymphotropic virus type 1 by dendritic cells. J Virol 2009; 83:10908-21. [PMID: 19692463 DOI: 10.1128/jvi.01054-09] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite the susceptibility of dendritic cells (DCs) to human T-cell lymphotropic virus type 1 (HTLV-1) infection and the defined role of these cells in disease pathogenesis, the mechanisms of viral binding to DCs have not been fully delineated. Recently, a glucose transporter, GLUT-1, heparan sulfate proteoglycans (HSPGs), and neuropilin-1 (NRP-1) were demonstrated to facilitate HTLV-1 entry into T cells. DCs express their own array of antigen receptors, the most important being the DC-specific intercellular adhesion molecule-3 (ICAM-3)-grabbing nonintegrin (DC-SIGN) with respect to retrovirus binding. Consequently, the role of DC-SIGN and other HTLV-1 attachment factors was analyzed in viral binding, transmission, and productive infection using monocyte-derived DCs (MDDCs), blood myeloid DCs, and B-cell lines expressing DC-SIGN. The relative expression of DC-SIGN, GLUT-1, HSPGs, and NRP-1 first was examined on both DCs and B-cell lines. Although the inhibition of these molecules reduced viral binding, HTLV-1 transmission from DCs to T cells was mediated primarily by DC-SIGN. DC-SIGN also was shown to play a role in the infection of MDDCs as well as model B-cell lines. The HTLV-1 infection of MDDCs also was achieved in blood myeloid DCs following the enhancement of virus-induced interleukin-4 production and subsequent DC-SIGN expression in this cell population. This study represents the first comprehensive analysis of potential HTLV-1 receptors on DCs and strongly suggests that DC-SIGN plays a critical role in HTLV-1 binding, transmission, and infection, thereby providing an attractive target for the development of antiretroviral therapeutics and microbicides.
Collapse
|
24
|
Manuel SL, Schell TD, Acheampong E, Rahman S, Khan ZK, Jain P. Presentation of human T cell leukemia virus type 1 (HTLV-1) Tax protein by dendritic cells: the underlying mechanism of HTLV-1-associated neuroinflammatory disease. J Leukoc Biol 2009; 86:1205-16. [PMID: 19656902 DOI: 10.1189/jlb.0309172] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
HTLV-1 is the etiologic agent of a debilitating neurologic disorder, HAM/TSP. This disease features a robust immune response including the oligoclonal expansion of CD8+ CTLs specific for the viral oncoprotein Tax. The key pathogenic process resulting in the proliferation of CTLs and the presentation of Tax peptide remains uncharacterized. We have investigated the role of APCs, particularly DCs, in priming of the anti-Tax CTL response under in vitro and in vivo conditions. We investigated two routes (direct vs. indirect) of Tax presentation using live virus, infected primary CD4+/CD25+ T cells, and the CD4+ T cell line (C8166, a HTLV-1-mutated line that only expresses Tax). Our results indicated that DCs are capable of priming a pronounced Tax-specific CTL response in cell cultures consisting of naïve PBLs as well as in HLA-A*0201 transgenic mice (line HHD II). DCs were able to direct the presentation of Tax successfully through infected T cells, live virus, and cell-free Tax. These observations were comparable with those made with a known stimulant of DC maturation, a combination of CD40L and IFN-gamma. Our studies clearly establish a role for this important immune cell component in HTLV-1 immuno/neuropathogenesis and suggest that modulation of DC functions could be an important tool for therapeutic interventions.
Collapse
Affiliation(s)
- Sharrón L Manuel
- Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | | | | | | | | | | |
Collapse
|
25
|
Sainathrao S, Mohan KVK, Atreya C. Gamma-phage lysin PlyG sequence-based synthetic peptides coupled with Qdot-nanocrystals are useful for developing detection methods for Bacillus anthracis by using its surrogates, B. anthracis-Sterne and B. cereus-4342. BMC Biotechnol 2009; 9:67. [PMID: 19624851 PMCID: PMC2722591 DOI: 10.1186/1472-6750-9-67] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 07/22/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Previous reports of site-directed deletion analysis on gamma (gamma)-phage lysin protein (PlyG) have demonstrated that removal of a short amino acid sequence in the C-terminal region encompassing a 10-amino acid motif (190LKMTADFILQ199) abrogates its binding activity specific to the cell wall of Bacillus anthracis. Whether short synthetic peptides representing the10-amino acid PlyG putative binding motif flanked by surrounding N- and C-terminal residues also selectively bind to the bacterial cell wall has not been evaluated. If such peptides do demonstrate selective binding to the cell wall, they could serve as bio-probes towards developing detection technologies for B. anthracis. Furthermore, by using B. anthracis (Sterne, 34F2), an animal vaccine and B. cereus-4342, a gamma-phage susceptible rare strain as surrogates of B. anthracis, development of proof-of-concepts for B. anthracis are feasible. RESULTS Using four different methods, we evaluated six synthetic peptides representing the putative binding motif including flanking sequences (PlyG-P1 through P6) for the bacterial cell wall binding capacity. Our analysis identified PlyG-P1, PlyG-P3 and PlyG-P5 to have binding capability to both B. anthracis (Sterne, 34F2) and B. cereus-4342. The peptides however did not bind to B. cereus-11778, B. thuringiensis, and B. cereus-10876 suggesting their specificity for B. anthracis-Sterne and B. cereus-4342. PlyG-P3 in combination with fluorescent light microscopy detected even a single bacterium in plasma spiked with the bacteria. CONCLUSION Overall, these studies illustrate that the short 10-amino acid sequence 'LKMTADFILQ' in fact is a stand-alone bacterial cell wall-binding motif of PlyG. In principle, synthetic peptides PlyG-P1, PlyG-P3 and PlyG-P5, especially PlyG-P3 coupled with Qdot-nanocrystals are useful as high-sensitivity bio-probes in developing detection technologies for B. anthracis.
