51
|
Tao R, Li L, Huang W, Zheng L. Activation of human dendritic cells by recombinant modified vaccinia virus Ankara vectors encoding survivin and IL-2 genes in vitro. Hum Gene Ther 2010; 21:98-108. [PMID: 19715401 DOI: 10.1089/hum.2009.113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) has attracted significant attention as a safe, promising vector for immunotherapy. However, the precise effects of MVA infection on immune responses in humans remain largely unknown. We constructed recombinant MVA (rMVA) encoding both a human tumor-associated antigen (survivin) and the proinflammatory cytokine interleukin (IL)-2 and investigated their effects on human monocyte-derived dendritic cells (DCs). The results showed that infection with rMVA slightly impaired the upregulation of CD83 and reduced the production of IL-10 in DCs after lipopolysaccharide stimulation. However, rMVA-infected DCs were still able to express high levels of target genes and the costimulatory molecules CD80 and CD86 and to produce significant amounts of the proinflammatory cytokine tumor necrosis factor alpha. Moreover, rMVA-infected DCs exhibited a greater capacity than uninfected cells to stimulate T-cell proliferation and to reverse MVA-induced apoptosis in syngeneic T cells. Coculture of lymphocytes with rMVA-infected DCs significantly increased cytotoxic potential and interferon gamma production by cytotoxic T cells. These findings suggest that rMVA encoding survivin and IL-2 can effectively stimulate the activation of human DCs and overcome defects such as impairment of DC maturation and apoptosis of lymphocytes that are caused by vector alone. Thus, this study may provide a rational basis for further optimization of MVA vector.
Collapse
Affiliation(s)
- Ran Tao
- State Key Laboratory of Biocontrol, Cancer Center, Sun Yat-Sen (Zhongshan) University , Guangzhou 510275, P.R. China
| | | | | | | |
Collapse
|
52
|
Bermúdez-Humarán LG, Langella P. Utilisation des bactéries lactiques comme vecteurs vaccinaux. REVUE FRANCOPHONE DES LABORATOIRES 2009; 2009:79-89. [PMID: 32518601 PMCID: PMC7270964 DOI: 10.1016/s1773-035x(09)70312-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 10/12/2009] [Indexed: 11/26/2022]
Abstract
Aujourd’hui, nous disposons de données suffisantes qui confortent l’intérêt d’utiliser des bactéries lactiques (BL), notamment des souches des lactocoques et lactobacilles, pour le développement de nouvelles stratégies de vaccination mucosale. Les BL sont des bactéries à Gram positif utilisées depuis des millénaires dans la production d’aliments fermentés. Elles sont donc de bonnes candidates pour le développement de nouvelles stratégies de vectorisation orale et constituent des alternatives attractives aux stratégies vaccinales basées sur des bactéries pathogènes atténuées dont l’utilisation présente des risques sanitaires. Ce chapitre passe en revue la recherche et les progrès les plus récents dans l’utilisation des BL comme vecteurs de délivrance de protéines d’intérêt médical pour développer de nouveaux vaccins.
Collapse
|
53
|
Borovkov A, Magee DM, Loskutov A, Cano JA, Selinsky C, Zsemlye J, Lyons CR, Sykes K. New classes of orthopoxvirus vaccine candidates by functionally screening a synthetic library for protective antigens. Virology 2009; 395:97-113. [PMID: 19800089 DOI: 10.1016/j.virol.2009.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 07/22/2009] [Accepted: 09/05/2009] [Indexed: 10/20/2022]
Abstract
The licensed smallpox vaccine, comprised of infectious vaccinia, is no longer popular as it is associated with a variety of adverse events. Safer vaccines have been explored such as further attenuated viruses and component designs. However, these alternatives typically provide compromised breadth and strength of protection. We conducted a genome-level screening of cowpox, the ancestral poxvirus, in the broadly immune-presenting C57BL/6 mouse as an approach to discovering novel components with protective capacities. Cowpox coding sequences were synthetically built and directly assayed by genetic immunization for open-reading frames that protect against lethal pulmonary infection. Membrane and non-membrane antigens were identified that partially protect C57BL/6 mice against cowpox and vaccinia challenges without adjuvant or regimen optimization, whereas the 4-pox vaccine did not. New vaccines might be developed from productive combinations of these new and existing antigens to confer potent, broadly efficacious protection and be contraindicated for none.
Collapse
Affiliation(s)
- Alexandre Borovkov
- Center for Innovations in Medicine at The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | | | | | | | | | | | | | | |
Collapse
|
54
|
Lace MJ, Yamakawa Y, Ushikai M, Anson JR, Haugen TH, Turek LP. Cellular factor YY1 downregulates the human papillomavirus 16 E6/E7 promoter, P97, in vivo and in vitro from a negative element overlapping the transcription-initiation site. J Gen Virol 2009; 90:2402-2412. [PMID: 19553391 DOI: 10.1099/vir.0.012708-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cellular factors that bind to cis sequences in the human papillomavirus 16 (HPV-16) upstream regulatory region (URR) positively and negatively regulate the viral E6 and E7 oncogene promoter, P97. DNase I footprinting has revealed the binding of cellular proteins to two previously undetected cis elements overlapping and 3′ of the transcription-initiation site of the P97 promoter. Mutations within homologous motifs found in both of these cis elements abolished their negative function in vivo and the binding of the same cellular complex in vitro. This factor was identified as YY1 by complex mobility and binding specificity in comparison with vaccinia virus-expressed, purified recombinant YY1 protein and by antigenic reactivity with YY1 antisera. Cis mutations in the ‘initiator’ YY1 site activated the P97 promoter in vivo and in vitro. P97 was also activated threefold in vitro by depletion of endogenous YY1 with wild-type, but not mutant, YY1 oligonucleotides from the IgH kappa E3′ enhancer. Furthermore, increasing concentrations of exogenous, purified recombinant YY1 repressed wild-type P97 transcript levels by up to threefold, but did not influence the P97 promoter mutated in the ‘initiator’ YY1 site. Thus, the promoter-proximal YY1 site was not necessary for correct transcription initiation at the P97 promoter, but was found to be required for downregulation of P97 transcription in vivo and in vitro. In contrast to other viral and cellular promoters, where YY1 is thought to function as a positive transcription-‘initiator’ factor, HPV-16 P97 transcription is downregulated by YY1 from a critical motif overlapping the transcription start site.
Collapse
Affiliation(s)
- Michael J. Lace
- Department of Pathology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
- Veterans Affairs Medical Center, 601 Highway 6 West, Iowa City, IA 52246, USA
| | - Yasushi Yamakawa
- Veterans Affairs Medical Center, 601 Highway 6 West, Iowa City, IA 52246, USA
| | - Masato Ushikai
- Veterans Affairs Medical Center, 601 Highway 6 West, Iowa City, IA 52246, USA
| | - James R. Anson
- Veterans Affairs Medical Center, 601 Highway 6 West, Iowa City, IA 52246, USA
| | - Thomas H. Haugen
- Department of Pathology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
- Veterans Affairs Medical Center, 601 Highway 6 West, Iowa City, IA 52246, USA
| | - Lubomir P. Turek
- Department of Pathology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242, USA
- Veterans Affairs Medical Center, 601 Highway 6 West, Iowa City, IA 52246, USA
| |
Collapse
|
55
|
Abstract
Oncolytic or replication-selective viruses have been used as powerful tools for the delivery of therapeutic genes to tumors. Because these vectors are capable of replicating within the tumor, the therapeutic gene is amplified within the target tissue itself, resulting in the spread of the virus both within the tumor, and sometimes also between tumors. Vaccinia virus holds many advantages when serving as the backbone for oncolytic viral strains, including a large cloning capacity (at least 25 kbp) (1); a short life-cycle (2, 3); extensive previous use in humans, with contraindications and adverse reactions well described and antivirals available (4); the potential for systemic (intravenous) delivery to distant tumors; and vaccinia strains have previously demonstrated antitumor benefits in clinical trials (5). Because vaccinia has no known receptor and is capable of infecting almost any cell type, tumor selectivity has to be engineered into vaccinia at steps after infection. We will therefore discuss potential viral virulence genes and metabolic targets that result in tumor-selective vaccinia strains. Because the virus has limited natural requirements for host cell proteins, and, instead, contains a large genome and multiple genes involved in virulence, a large number of possible attenuating gene deletions can result in the production of viral strains reliant on inherent properties of the host cell for replication. The protocols for producing viral gene deletions and constructing viral gene expression vectors have been well established for vaccinia and are summarized briefly in this chapter. Basic assays for testing the tumor selectivity and therapeutic index of new oncolytic constructs in vitro will be covered. In addition, we describe how bioluminescence imaging can be incorporated into preclinical testing of vaccinia gene expression strains to examine the timing, biodistribution, and kinetics of viral gene expression noninvasively after delivery of the viral agents to tumor-bearing mice via different routes.
Collapse
|
56
|
Transcriptionally regulated, prostate-targeted gene therapy for prostate cancer. Adv Drug Deliv Rev 2009; 61:572-88. [PMID: 19393705 DOI: 10.1016/j.addr.2009.03.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 03/10/2009] [Indexed: 01/08/2023]
Abstract
Prostate cancer is the most frequently diagnosed cancer and the second leading cause of cancer deaths in American males today. Novel and effective treatment such as gene therapy is greatly desired. The early viral based gene therapy uses tissue-nonspecific promoters, which causes unintended toxicity to other normal tissues. In this chapter, we will review the transcriptionally regulated gene therapy strategy for prostate cancer treatment. We will describe the development of transcriptionally regulated prostate cancer gene therapy in the following areas: (1) Comparison of different routes for best viral delivery to the prostate; (2) Study of transcriptionally regulated, prostate-targeted viral vectors: specificity and activity of the transgene under several different prostate-specific promoters were compared in vitro and in vivo; (3) Selection of therapeutic transgenes and strategies for prostate cancer gene therapy (4) Oncolytic virotherapy for prostate cancer. In addition, the current challenges and future directions in this field are also discussed.
Collapse
|
57
|
Ilyinskii PO, Nurminskaya MV, Gabai VL, Prilipov AG, Usachev EV, Zakharova LG, Thoidis G, Altstein AD, Shneider AM. Effective expression of recombinant cytotoxic protein via its attachment to a polyglutamine domain. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2009; 13:211-7. [PMID: 19441878 DOI: 10.1089/omi.2009.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Inadvertent cytotoxicity may hinder the expression of many recombinant proteins that are of industrial or medicinal importance. Here, we show that covalent binding of the influenza A cytotoxic protein M2 to a polyglutamine domain (polyQ-M2; QM2) results in significant delay of its cytotoxic effects when compared to wild-type protein (M2wt). We also show that while expression of recombinant M2wt from A/WSN/1933 strain could not be attained in vaccinia virus (VV), polyQ-M2 was successfully expressed in this system. Moreover, we demonstrate that in cell culture, the polyQ domain is cleaved off following 48 h of expression, thus releasing free and active M2. Similarly, we show the spontaneous cleavage and polyQ release from fusion with another distinct polypeptide, green fluorescent protein (GFP). Expression of M2 from QM2 construct was more prolonged than one based on M2wt-expressing construct, markedly exceeding it at the later time points. Therefore, cell death caused by a toxic polypeptide may be suppressed via genetic fusion with polyQ, resulting in its enhanced expression, followed by slow release of the free polypeptide from the fusion. Collectively, covalent fusion with polyQ or other aggregate-forming domains presents a novel approach for industrial production of cytotoxic proteins and also holds promise for gene therapy applications.
Collapse
|
58
|
Chen X, Bai F, Sokol L, Zhou J, Ren A, Painter JS, Liu J, Sallman DA, Chen YA, Yoder JA, Djeu JY, Loughran TP, Epling-Burnette PK, Wei S. A critical role for DAP10 and DAP12 in CD8+ T cell-mediated tissue damage in large granular lymphocyte leukemia. Blood 2009; 113:3226-34. [PMID: 19075187 PMCID: PMC2665892 DOI: 10.1182/blood-2008-07-168245] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Accepted: 11/23/2008] [Indexed: 01/02/2023] Open
Abstract
Large granular lymphocyte (LGL) leukemia, or LGLL, is characterized by increased numbers of circulating clonal LGL cells in association with neutropenia, anemia, rheumatoid arthritis, and pulmonary artery hypertension (PAH). Emerging evidence suggests that LGLL cells with a CD8(+)CD28(null) phenotype induce these clinical manifestations through direct destruction of normal tissue. Compared with CD8(+)CD28(null) T cells from healthy controls, CD8(+)CD28(null) T cells from LGLL patients have acquired the ability to directly lyse pulmonary artery endothelial cells and human synovial cells. Here, we show that LGLL cells from patients possess enhanced cytotoxic characteristics and express elevated levels of activating natural killer receptors as well as their signaling partners, DAP10 and DAP12. Moreover, downstream targets of DAP10 and DAP12 are constitutively activated in LGLL cells, and expression of dominant-negative DAP10 and DAP12 dramatically reduces their lytic capacity. These are the first results to show that activating NKR-ligand interactions play a critical role in initiating the DAP10 and DAP12 signaling events that lead to enhanced lytic potential of LGLL cells. Results shown suggest that inhibitors of DAP10 and DAP12 or other proteins involved in this signaling pathway will be attractive therapeutic targets for the treatment of LGLL and other autoimmune diseases and syndromes.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/physiology
- CD28 Antigens/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cytotoxicity, Immunologic/genetics
- Endothelial Cells/immunology
- Endothelial Cells/pathology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Humans
- K562 Cells
- Leukemia, Large Granular Lymphocytic/genetics
- Leukemia, Large Granular Lymphocytic/immunology
- Leukemia, Large Granular Lymphocytic/metabolism
- Leukemia, Large Granular Lymphocytic/pathology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Membrane Proteins/physiology
- Phosphatidylinositol 3-Kinases/metabolism
- Pulmonary Artery/immunology
- Pulmonary Artery/pathology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/physiology
- Receptors, Natural Killer Cell/metabolism
- Signal Transduction/genetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Xianghong Chen
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Aspects of Microparticle Utilization for Potentiation of Novel Vaccines: Promises and Risks. NATO SCIENCE FOR PEACE AND SECURITY SERIES B: PHYSICS AND BIOPHYSICS 2009. [PMCID: PMC7122810 DOI: 10.1007/978-90-481-2523-4_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Many recombinant vaccines against novel (HIV, HCV) or ever-changing (influenza) infectious agents require the presence of adjuvants/delivery vehicles to induce strong immune responses. The necessity of their improvement led to the major effort towards development of vaccine delivery systems that are generally particulate (e.g., nano- and microparticles) and have comparable dimensions to the pathogens (viruses or bacteria). The mode of action of these adjuvants is not fully understood but implies the stimulation of the innate or antigen-specific immune responses, and/or the increase of antigen uptake or processing by antigen-presenting cells (APC). Moreover, enhancement of adjuvant activity through the use of micro- and nanoparticulate delivery systems often resulted from the synergistic effects producing immune responses stronger than those elicited by the adjuvant or delivery system alone. Among particulate adjuvants, biodegradable micro- and nanoparticles of poly(D,L-lactide-co-glycoside) (PLGA) or poly(D,L-lactide) (PLA) have been reported to enhance both humoral and cellular immune responses against an encapsulated protein antigen. Cationic and anionic polylactide co-glycolide (PLG) microparticles have been successfully used to adsorb a variety of agents, which include plasmid DNA, recombinant proteins and adjuvant active oligonucleotides and are also currently tested in several vaccine applications. Another approach envisions specific targeting of APC, especially peripheral DC and exploitation of particulate systems that are small enough for lymphatic uptake (polystyrene nanobeads). Micro- and nanoparticles offer the possibility of enhancement of their uptake by appropriate cells through manipulation of their surface properties. Still, questions regarding toxicity and molecular interaction between micro- and nano-particles and immune cells, tissues and whole organisms remain to be addressed. These risks and other possible side effects should be assessed in detail especially if mass-production and massive administration of such preparations is to be considered.
Collapse
|
60
|
Charalampopoulos D, Rastall RA. Development of Mucosal Vaccines Based on Lactic Acid Bacteria. PREBIOTICS AND PROBIOTICS SCIENCE AND TECHNOLOGY 2009. [PMCID: PMC7121035 DOI: 10.1007/978-0-387-79058-9_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Today, sufficient data are available to support the use of lactic acid bacteria (LAB), notably lactococci and lactobacilli, as delivery vehicles for the development of new mucosal vaccines. These non-pathogenic Gram-positive bacteria have been safely consumed by humans for centuries in fermented foods. They thus constitute an attractive alternative to the attenuated pathogens (most popular live vectors actually studied) which could recover their pathogenic potential and are thus not totally safe for use in humans. This chapter reviews the current research and advances in the use of LAB as live delivery vectors of proteins of interest for the development of new safe mucosal vaccines. The use of LAB as DNA vaccine vehicles to deliver DNA directly to antigen-presenting cells of the immune system is also discussed.
Collapse
Affiliation(s)
| | - Robert A. Rastall
- Department of Food Biosciences, University of Reading Whiteknights, Reading, UK
| |
Collapse
|
61
|
Subcellular forms and biochemical events triggered in human cells by HCV polyprotein expression from a viral vector. Virol J 2008; 5:102. [PMID: 18793431 PMCID: PMC2553408 DOI: 10.1186/1743-422x-5-102] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 09/15/2008] [Indexed: 12/16/2022] Open
Abstract
To identify the subcellular forms and biochemical events induced in human cells after HCV polyprotein expression, we have used a robust cell culture system based on vaccinia virus (VACV) that efficiently expresses in infected cells the structural and nonstructural proteins of HCV from genotype 1b (VT7-HCV7.9). As determined by confocal microscopy, HCV proteins expressed from VT7-HCV7.9 localize largely in a globular-like distribution pattern in the cytoplasm, with some proteins co-localizing with the endoplasmic reticulum (ER) and mitochondria. As examined by electron microscopy, HCV proteins induced formation of large electron-dense cytoplasmic structures derived from the ER and containing HCV proteins. In the course of HCV protein production, there is disruption of the Golgi apparatus, loss of spatial organization of the ER, appearance of some "virus-like" structures and swelling of mitochondria. Biochemical analysis demonstrate that HCV proteins bring about the activation of initiator and effector caspases followed by severe apoptosis and mitochondria dysfunction, hallmarks of HCV cell injury. Microarray analysis revealed that HCV polyprotein expression modulated transcription of genes associated with lipid metabolism, oxidative stress, apoptosis, and cellular proliferation. Our findings demonstrate the uniqueness of the VT7-HCV7.9 system to characterize morphological and biochemical events related to HCV pathogenesis.