Collapse
Affiliation(s)
- Shilpakala Sainathrao
- Section of Cell biology, Laboratory of Cellular Hematology, Center for Biologics Evaluation and Research, FDA, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
26
|
Douma K, Prinzen L, Slaaf DW, Reutelingsperger CPM, Biessen EAL, Hackeng TM, Post MJ, van Zandvoort MAMJ. Nanoparticles for optical molecular imaging of atherosclerosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2009; 5:544-557. [PMID: 19226595 DOI: 10.1002/smll.200801079] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Molecular imaging contributes to future personalized medicine dedicated to the treatment of cardiovascular disease, the leading cause of mortality in industrialized countries. Endoscope-compatible optical imaging techniques would offer a stand-alone alternative and high spatial resolution validation technique to clinically accepted imaging techniques in the (intravascular) assessment of vulnerable atherosclerotic lesions, which are predisposed to initiate acute clinical events. Efficient optical visualization of molecular epitopes specific for vulnerable atherosclerotic lesions requires targeting of high-quality optical-contrast-enhancing particles. In this review, we provide an overview of both current optical nanoparticles and targeting ligands for optical molecular imaging of atherosclerotic lesions and speculate on their applicability in the clinical setting.
Collapse
Affiliation(s)
- Kim Douma
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Williams Y, Byrne S, Bashir M, Davies A, Whelan A, Gun'ko Y, Kelleher D, Volkov Y. Comparison of three cell fixation methods for high content analysis assays utilizing quantum dots. J Microsc 2008; 232:91-8. [PMID: 19017205 DOI: 10.1111/j.1365-2818.2008.02083.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Semiconductor nanoparticles or quantum dots are being increasingly utilized as fluorescent probes in cell biology both in live and fixed cell assays. Quantum dots possess an immense potential for use in multiplexing assays that can be run on high content screening analysers. Depending on the nature of the biological target under investigation, experiments are frequently required on cells retaining an intact cell membrane or also on those that have been fixed and permeabilized to expose intracellular antigens. Fixation of cell lines before or after the addition of quantum dots may affect their localization, emission properties and stability. Using a high content analysis platform we perform a quantitative comparative analysis of three common fixation techniques in two different cell lines exposed to carboxylic acid stabilized CdTe quantum dots. Our study demonstrates that in prefixed and permeabilized cells, quantum dots are readily internalized regardless of cell type, and their intracellular location is primarily determined by the properties of the quantum dots themselves. However, if the fixation procedures are preformed on live cells previously incubated with quantum dots, other important factors have to be considered. The choice of the fixative significantly influences the fluorescent characteristics of the quantum dots. Fixatives, regardless of their chemical nature, negatively affected quantum dots fluorescence intensity. Comparative analysis of gluteraldehyde, methanol and paraformaldehyde demonstrated that 2% paraformaldehyde was the fixative of choice. The presence of protein in the media did not significantly alter the quantum dot fluorescence. This study indicates that multiplexing assays utilizing quantum dots, despite being a cutting edge tool for high content cell imaging, still require careful consideration of the basic steps in biological sample processing.