Collapse
|
62
|
JNK-deficiency enhanced oncolytic vaccinia virus replication and blocked activation of double-stranded RNA-dependent protein kinase. Cancer Gene Ther 2008; 15:616-24. [PMID: 18535619 DOI: 10.1038/cgt.2008.32] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vaccinia virus has recently been used as an expression vector for gene delivery and an oncolytic agent for cancer therapy. Although it has been established that interferon-induced double-stranded RNA (dsRNA)-activated protein kinase (PKR) and RNase L interfere with viral replication, little else is known about the other host factors that might affect viral replication and virus-mediated host cell killing. In this study, we evaluated the roles of c-Jun NH2-terminal kinase (JNK) in oncolytic vaccinia virus replication and vaccinia virus-mediated host cell killing. We found that JNK knockout mouse embryonic fibroblasts (MEFs) were more susceptible to oncolytic vaccinia virus infection than wild-type MEFs. Moreover, viral replication and the production of infectious viral progeny were up to 100-fold greater in JNK-deficient MEFs than in wild-type MEFs. A similar result was observed for wild-type vaccinia virus. The increased killing of infected cells and the production of viral progeny was also observed in wild-type MEFs that had been treated with JNK inhibitors and in human colon cancer cells that had been transfected with dominant-negative JNK constructs. Moreover, testing on several human lung cancer cell lines and HeLa cells showed an inverse correlation between levels of JNK expression and susceptibility to oncolytic vaccinia virus. Our study also revealed that oncolytic virus infection-mediated PKR activation was blocked or diminished in JNK-deficient MEFs. The adenovirus-mediated ectopic expression of human PKR in JNK-deficient MEFs reduced vaccinia virus replication to the levels observed in wild-type MEFs, indicating that JNK is required for vaccinia virus to efficiently activate PKR. Our results demonstrated that the cellular status of JNK function can dramatically affect oncolytic vaccinia virus replication and vaccinia virus-mediated host cell killing. This finding may enable further improvements in oncolytic virotherapy using vaccinia virus.
Collapse
|
63
|
Abstract
BACKGROUND Current therapies for multiple myeloma (MM) are not curative, thus novel targeted therapeutics are being developed. One such targeted therapy is oncolytic virotherapy, wherein viruses specifically infect and kill the malignant plasma cells, leaving normal cells intact. OBJECTIVE This review provides an overview of the mechanisms and results of the oncolytic viruses being used to date and discusses the recent advances in the field of virotherapy for MM. METHODS All papers using viruses to treat MM were identified and screened. Only papers describing replicating, oncolytic viruses were reviewed. RESULTS/CONCLUSIONS Several viruses are currently being developed preclinically and clinically to treat MM, including measles virus, vesicular stomatitis virus, coxsackievirus A21 and vaccinia virus. Other viruses are being used preclinically to purge myeloma cells from autologous bone marrow transplants. Efforts to improve myeloma-specific targeting, avoid the antiviral immune response and evaluate combination therapies for MM are ongoing.
Collapse
Affiliation(s)
- Amaalia E Stief
- Experimental Therapeutics, Toronto General Research Institute, 67 College Street, Room 4-408, Toronto, ON, M5G 2M1, Canada
| | | |
Collapse
|
64
|
|
65
|
Grosenbach DW, Jordan R, King DS, Berhanu A, Warren TK, Kirkwood-Watts DL, Tyavanagimatt S, Tan Y, Wilson RL, Jones KF, Hruby DE. Immune responses to the smallpox vaccine given in combination with ST-246, a small-molecule inhibitor of poxvirus dissemination. Vaccine 2007; 26:933-46. [PMID: 18226434 PMCID: PMC2757089 DOI: 10.1016/j.vaccine.2007.11.095] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Revised: 10/26/2007] [Accepted: 11/29/2007] [Indexed: 11/24/2022]
Abstract
The re-emerging threat of smallpox and the emerging threat of monkeypox highlight the need for effective poxvirus countermeasures. Currently approved smallpox vaccines have unacceptable safety profiles and, consequently, the general populace is no longer vaccinated, leading to an increasingly susceptible population. ST-246, a small-molecule inhibitor of poxvirus dissemination, has been demonstrated in various animal models to be safe and effective in preventing poxviral disease. This suggests that it may also be used to improve the safety of the traditional smallpox vaccine provided that it does not inhibit vaccine-induced protective immunity. In this study, we compared the immune responses elicited by the smallpox vaccine alone or in combination with ST-246 in mice. Normal lesion formation following dermal scarification with the attenuated New York City Board of Health strain (Dryvax), commonly referred to as a vaccine “take”, was not inhibited although severe lesions and systemic disease due to vaccination with the virulent Western Reserve (VV-WR) strain were prevented. The vaccine given with ST-246 did not affect cellular immune responses or neutralizing antibody titers although anti-vaccinia ELISA titers were slightly reduced. Vaccination in combination with ST-246 provided equivalent short- and long-term protection against lethal intranasal challenge with VV-WR when compared to vaccine alone. These results suggest that ST-246 does not compromise protective immunity elicited by the vaccine and provide the basis for future studies examining the efficacy of ST-246 in preventing or treating adverse events due to vaccination.
Collapse
Affiliation(s)
- Douglas W Grosenbach
- SIGA Technologies, 4575 SW Research Way, Suite 230, Corvallis, OR 97333, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Kim D, Hung CF, Wu TC. Monitoring the trafficking of adoptively transferred antigen- specific CD8-positive T cells in vivo, using noninvasive luminescence imaging. Hum Gene Ther 2007; 18:575-88. [PMID: 17576157 DOI: 10.1089/hum.2007.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Understanding of the trafficking of antigen-specific CD8(+) T cells in vivo will provide insight about how our immune system controls infectious diseases and cancers. In the current study we used a luciferase-expressing human papillomavirus type 16 (HPV-16) E7-specific CD8(+) T cell for adoptive transfer to control E7-expressing TC-1 tumor cells. We used noninvasive luminescence imaging to monitor the trafficking of E7-specific CD8(+) T cells over time. We also boosted the luciferase-expressing E7-specific CD8(+) T cells in vivo, using E7-expressing vaccinia. We found that injected E7-specific T cells preferentially migrated to the E7-expressing tumor site but not to the E7-negative control tumor site, and increased in number at the tumor site over time. In addition, vaccination with E7-expressing vaccinia led to a significant increase in the number of E7-specific CD8(+) T cells at the tumor site, resulting in a significant antitumor effect compared with vaccination with wild-type vaccinia. Thus, our data suggest that the antitumor effects generated by adoptive transfer of E7-specific CD8(+) T cells can be significantly enhanced by vaccination with E7-expressing vaccinia and that our system represents a plausible approach to investigate the trafficking and biology of antigen-specific T cells in vivo.
Collapse
Affiliation(s)
- Daejin Kim
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA
| | | | | |
Collapse
|
67
|
Abstract
Vaccines that comprise attenuated viral vectors encoding antigens from target pathogens generate potent T-cell responses. One such pathogen is malaria, and in particular the liver stage of its life cycle. Immunogenicity and efficacy studies in animals and humans have revealed the generation of memory T cells of both the central and effector phenotypes, depending on the viral vectors used in the malaria vaccination regime (viral species and serotype, combination and sequence for prime-boost) and suggest a divergence in their protective role. Being able to influence the memory T-cell make-up in a rational manner may allow us to develop more efficacious vaccines.
Collapse
|
68
|
Okamura T, Someya K, Matsuo K, Hasegawa A, Yamamoto N, Honda M. Recombinant vaccinia DIs expressing simian immunodeficiency virus gag and pol in mammalian cells induces efficient cellular immunity as a safe immunodeficiency virus vaccine candidate. Microbiol Immunol 2007; 50:989-1000. [PMID: 17179668 DOI: 10.1111/j.1348-0421.2006.tb03867.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A highly attenuated vaccinia virus substrain of Dairen-I (DIs) shows promise as a candidate vector for eliciting positive immunity against immune deficiency virus. DIs was randomly obtained by serial 1-day egg passages of a chorioarantoic membrane-adapted Dairen strain (DIE), resulting in substantial genomic deletion, including various genes regulating the virus-host-range. To investigate the impact of that deletion and of the subsequent insertion of a foreign gene into that region of DIs on the ability of the DIs recombinant to induce antigen-specific immunity, we generated a recombinant vaccinia DIs expressing fulllength gag and pol genes of simian immunodeficiency virus (SIV) (rDIsSIV gag/pol) and studied the biological and immunological characteristics of the recombinant natural mutant. The rDIsSIV gag/pol developed a tiny plaque on the chick embryo fibroblast (CEF). Viral particles of rDIsSIV gag/pol as well as SIV Gag-like particles were electromicroscopically detected in the cytoplasm. Interestingly, the recombinant DIs strain grows well in CEF cells but not in mammalian cells. While rDIsSIV gag/pol produces SIV proteins in mammalian HeLa and CV-1 cells, recombinant modified vaccinia Ankara strain (MVA) expressing SIV gag and pol genes (MVA/SIV239 gag/pol) clearly replicates in HeLa and CV-1 cell lines under synchronized growth conditions and produces the SIV protein in all cell lines. Moreover, intradermal administration of rDIsSIV gag/pol or of MVA/SIV239 gag/pol elicited similar levels of IFN-gamma spot-forming cells specific for SIV Gag. If the non-productive infection characteristically induced by recombinant DIs is sufficient to trigger immune induction, as we believe it is, then a human immunodeficiency virus vaccine employing the DIs recombinant would have the twin advantages of being both effective and safe.
Collapse
Affiliation(s)
- Tomotaka Okamura
- AIDS Research Center, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
69
|
Tsen KT, Dykeman EC, Sankey OF, Tsen SWD, Lin NT, Kiang JG. Probing the low-frequency vibrational modes of viruses with Raman scattering--bacteriophage M13 in water. JOURNAL OF BIOMEDICAL OPTICS 2007; 12:024009. [PMID: 17477724 DOI: 10.1117/1.2718935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Raman spectroscopy is used to study low-wave-number (</=20 cm(-1)) acoustic vibrations of the M13 phage. A well-defined Raman line is observed at around 8.5 cm(-1). The experimental results are compared with theoretical calculations based on an elastic continuum model and appropriate Raman selection rules derived from a bond polarizability model. The observed Raman mode is shown to belong to one of the Raman-active axial modes of the M13 phage protein coat. It is expected that the detection and characterization of this low-frequency vibrational mode can be used for applications in biomedical nanotechnology such as for monitoring the process of virus functionalization and self-assembly.
Collapse
Affiliation(s)
- Kong T Tsen
- Arizona State University, Department of Physics, Tempe, Arizona 85287-1504, USA.
| | | | | | | | | | | |
Collapse
|
70
|
Abstract
Prostate cancer is a major cause of mortality in men in the Western world. Although treatment of early stage prostate cancer with radiation therapy or prostatectomy is efficient in most cases, some patients develop a fatal hormone-refractory disease. Treatments in this case are limited to aggressive chemotherapies, which can reduce serum prostate-specific antigen (PSA) levels in some patients. Taxane- and platinum-compound-based chemotherapies produce a survival benefit of only a few months. Therefore, it is crucial to develop novel, well tolerated treatment strategies. Over the past years, immunotherapy of hormone-refractory prostate cancer has been studied in numerous clinical trials. The fact that the prostate is a non-essential organ makes prostate cancer an excellent target for immunotherapy. Administration of antibodies targeting the human epidermal growth factor receptor-2 or the prostate-specific membrane antigen led to stabilisation of PSA levels in several patients. Vaccination of prostate cancer patients with irradiated allogeneic prostate cell lines has demonstrated that whole cell-based vaccines can significantly attenuate increases in PSA. Two different recombinant viral expression vectors have been applied in prostate cancer treatment: poxvirus and adenovirus vectors. Both vaccines have the advantages of using a natural method to induce immune responses and achieving high levels of transgene expression. Vaccinia viruses in combination with recombinant fowlpox or canarypox virus have been used to express recombinant PSA. Several studies demonstrated that this approach is safe and can lead to stabilisation of PSA values. A very promising approach in prostate cancer immunotherapy is vaccination of patients with dendritic cells. Thereby, peptides, recombinant proteins, tumour lysates or messenger RNA have been used to deliver antigens to autologous dendritic cells. Loading of dendritic cells with up to five different peptides derived from multiple proteins expressed in prostate cancer demonstrated that cytotoxic T-cell responses could be elicited in prostate cancer patients. Sipuleucel-T (APC8015), an immunotherapy product consisting of antigen-presenting cells, loaded ex vivo with a recombinant fusion protein consisting of prostatic acid phosphatase linked to granulocyte-macrophage colony-stimulating factor, demonstrated in a phase III, placebo-controlled trial an improvement in median time to disease progression. The improvement in overall survival was 4.5 months for sipuleucel-T-treated patients compared with the placebo group. Although there is a minor increase in overall survival of metastatic prostate cancer patients with some approaches, more effective therapeutic strategies need to be developed.
Collapse
Affiliation(s)
- Michael Basler
- Division of Immunology, Department of Biology, University of Constance, Konstanz, Germany.
| | | |
Collapse
|
71
|
Dénes B, Yu J, Fodor N, Takátsy Z, Fodor I, Langridge WHR. Suppression of hyperglycemia in NOD mice after inoculation with recombinant vaccinia viruses. Mol Biotechnol 2006; 34:317-27. [PMID: 17284779 DOI: 10.1385/mb:34:3:317] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/11/2022]
Abstract
In autoimmune (type 1) diabetes, autoreactive lymphocytes destroy pancreatic beta-cells responsible for insulin synthesis. To assess the feasibility of gene therapy for type 1 diabetes, recombinant vaccinia virus (rVV) vectors were constructed expressing pancreatic islet autoantigens proinsulin (INS) and a 55-kDa immunogenic peptide from glutamic acid decarboxylase (GAD), and the immunomodulatory cytokine interleukin (IL)-10. To augment the beneficial effects of recombinant virus therapy, the INS and GAD genes were fused to the C terminus of the cholera toxin B subunit (CTB). Five-week-old non-obese diabetic (NOD) mice were injected once with rVV. Humoral antibody immune responses and hyperglycemia in the infected mice were analyzed. Only 20% of the mice inoculated with rVV expressing the CTB::INS fusion protein developed hyperglycemia, in comparison to 70% of the mice in the uninoculated animal group. Islets from pancreatic tissues isolated from euglycemic mice from this animal group showed no sign of inflammatory lymphocyte invasion. Inoculation with rVV producing CTB::GAD or IL-10 was somewhat less effective in reducing diabetes. Humoral antibody isotypes of hyperglycemic and euglycemic mice from all treated groups possessed similar IgG1/IgG2c antibody titer ratios from 19 to 32 wk after virus inoculation. In comparison with uninoculated mice, 11-wk-old NOD mice injected with virus expressing CTB::INS were delayed in diabetes onset by more than 4 wk. The experimental results demonstrate the feasibility of using rVV expressing CTB::INS fusion protein to generate significant protection and therapy against type 1 diabetes onset and progression.
Collapse
Affiliation(s)
- Béla Dénes
- Center for Molecular Biology and Gene Therapy, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, CA 92350, USA.
| | | | | | | | | | | |
Collapse
|
72
|
Kremer M, Suezer Y, Martinez-Fernandez Y, Münk C, Sutter G, Schnierle BS. Vaccinia virus replication is not affected by APOBEC3 family members. Virol J 2006; 3:86. [PMID: 17052331 PMCID: PMC1635045 DOI: 10.1186/1743-422x-3-86] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Accepted: 10/19/2006] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND The APOBEC3G protein represents a novel innate defense mechanism against retroviral infection. It facilitates the deamination of the cytosine residues in the single stranded cDNA intermediate during early steps of retroviral infection. Most poxvirus genomes are relatively A/T-rich, which may indicate APOBEC3G-induced mutational pressure. In addition, poxviruses replicate exclusively in the cytoplasm where APOBEC3G is located. It was therefore tempting to analyze whether vaccinia virus replication is affected by APOBEC3G. RESULTS The replication of vaccinia virus, a prototype poxvirus, was not, however, inhibited in APOBEC3G-expressing cells, nor did other members of the APOBEC3 family alter vaccinia virus replication. HIV counteracts APOBEC3G by inducing its degradation. However, Western blot analysis showed that the levels of APOBEC3G protein were not affected by vaccinia virus infection. CONCLUSION The data indicate that APOBEC3G is not a restriction factor for vaccinia virus replication nor is vaccinia virus able to degrade APOBEC3G.
Collapse
Affiliation(s)
- Melanie Kremer
- Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51–59, 63225 Langen, Germany
| | | | | | - Carsten Münk
- Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51–59, 63225 Langen, Germany
| | - Gerd Sutter
- Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51–59, 63225 Langen, Germany
| | | |
Collapse
|
73
|
Denes B, Krausova V, Fodor N, Timiryasova T, Henderson D, Hough J, Yu J, Fodor I, Langridge WHR. Protection of NOD mice from type 1 diabetes after oral inoculation with vaccinia viruses expressing adjuvanted islet autoantigens. J Immunother 2005; 28:438-48. [PMID: 16113600 DOI: 10.1097/01.cji.0000171315.82997.9a] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Oral administration of autoantigens and allergens can delay or suppress clinical disease in experimental autoimmune and allergic disorders. However, repeated feeding of large amounts of the tolerogens is required over long periods and is only partially effective in animals systemically sensitized to the ingested antigen. Enhanced suppression of type 1 autoimmune diabetes insulitis and hyperglycemia was demonstrated in both naive and immune animals following oral inoculation with plant-based antigens coupled to the cholera toxin B subunit (CTB). Thus, plant-synthesized antigens linked to the CTB adjuvant, can enhance suppression of inflammatory TH1 lymphocyte-mediated autoreactivity in both naive and immune animals. To stimulate adjuvant-autoantigen fusion protein biosynthesis in the gut mucosae, the authors evaluated oral inoculation of juvenile non-obese diabetic (NOD) mice with recombinant vaccinia virus (rVV) expressing fusion genes encoding CTB linked to the pancreatic islet autoantigens proinsulin (INS) and a 55-kDa C-terminal peptide from glutamate decarboxylase (GAD55). Hyperglycemia in both rVV-CTB:: INS and rVV-CTB:: GAD inoculated mice was substantially reduced in comparison with the uninoculated mouse control. Oral inoculation with rVV carrying the CTB::INS fusion gene generated a significant reduction in insulitis. An increase in IgG1 in comparison with IgG2c antibody isotype titers in rVV-CTB::INS infected mice suggested possible activation of autoantigen specific Th2 lymphocytes. The experimental results demonstrate feasibility of using vaccinia virus oral delivery of adjuvanted autoantigens to the mucosae of prediabetic mice for suppression and therapy of type 1 autoimmune diabetes.