Collapse
Affiliation(s)
- Y Williams
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College, Dublin 2, Dublin, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Spirig R, van Kooten C, Obregon C, Nicod L, Daha M, Rieben R. The complement inhibitor low molecular weight dextran sulfate prevents TLR4-induced phenotypic and functional maturation of human dendritic cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:878-90. [PMID: 18606639 DOI: 10.4049/jimmunol.181.2.878] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Low molecular weight dextran sulfate (DXS) has been reported to inhibit the classical, alternative pathway as well as the mannan-binding lectin pathway of the complement system. Furthermore, it acts as an endothelial cell protectant inhibiting complement-mediated endothelial cell damage. Endothelial cells are covered with a layer of heparan sulfate (HS), which is rapidly released under conditions of inflammation and tissue injury. Soluble HS induces maturation of dendritic cells (DC) via TLR4. In this study, we show the inhibitory effect of DXS on human DC maturation. DXS significantly prevents phenotypic maturation of monocyte-derived DC and peripheral myeloid DC by inhibiting the up-regulation of CD40, CD80, CD83, CD86, ICAM-1, and HLA-DR and down-regulates DC-SIGN in response to HS or exogenous TLR ligands. DXS also inhibits the functional maturation of DC as demonstrated by reduced T cell proliferation, and strongly impairs secretion of the proinflammatory mediators IL-1beta, IL-6, IL-12p70, and TNF-alpha. Exposure to DXS leads to a reduced production of the complement component C1q and a decreased phagocytic activity, whereas C3 secretion is increased. Moreover, DXS was found to inhibit phosphorylation of IkappaB-alpha and activation of NF-kappaB. These findings suggest that DXS prevents TLR-induced maturation of human DC and may therefore be a useful reagent to impede the link between innate and adaptive immunity.
Collapse
Affiliation(s)
- Rolf Spirig
- Departmentof Clinical Research, Laboratory of Cardiovascular Surgery, University of Bern, Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
29
|
Jones KS, Petrow-Sadowski C, Huang YK, Bertolette DC, Ruscetti FW. Cell-free HTLV-1 infects dendritic cells leading to transmission and transformation of CD4(+) T cells. Nat Med 2008; 14:429-36. [PMID: 18376405 DOI: 10.1038/nm1745] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Accepted: 02/29/2008] [Indexed: 11/09/2022]
Abstract
Cell-free human T-lymphotropic virus type 1 (HTLV-1) virions are poorly infectious in vitro for their primary target cells, CD4(+) T cells. Here, we show that HTLV-1 can efficiently infect myeloid and plasmacytoid dendritic cells (DCs). Moreover, DCs exposed to HTLV-1, both before and after being productively infected, can rapidly, efficiently and reproducibly transfer virus to autologous primary CD4(+) T cells. This DC-mediated transfer of HTLV-1 involves heparan sulfate proteoglycans and neuropilin-1 and results in long-term productive infection and interleukin-2-independent transformation of the CD4(+) T cells. These studies, along with observations of HTLV-1-infected DCs in the peripheral blood of infected individuals, indicate that DCs have a central role in HTLV-1 transmission, dissemination and persistence in vivo. In addition to altering the current paradigm concerning how HTLV-1 transmission occurs, these studies suggest that impairment of DC function after HTLV-1 infection plays a part in pathogenesis.
Collapse
Affiliation(s)
- Kathryn S Jones
- Basic Research Program, Science Applications International Corporation-Frederick, Frederick, Maryland 21702, USA.
| | | | | | | | | |
Collapse
|
30
|
Shen J, Xu F, Jiang H, Wang Z, Tong J, Guo P, Ding S. Characterization and application of quantum dot nanocrystal–monoclonal antibody conjugates for the determination of sulfamethazine in milk by fluoroimmunoassay. Anal Bioanal Chem 2007; 389:2243-50. [PMID: 17899028 DOI: 10.1007/s00216-007-1609-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 08/29/2007] [Accepted: 09/04/2007] [Indexed: 10/22/2022]
Abstract
Quantum dot (Qdot) nanocrystals have been increasingly used as fluorescence labels in fluoroimmunoassays recently because of their excellent optical characteristics. In this paper, a new monoclonal antibody (MAb) against sulfamethazine (SMZ) was successfully produced and linked to Qdot nanocrystals by covalent coupling. The Qdot-MAb conjugates were characterized by SDS-PAGE and high-performance capillary electrophoresis (HPCE). An enzyme-linked immunosorbent assay (ELISA) method was utilized to evaluate the antigen-antibody binding affinity and then a novel direct competitive fluorescence-linked immunosorbent assay (cFLISA) for the detection of SMZ in milk by using Qdots as fluorescent labels was evaluated. The results showed that the 50% inhibition values (IC50) of the cFLISA were 4.3 ng/mL in milk and 5.2 ng/mL in PBS, and the limits of detection (LODs) were 0.6 ng/mL in milk and 0.4 ng/mL in PBS, respectively. The recoveries of SMZ from spiked milk samples at levels of 10-100 ng/mL ranged from 94 to 106%, with coefficients of variation (CVs) of 2.1-9.2%.
Collapse
Affiliation(s)
- Jianzhong Shen
- National Reference Laboratories for Veterinary Drug Residue, Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing, 100094, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|