Collapse
Affiliation(s)
- Bela Denes
- Center for Molecular Biology and Gene Therapy, Department of Biochemistry and Microbiology, Loma Linda University, Loma Linda, CA 92350, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
74
|
McCart JA, Mehta N, Scollard D, Reilly RM, Carrasquillo JA, Tang N, Deng H, Miller M, Xu H, Libutti SK, Alexander HR, Bartlett DL. Oncolytic vaccinia virus expressing the human somatostatin receptor SSTR2: molecular imaging after systemic delivery using 111In-pentetreotide. Mol Ther 2005; 10:553-61. [PMID: 15336655 DOI: 10.1016/j.ymthe.2004.06.158] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2004] [Revised: 06/07/2004] [Accepted: 06/09/2004] [Indexed: 11/22/2022] Open
Abstract
Oncolytic vaccinia viruses (VV) have demonstrated tumor specificity, high levels of transgene expression, and anti-tumor effects. The ability to visualize vector biodistribution noninvasively will be necessary as gene therapy vectors come to clinical trials, and the creation of a VV that can both treat tumors and permit noninvasive imaging after systemic delivery is therefore an exciting concept. To facilitate imaging, a VV expressing the human somatostatin receptor type 2 (SSTR2) was created. Cells infected with the SSTR2-expressing VV or controls were incubated with the somatostatin analog 111In-pentetreotide with or without an excess of nonradiolabeled pentetreotide. The SSTR2-infected cells bound 111In-pentetreotide sixfold more efficiently than control virus-infected cells and this binding was specifically blocked by nonradiolabeled pentetreotide. Nude mice bearing subcutaneous murine colon CA xenografts were injected intraperitoneally with the SSTR2-expressing VV or control VV. After 6 days, mice were injected with 111In-pentetreotide and imaged. Mice were sacrificed and organs collected and counted in a gamma counter. The uptake of radioactivity in tumors and normal tissues (percentage injected dose per gram) and tumor-to-normal tissue ratios were determined. Tumors infected with the SSTR2-expressing VV accumulated significantly higher concentrations of radioactivity compared to tumors in animals receiving the control virus. SSTR2-infected tumors were visible on imaging 6 days after VV injection and could be visualized for up to 3 weeks post-viral injection using repeat injections of 111In-pentetreotide. This reporter gene imaging strategy could be a very effective method to visualize vector distribution, expression, and persistence over time and enhances the potential of VV as a novel anti-cancer therapeutic.
Collapse
Affiliation(s)
- J Andrea McCart
- Division of Experimental Therapeutics, Toronto General Research Institute, Toronto, ON, Canada.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Terasaka K, Blakeslee JJ, Titapiwatanakun B, Peer WA, Bandyopadhyay A, Makam SN, Lee OR, Richards EL, Murphy AS, Sato F, Yazaki K. PGP4, an ATP binding cassette P-glycoprotein, catalyzes auxin transport in Arabidopsis thaliana roots. THE PLANT CELL 2005; 17:2922-39. [PMID: 16243904 PMCID: PMC1276020 DOI: 10.1105/tpc.105.035816] [Citation(s) in RCA: 262] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Members of the ABC (for ATP binding cassette) superfamily of integral membrane transporters function in cellular detoxification, cell-to-cell signaling, and channel regulation. More recently, members of the multidrug resistance P-glycoprotein (MDR/PGP) subfamily of ABC transporters have been shown to function in the transport of the phytohormone auxin in both monocots and dicots. Here, we report that the Arabidopsis thaliana MDR/PGP PGP4 functions in the basipetal redirection of auxin from the root tip. Reporter gene studies showed that PGP4 was strongly expressed in root cap and epidermal cells. PGP4 exhibits apolar plasma membrane localization in the root cap and polar localization in tissues above. Root gravitropic bending and elongation as well as lateral root formation were reduced in pgp4 mutants compared with the wild type. pgp4 exhibited reduced basipetal auxin transport in roots and a small decrease in shoot-to-root transport consistent with a partial loss of the redirective auxin sink in the root. Seedlings overexpressing PGP4 exhibited increased shoot-to-root auxin transport. Heterologous expression of PGP4 in mammalian cells resulted in 1-N-naphthylthalamic acid-reversible net uptake of [3H]indole-3-acetic acid. These results indicate that PGP4 functions primarily in the uptake of redirected or newly synthesized auxin in epidermal root cells.
Collapse
Affiliation(s)
- Kazuyoshi Terasaka
- Laboratory of Molecular and Cellular Biology of Totipotency, Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kitashirakawa, Kyoto 606-8502, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Razumov IA, Gileva IP, Vasil'eva MA, Nepomnyashchikh TS, Mishina MN, Belanov EF, Kochneva GV, Konovalov EE, Shchelkunov SN, Loktev VB. Neutralizing Monoclonal Antibodies Cross-React with Fusion Proteins Encoded by 129L of the Ectromelia Virus and A30L of the Variola Virus. Mol Biol 2005. [DOI: 10.1007/s11008-005-0113-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
77
|
Geisler M, Blakeslee JJ, Bouchard R, Lee OR, Vincenzetti V, Bandyopadhyay A, Titapiwatanakun B, Peer WA, Bailly A, Richards EL, Ejendal KFK, Smith AP, Baroux C, Grossniklaus U, Müller A, Hrycyna CA, Dudler R, Murphy AS, Martinoia E. Cellular efflux of auxin catalyzed by the Arabidopsis MDR/PGP transporter AtPGP1. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2005; 44:179-94. [PMID: 16212599 DOI: 10.1111/j.1365-313x.2005.02519.x] [Citation(s) in RCA: 391] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Directional transport of the phytohormone auxin is required for the establishment and maintenance of plant polarity, but the underlying molecular mechanisms have not been fully elucidated. Plant homologs of human multiple drug resistance/P-glycoproteins (MDR/PGPs) have been implicated in auxin transport, as defects in MDR1 (AtPGP19) and AtPGP1 result in reductions of growth and auxin transport in Arabidopsis (atpgp1, atpgp19), maize (brachytic2) and sorghum (dwarf3). Here we examine the localization, activity, substrate specificity and inhibitor sensitivity of AtPGP1. AtPGP1 exhibits non-polar plasma membrane localization at the shoot and root apices, as well as polar localization above the root apex. Protoplasts from Arabidopsis pgp1 leaf mesophyll cells exhibit reduced efflux of natural and synthetic auxins with reduced sensitivity to auxin efflux inhibitors. Expression of AtPGP1 in yeast and in the standard mammalian expression system used to analyze human MDR-type proteins results in enhanced efflux of indole-3-acetic acid (IAA) and the synthetic auxin 1-naphthalene acetic acid (1-NAA), but not the inactive auxin 2-NAA. AtPGP1-mediated efflux is sensitive to auxin efflux and ABC transporter inhibitors. As is seen in planta, AtPGP1 also appears to mediate some efflux of IAA oxidative breakdown products associated with apical sites of high auxin accumulation. However, unlike what is seen in planta, some additional transport of the benzoic acid is observed in yeast and mammalian cells expressing AtPGP1, suggesting that other factors present in plant tissues confer enhanced auxin specificity to PGP-mediated transport.
Collapse
Affiliation(s)
- Markus Geisler
- Basel-Zurich Plant Science Center, University of Zurich, Institute of Plant Biology, CH-8007 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Feder-Mengus C, Schultz-Thater E, Oertli D, Marti WR, Heberer M, Spagnoli GC, Zajac P. Nonreplicating recombinant vaccinia virus expressing CD40 ligand enhances APC capacity to stimulate specific CD4+ and CD8+ T cell responses. Hum Gene Ther 2005; 16:348-60. [PMID: 15812230 DOI: 10.1089/hum.2005.16.348] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recombinant poxviruses expressing immunomodulatory molecules together with specific antigens represent powerful vaccines for cancer immunotherapy. Recently, we and others have demonstrated, in vitro and in vivo, that coexpression of CD80 and CD86 costimulatory molecules enhances the immunogenic capacity of a recombinant vaccinia virus (rVV) encoding different tumor-associated antigens. To further investigate the capacity of these vectors to provide ligands for different costimulatory pathways relevant in the generation of T cell responses, we constructed a recombinant virus (rVV) expressing CD40 ligand or CD154 (CD154rVV). Upon binding the CD40 receptor expressed on antigen presenting cells (APC), this molecule, physiologically expressed on activated CD4+ T cells, increases their antigen presentation and immunostimulatory capacities. Therefore, we evaluated the effects of CD154rVV infection on APC activation and its consequences on T cell stimulation. CD154rVV infection of autologous fibroblasts, monocytes, or iDC promoted the expression of a number of cytokines, including GM-CSF, TNF-alpha, and IL-15 in iDC. Most importantly, IL-12 p40 gene expression and protein secretion were induced by CD154rVV but not by wild-type VV (WT VV) in either CD14+ cells or iDC, and these effects could be blocked by anti-CD40 monoclonal antibodies. Furthermore, phenotypic characterization of CD154rVV infected iDC revealed enhanced expression of CD83 and CD86 surface markers as compared with wild-type vaccinia virus infection. As expected, VV infection triggered cytokines gene expression in cultures including APC and T cells from VV immune donors. However, cytokine genes typically expressed by T cell receptor triggered T cells such as those encoding IL-2 and IFN-gamma, or T cell proliferation, were detectable to a significantly higher extent in CD154rVV infected cultures, as compared with WT VV. Activation of specific CD8+ T cells was then investigated using MART-1/Melan-A(27-35) epitope as the model of tumor-associated antigen (TAA). In the presence of CD154rVV activated APCs, significantly higher numbers of specific cytotoxic CD8+ T cells were detected, as compared with cultures performed in the presence of WT VV or in the absence of virus. Taken together, these data indicate that functional CD154 expression from rVV infected cells promotes APC activation, thereby enhancing antigen-specific T cell generation. Such a recombinant vector might help bypass the requirement for activated helper cells during CTL priming, thus qualifying as a potentially relevant vector in the generation of CD8+ T cell responses in cancer immunotherapy.
Collapse
Affiliation(s)
- Chantal Feder-Mengus
- Oncology Group, Department of Surgery, Institut für Chirurgische Forschung und Spitalmanagement, Basel University Hospital Research Center, CH-4031 Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
79
|
Allen GA, Denes B, Fodor I, De Leon M. Vaccinia virus infection and gene transduction in cultured neurons. Microbes Infect 2005; 7:1087-96. [PMID: 15893497 DOI: 10.1016/j.micinf.2005.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2004] [Revised: 11/09/2004] [Accepted: 02/24/2005] [Indexed: 11/23/2022]
Abstract
The study of neurons in culture would benefit from the development of a gene transduction system capable of delivering foreign genes at high efficiency, as transduction of primary neurons with existing systems is inefficient. The efficacy of lytic vaccinia virus (VV) infection of primary retinal cultures and PC12 cells (a model of neuronal differentiation) was examined in order to determine the efficiency of gene transduction using VV in neuronal primary culture. VV was able to infect retinal cells and PC12 cells and express transgenes of Escherichia coli beta-galactosidase (lacZ) and epithelial fatty acid binding protein (E-FABP) in a virus dose-dependent manner. Most (50-100%) of the retinal cells were positive for transgene protein at multiplicities of infection (MOI) between 10 and 100 plaque-forming units (PFU), while over 50% of VV-infected PC12 cells expressed the virus encoded gene at an MOI = 10. The production of foreign mRNA and protein by VV following infection was verified by PCR and Western blot. Because VV is a lytic virus, cytopathic effects were examined. Retinal cultures maintained for 0.5 days in vitro showed greater than 90% survival at 24 h post-infection, while 14-day cultures were equally viable for 48 h. Retinal ganglion cells and differentiated PC12 cells appear to be more protected against lytic VV infection than proliferating glial and undifferentiated PC12 cells. These data suggest that VV may be a useful vector for delivering foreign genes to neuronal cells with an efficient transient transgene expression.
Collapse
Affiliation(s)
- Gregory A Allen
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | | | | | | |
Collapse
|
80
|
Kaufman HL, Deraffele G, Mitcham J, Moroziewicz D, Cohen SM, Hurst-Wicker KS, Cheung K, Lee DS, Divito J, Voulo M, Donovan J, Dolan K, Manson K, Panicali D, Wang E, Hörig H, Marincola FM. Targeting the local tumor microenvironment with vaccinia virus expressing B7.1 for the treatment of melanoma. J Clin Invest 2005; 115:1903-12. [PMID: 15937544 PMCID: PMC1142116 DOI: 10.1172/jci24624] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2005] [Accepted: 03/31/2005] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy for the treatment of metastatic melanoma remains a major clinical challenge. The melanoma microenvironment may lead to local T cell tolerance in part through downregulation of costimulatory molecules, such as B7.1 (CD80). We report the results from the first clinical trial, to our knowledge, using a recombinant vaccinia virus expressing B7.1 (rV-B7.1) for monthly intralesional vaccination of accessible melanoma lesions. A standard 2-dose-escalation phase I clinical trial was conducted with 12 patients. The approach was well tolerated with only low-grade fever, myalgias, and fatigue reported and 2 patients experiencing vitiligo. An objective partial response was observed in 1 patient and disease stabilization in 2 patients, 1 of whom is alive without disease 59 months following vaccination. All patients demonstrated an increase in postvaccination antibody and T cell responses against vaccinia virus. Systemic immunity was tested in HLA-A*0201 patients who demonstrated an increased frequency of gp100 and T cells specific to melanoma antigen recognized by T cells 1 (MART-1), also known as Melan-A, by ELISPOT assay following local rV-B7.1 vaccination. Local immunity was evaluated by quantitative real-time RT-PCR, which suggested that tumor regression was associated with increased expression of CD8 and IFN-gamma. The local delivery of vaccinia virus expressing B7.1 was well tolerated and represents an innovative strategy for altering the local tumor microenvironment in patients with melanoma.
Collapse
Affiliation(s)
- Howard L Kaufman
- Department of Surgery, Columbia University, New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Ma X, Sayeski PP. Vaccinia virus-mediated high level expression and single step purification of recombinant Jak2 protein. Protein Expr Purif 2005; 35:181-9. [PMID: 15135391 DOI: 10.1016/j.pep.2004.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2003] [Revised: 01/16/2004] [Indexed: 11/21/2022]
Abstract
Jak2 functions as a non-receptor tyrosine kinase and has been linked to pathologies such as cancer and cardiovascular disease. Because of this, many studies have tried to better understand its function. Unfortunately, very little information is known about its catalytic or biochemical properties as purification of significant amounts of functional Jak2 protein has been exceedingly difficult. Here, Jak2 was expressed in BSC-40 cells using a vaccinia virus-mediated expression system. Significant amounts ( approximately 10microg) of Jak2 protein were expressed from a single 100-mm cell culture dish. The protein was first harvested using three different methods of extraction to determine the relative efficiency of each lysis method with respect to Jak2 protein yield and catalytic activity. We found that lysis methods utilizing detergents increased the efficiency of protein extraction about 3-fold when compared to a method lacking detergent. However, with respect to catalytic activity, Jak2 isolated from cells using detergent-containing lysis buffers had significantly less catalytic activity than when compared to the method that was detergent free. Expression was then scaled up and Jak2 protein was purified via a one step immunoaffinity purification scheme using both the detergent-free and a modified detergent-containing method of extraction that maintained catalytic activity. In vitro kinase assays demonstrated that the purified product was highly catalytic as measured by its ability to tyrosine phosphorylate Stat1. Collectively, the results show that (1) Jak2 can be expressed at very high levels in mammalian cells, (2) it can be purified to homogeneity via a single step purification scheme, and (3) the purified product is biologically active.
Collapse
Affiliation(s)
- Xianyue Ma
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, P.O. Box 100274, Gainesville, FL 32610, USA
| | | |
Collapse
|
82
|
Stambas J, Brown SA, Gutierrez A, Sealy R, Yue W, Jones B, Lockey TD, Zirkel A, Freiden P, Brown B, Surman S, Coleclough C, Slobod KS, Doherty PC, Hurwitz JL. Long lived multi-isotype anti-HIV antibody responses following a prime-double boost immunization strategy. Vaccine 2005; 23:2454-64. [PMID: 15752831 DOI: 10.1016/j.vaccine.2004.10.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2004] [Revised: 10/10/2004] [Accepted: 10/28/2004] [Indexed: 10/26/2022]
Abstract
Despite decades of work, an effective HIV vaccine remains elusive. In an effort to elicit protective immunity, investigators have sought to define vaccines able to elicit durable HIV-specific B-cell and T-cell activities. Additionally, vaccines are sought which can induce antibodies of a variety of isotypes, as each isotype possesses unique attributes in terms of opsonization, Fc receptor binding capacity, complement fixation and location. One prominent new vaccine strategy, applied to numerous distinct antigenic systems is the prime boost-regimen, with DNA, vaccinia virus (VV), and/or purified recombinant protein. To examine the durability, location and isotype distribution of responses induced by prime-boost regimens, we tested successive immunizations with DNA, VV and protein (D-V-P), comparing three forms of protein inoculations: (i) purified protein administered intramuscularly with complete Freunds adjuvant, (ii) purified protein administered intranasally, and (iii) purified protein conjugated to oxidized mannan, administered intranasally. We found that all three protocols elicited serum antibodies of multiple isotypes, with serum IgA being most prominent among mice immunized with mannan-conjugated protein. All D-V-P protocols, regardless of protein form or route, also elicited antibody responses at mucosal surfaces. In bronchoalveolar lavage, a tendency toward IgA production was again most prominent in mice boosted with the protein-mannan conjugate. Both B-cell and T-cell responses were sustained for more than 1 year post-immunization following each form of vaccination. Contemporaneous with long-lasting serum and mucosal antibodies were antibody forming cells in the bone marrow of primed animals. Results highlight the D-V-P vaccination strategy as a promising approach for attaining durable, multi-isotype B-cell and T-cell activities toward HIV.
Collapse
Affiliation(s)
- J Stambas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Abstract
Gene therapy is a promising approach, yet so far it has shown limited effectiveness in many clinical trials, mainly due to insufficient gene transduction. Recombinant vaccinia virus (rVV) has been well developed as a gene delivery vector, initially for protein expression in mammalian cells. rVV has been further developed to express antigens in vivo in generating immunity for protection against specific infectious diseases and cancer. rVVs, as non-replicating viral vectors, have been demonstrated for their great potential as vaccines, for their diminished cytopathic effects, high levels of protein expression and strong immunogenicity, and they are relatively safe in animals and in human patients. A number of clinical trials using rVVs as vaccines have shown promising results for treating infectious diseases and cancer. In the last few years, due to its exceptional ability to replicate in tumour cells, the Western Reserve strain vaccinia has been explored as a replicating oncolytic virus for cancer virotherapy. As more is learned about the functions of viral gene products in controlling the mammalian cell cycle and in disabling cellular defence mechanisms, specific viral functions can be augmented or eliminated to enhance antitumour efficacy and improve tumour cell targeting. General mechanisms by which this oncolytic virus achieves the antitumour efficacy and specificity are reviewed. Specifically, the deletion of the viral genes for thymidine kinase and vaccinia growth factor resulted in a vaccinia mutant with enhanced tumour targeting activity and fully retaining its efficiency of replication in cancer cells. Other potential strategies for improving this vector for gene delivery will also be discussed in this review.
Collapse
Affiliation(s)
- Z Sheng Guo
- University of Pittsburgh, Division of Surgical Oncology, 5150 Center Avenue, Suite 459, Pittsburgh, PA 15232, USA
| | | |
Collapse
|
84
|
Bleckwenn NA, Golding H, Bentley WE, Shiloach J. Production of recombinant proteins by vaccinia virus in a microcarrier based mammalian cell perfusion bioreactor. Biotechnol Bioeng 2005; 90:663-74. [PMID: 15858791 DOI: 10.1002/bit.20423] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The HeLa cell-vaccinia virus expression system was evaluated for the production of recombinant proteins (enhanced green fluorescent protein (EGFP) and HIV envelope coat protein, gp120) using microcarriers in 1.5 L perfused bioreactor cultures. Perfusion was achieved by use of an alternating tangential flow device (ATF), increasing the length of the exponential phase by 50 h compared to batch culture and increasing the maximum cell density from 1.5x10(6) to 4.4x10(6) cell/mL. A seed train expansion method using cells harvested from microcarrier culture and reseeding onto fresh carriers was developed. EGFP was first used as a model protein to study process parameters affecting protein yield, specifically dissolved oxygen (DO) and temperature during the production phase. The highest level of EGFP, 12+/-1.5 microg/10(6) infected cells, was obtained at 50% DO and 31 degrees C. These setpoints were then used to produce glycoprotein, gp120, which was purified and deglycosylated, revealing a significant amount of N-linked glycosylation. Also, biological activity was assayed, resulting in an ID50 of 3.1 microg/mL, which is comparable to previous reports.
Collapse
Affiliation(s)
- Nicole A Bleckwenn
- Biotechnology Unit, NIDDK, National Institutes of Health, DHHS, Bldg. 14A Rm. 173, MSC 5522, 9000 Rockville Pike, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
85
|
Flanagan K, Glover RT, Hörig H, Yang W, Kaufman HL. Local delivery of recombinant vaccinia virus expressing secondary lymphoid chemokine (SLC) results in a CD4 T-cell dependent antitumor response. Vaccine 2004; 22:2894-903. [PMID: 15246626 DOI: 10.1016/j.vaccine.2003.12.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2003] [Revised: 12/14/2003] [Accepted: 12/14/2003] [Indexed: 10/26/2022]
Abstract
Secondary lymphoid chemokine (SLC) attracts mature dendritic cells (DCs) and naïve T cells. Co-localization of these cells within local tumor environments may enhance the induction of tumor-specific T cells. However, the presence of danger signals or other DC maturation signals are required to optimize T-cell priming. We hypothesized that expression of SLC in vaccinia virus would provide local chemokine delivery and adjuvant factors. A recombinant vaccinia virus expressing murine SLC (rVmSLC) was constructed and characterized. SLC expression was confirmed by Western blot analysis and functional activity was determined by in vitro chemotaxis assay. Supernatants from rVmSLC-infected cells attracted CD4 T cells, and also induced the migration of CD8 T cells and DCs. Although poxviruses are known to express several chemokine-binding proteins, systemic injection of rVmSLC was well tolerated in mice up to a dose of 1 x 10(7) pfu and did not significantly alter vaccinia-specific T-cell immunity. Local injection of rVmSLC into established tumors derived from the murine colon cancer line, CT26, resulted in enhanced infiltration of CD4 T cells, which correlated with inhibition of tumor growth. The central role of CD4 T cells was further demonstrated by loss of anti-tumor activity in CD4 T-cell depleted mice. Intratumoral delivery of SLC using a poxviral vaccine extends the use of SLC in anti-tumor therapies and may present an effective alternative for improving the immunotherapy of cancer alone or in combination with other anti-tumor agents for clinical therapy.
Collapse
MESH Headings
- Animals
- Blotting, Southern
- Blotting, Western
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line
- Chemokine CCL21
- Chemokines, CC/biosynthesis
- Chemokines, CC/genetics
- Chemotaxis, Leukocyte
- Colonic Neoplasms/immunology
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Female
- Flow Cytometry
- Immunohistochemistry
- Mice
- Mice, Inbred BALB C
- Microinjections
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
- Vaccinia virus/genetics
- Vaccinia virus/immunology
- Viral Vaccines/adverse effects
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Kenneth Flanagan
- Department of Pathology, Columbia University, 177 Fort Washington Avenue, MHB 7-SK, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
86
|
Gómez CE, Abaitua F, Rodríguez D, Esteban M. Efficient CD8+ T cell response to the HIV-env V3 loop epitope from multiple virus isolates by a DNA prime/vaccinia virus boost (rWR and rMVA strains) immunization regime and enhancement by the cytokine IFN-γ. Virus Res 2004; 105:11-22. [PMID: 15325077 DOI: 10.1016/j.virusres.2004.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2004] [Revised: 03/30/2004] [Accepted: 04/02/2004] [Indexed: 12/16/2022]
Abstract
The cytotoxic T-lymphocyte response (CTL) has been shown to be determinant in the clearance of many viral infections and hence, vaccine candidates against AIDS are designed to enhance this arm of the immune system. In this study, we have analyzed the antigen specific immune responses triggered in mice by different combinations of vaccine vehicles expressing the multiepitope polypeptide TAB13. This chimeric protein contains the V3 region of the gp120 from eight different HIV-1 isolates and was efficiently expressed by a DNA vector (DNA-TAB), and also by vaccinia virus recombinants (rVV) based either on the attenuated modified vaccinia virus Ankara (MVA-TAB) or Western Reserve (VV-TAB) strains. Inoculation of a DNA-TAB vector in priming followed by a booster with VV-TAB or MVA-TAB induces a humoral immune response against TAB13 protein and efficiently enhanced the CD8+ T cell response against V3 epitopes from HIV-1 isolates LR150, MN, and IIIB in comparison with animals immunized with two doses of DNA-TAB. A protocol that incorporates a DNA vector expressing IFN-gamma (DNA-IFN-gamma) with DNA-TAB in the priming, followed by a booster with MVA-TAB, triggered the highest values of specific CD8+ T cell response. By examining the cytokine pattern, the immune response induced by these vaccination approaches was predominantly of Th-1 type. These findings establish safe strategies for the enhanced generation of T cell mediated immunity to HIV-1 that can benefit in the design of an effective vaccine against AIDS.
Collapse
MESH Headings
- AIDS Vaccines/administration & dosage
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Amino Acid Sequence
- Animals
- Base Sequence
- CD8-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Cricetinae
- Cytokines/analysis
- Epitopes/genetics
- Epitopes/immunology
- Female
- HIV Envelope Protein gp120/genetics
- HIV Envelope Protein gp120/immunology
- HIV-1/immunology
- Immunization, Secondary
- Injections, Intramuscular
- Injections, Intraperitoneal
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Mice
- Mice, Inbred BALB C
- Molecular Sequence Data
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Vaccination/methods
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccinia virus/genetics
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Campus Universidad Autónoma, 28049 Madrid, Spain
| | | | | | | |
Collapse
|
87
|
Belyakov IM, Hammond SA, Ahlers JD, Glenn GM, Berzofsky JA. Transcutaneous immunization induces mucosal CTLs and protective immunity by migration of primed skin dendritic cells. J Clin Invest 2004; 113:998-1007. [PMID: 15057306 PMCID: PMC379323 DOI: 10.1172/jci20261] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2003] [Accepted: 01/13/2004] [Indexed: 12/16/2022] Open
Abstract
Transcutaneous immunization (TCI), the application of vaccines on the skin, induces robust systemic and mucosal antibodies in animal models and in humans. The means by which mucosal immune responses to vaccine antigens are elicited by TCI has not been well characterized. We examined the effect of TCI with an HIV peptide vaccine on the induction of mucosal and systemic CTL responses and protective immunity against mucosal challenge with live virus in mice. Robust HIV-specific CTL responses in the spleen and in the gut mucosa were detected after TCI. The responses were dependent upon the addition of an adjuvant and resulted in protection against mucosal challenge with recombinant vaccinia virus encoding HIV gp160. Although it is clear that adjuvant-activated DCs migrated mainly to draining lymph nodes, coculture with specific T cells and flow cytometry studies with DCs isolated from Peyer's patches after TCI suggested that activated DCs carrying skin-derived antigen also migrated from the skin to immune-inductive sites in gut mucosa and presented antigen directly to resident lymphocytes. These results and previous clinical trial results support the observation that TCI is a safe and effective strategy for inducing strong mucosal antibody and CTL responses.
Collapse
Affiliation(s)
- Igor M Belyakov
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1578, USA.
| | | | | | | | | |
Collapse
|
88
|
Hu Y, Riesland L, Paterson AJ, Kudlow JE. Phosphorylation of mouse glutamine-fructose-6-phosphate amidotransferase 2 (GFAT2) by cAMP-dependent protein kinase increases the enzyme activity. J Biol Chem 2004; 279:29988-93. [PMID: 15133036 DOI: 10.1074/jbc.m401547200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A protein encoded by a new gene with approximately 75% homology to glutamine-fructose-6-phosphate amidotransferase (GFAT) was termed GFAT2 on the basis of this similarity. The mouse GFAT2 cDNA was cloned, and the protein was expressed with either an N-terminal glutathione S-transferase or His tag. The purified protein expressed in mammalian cells had GFAT activity. The Km values for the two substrates of reaction, fructose 6-phosphate and glutamine, were determined to be 0.8 mm for fructose 6-phosphate and 1.2 mm for glutamine, which are within the ranges determined for GFAT1. The protein sequence around the serine 202 of GFAT2 was conserved to the serine 205 of GFAT1, whereas the serine at 235 in GFAT1 was not present in GFAT2. Previously we showed that phosphorylation of serine 205 in GFAT1 by the catalytic subunit of cAMP-dependent protein kinase (PKA) inhibits its activity. Like GFAT1, GFAT2 was phosphorylated by PKA, but GFAT2 activity increased approximately 2.2-fold by this modification. When serine 202 of GFAT2 was mutated to an alanine, the enzyme not only became resistant to phosphorylation, but also the increase in activity in response to PKA also was blocked. These results indicated that the phosphorylation of serine 202 was necessary and sufficient for these alterations by PKA. GFAT2 was modestly inhibited (15%) by UDP-GlcNAc but not through detectable O-glycosylation. GFAT2 is, therefore, an isoenzyme of GFAT1, but its regulation by cAMP is the opposite, allowing differential regulation of the hexosamine pathway in specialized tissues.
Collapse
Affiliation(s)
- Yong Hu
- Department of Cell Biology, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | |
Collapse
|
89
|
Di Nicola M, Carlo-Stella C, Anichini A, Mortarini R, Guidetti A, Tragni G, Gallino F, Del Vecchio M, Ravagnani F, Morelli D, Chaplin P, Arndtz N, Sutter G, Drexler I, Parmiani G, Cascinelli N, Gianni AM. Clinical protocol. Immunization of patients with malignant melanoma with autologous CD34(+) cell-derived dendritic cells transduced ex vivo with a recombinant replication-deficient vaccinia vector encoding the human tyrosinase gene: a phase I trial. Hum Gene Ther 2004; 14:1347-60. [PMID: 14503969 DOI: 10.1089/104303403322319426] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Massimo Di Nicola
- Cristina Gandini Bone Marrow Transplantation Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan I-20133, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Heinemann L, Dillon S, Crawford A, Bäckström BT, Hibma MH. Flow cytometric quantitation of the protective efficacy of dendritic cell based vaccines in a human papillomavirus type 16 murine challenge model. J Virol Methods 2004; 117:9-18. [PMID: 15019255 DOI: 10.1016/j.jviromet.2003.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2003] [Revised: 11/18/2003] [Accepted: 11/20/2003] [Indexed: 11/23/2022]
Abstract
A murine model for the assessment of protective immunity to human papillomavirus (HPV) type 16, a virus that does not naturally infect mice, is described. In this system, protection was tested following intranasal challenge of mice with a recombinant vaccinia virus expressing both the selected HPV antigen and a beta-galactosidase (beta-gal) reporter. The extent of viral infectivity was determined by measuring beta-gal positive lung cells using flow cytometry. The efficacy of this model to measure protective immunity was evaluated by priming mice with the beta-gal vaccinia virus then challenging the mice with the same virus. Vaccinia primed mice had negligible numbers of beta-gal positive cells in the lung 5 days following viral challenge indicating protection, whereas around 50% of cells were infected in immunologically naive, challenged mice. The protective efficacy of two dendritic cell vaccines for HPV16 was measured in this model. Both vaccines provided some protection to subsequent viral challenge, compared with their controls. Although this protection model was applied to HPV in this study, it may also have broad application to other viruses that do not infect mice naturally.
Collapse
Affiliation(s)
- Lucy Heinemann
- Virus Research Unit, Department of Microbiology, University of Otago, P.O. Box 56, Dunedin 9001, New Zealand
| | | | | | | | | |
Collapse
|
91
|
Li Z, Rubin SA, Taffs RE, Merchlinsky M, Ye Z, Carbone KM. Mouse neurotoxicity test for vaccinia-based smallpox vaccines. Vaccine 2004; 22:1486-93. [PMID: 15063573 DOI: 10.1016/j.vaccine.2003.10.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2003] [Revised: 10/13/2003] [Accepted: 10/14/2003] [Indexed: 10/26/2022]
Abstract
The only US FDA licensed smallpox vaccine, Dryvax, was associated with rare but serious neurological adverse events. After smallpox was eradicated in the United States, mass vaccination ceased in 1971. As counter-bioterrorism/biowarfare measures, new smallpox vaccines are now being investigated. However, there are no established pre-clinical neurotoxicity assays with which to evaluate these new vaccines prior to licensure. Here we report the development and initial characterization of a small animal neurotoxicity assay for vaccinia-based smallpox vaccines using Dryvax virus as a reference vaccine strain and the neuroadapted Western Reserve (WR) strain as a neurotoxic positive control. In neonatally inoculated mice, the WR strain produced significantly greater and more rapid onset of mortality than the Dryvax vaccine reference. Expression of virus antigen in neural cells and infectious virus replication in the brain was also significantly different between the two strains. In addition, the appearance of high titer virus antibody correlated with the clearance of virus from brain. With further validation, this assay incorporating a licensed vaccine reference standard and positive control strain may provide important pre-clinical neurotoxicity data on new vaccinia-based smallpox vaccine strains.
Collapse
Affiliation(s)
- Zhongqi Li
- Laboratory of Pediatric and Respiratory Viral Diseases, OD/Center for Biologics Evaluation and Research, Food and Drug Administration, HFM-460, Bldg. 29B, Room 5NN22, 8800 Rockville Pike, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
92
|
Zhang F, Su K, Yang X, Bowe DB, Paterson AJ, Kudlow JE. O-GlcNAc modification is an endogenous inhibitor of the proteasome. Cell 2004; 115:715-25. [PMID: 14675536 DOI: 10.1016/s0092-8674(03)00974-7] [Citation(s) in RCA: 335] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The ubiquitin proteasome system classically selects its substrates for degradation by tagging them with ubiquitin. Here, we describe another means of controlling proteasome function in a global manner. The 26S proteasome can be inhibited by modification with the enzyme, O-GlcNAc transferase (OGT). This reversible modification of the proteasome inhibits the proteolysis of the transcription factor Sp1 and a hydrophobic peptide through inhibition of the ATPase activity of 26S proteasomes. The Rpt2 ATPase in the mammalian proteasome 19S cap is modified by O-GlcNAc in vitro and in vivo and as its modification increases, proteasome function decreases. This mechanism may couple proteasomes to the general metabolic state of the cell. The O-GlcNAc modification of proteasomes may allow the organism to respond to its metabolic needs by controlling the availability of amino acids and regulatory proteins.
Collapse
Affiliation(s)
- Fengxue Zhang
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
93
|
Breckpot K, Heirman C, Neyns B, Thielemans K. Exploiting dendritic cells for cancer immunotherapy: genetic modification of dendritic cells. J Gene Med 2004; 6:1175-88. [PMID: 15468193 DOI: 10.1002/jgm.615] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs) are pivotal regulators of immune reactivity and immune tolerance. The observation that DCs can recruit naive T cells has invigorated cancer immunology and led to the proposal of DCs as the basis for vaccines designed for the treatment of cancer. Designing effective strategies to load DCs with antigens is a challenging field of research. The successful realization of gene transfer to DCs will be highly dependent on the employed vector system. Here, we review various viral and non-viral gene transfer systems, and discuss their distinct characteristics and possible advantages and disadvantages in respect to their use in DC-based immunotherapy.
Collapse
Affiliation(s)
- Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | | | | | | |
Collapse
|
94
|
Shalaby KA, Yin L, Thakur A, Christen L, Niles EG, LoVerde PT. Protection against Schistosoma mansoni utilizing DNA vaccination with genes encoding Cu/Zn cytosolic superoxide dismutase, signal peptide-containing superoxide dismutase and glutathione peroxidase enzymes. Vaccine 2003; 22:130-6. [PMID: 14604580 DOI: 10.1016/s0264-410x(03)00535-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Protection against Schistosoma mansoni infection in C57BL/6 female mice was evaluated by two DNA vaccination strategies. Mice were either vaccinated by intramuscular injection with pcDNAI/Amp constructs encoding either Cu/Zn cytosolic superoxide dismutase (CT-SOD), signal peptide-containing SOD (SP-SOD), glutathione peroxidase (GPX(bb)) or a mutated form of GPX (GPX(m)), or primed with naked DNA constructs and boosted with recombinant vaccinia virus (RVV) containing the same genes. Animals were then challenged with S. mansoni and the level of protection was assessed as the reduction in worm burden. CT-SOD showed significant levels of protection compared to the control group, ranging between 44 and 60%, while SP-SOD exhibited from 22 to 45%. GPX(bb) showed levels of protection (23-55%) higher than GPX(m) (25-34%). The prime-boost strategy gave the same results as naked DNA or recombinant vaccinia virus alone except in the case of GPX, where the protection was 85%.
Collapse
Affiliation(s)
- Kamal A Shalaby
- Department of Microbiology, and Immunology, School of Medicine and Biomedical Sciences, State University of New York, 138 Farber Hall, Buffalo, NY 14214, USA
| | | | | | | | | | | |
Collapse
|
95
|
Park SH, Yang SH, Lee CG, Youn JW, Chang J, Sung YC. Efficient induction of T helper 1 CD4+ T-cell responses to hepatitis C virus core and E2 by a DNA prime-adenovirus boost. Vaccine 2003; 21:4555-64. [PMID: 14575768 DOI: 10.1016/s0264-410x(03)00499-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hepatitis C virus (HCV) is an important causative agent of liver disease, but currently there is no available prophylactic vaccine against HCV infection. Here, we investigated the HCV E2- and core-specific T-cell responses induced by DNA (D) and/or recombinant adenovirus (A) vaccines. In single (D versus A) or double immunizations (D-D versus A-A), the recombinant adenovirus vaccines induced higher levels of IFN-gamma secreting T-cell response and cytotoxic T lymphocytes (CTL) response than the DNA vaccines. However, a heterologous (D-A) regimen elicited the highest level of T helper 1 (Th1) CD4(+) T-cell responses. Furthermore, three E2-specific CTL epitopes were mapped using a peptide pool spanning the E2 protein sequence (a.a. 384-713) in BALB/c mice, and one of these (E2 405-414: SGPSQKIQLV) was shown to be immunodominant. Interestingly, no significant differences were found in the repertoire of E2-specific T-cell responses or in the immunodominance hierarchy of the three epitopes induced by D-D, D-A, A-A, and A-D, indicating that the breadth and hierarchy of T-cell responses is independent of these different vaccination regimens. In conclusion, the heterologous DNA prime-recombinant adenovirus boost regimen described offers an efficient promising strategy for the development of an effective T-cell-based HCV vaccine.
Collapse
Affiliation(s)
- Su-Hyung Park
- National Research Laboratory of DNA Medicine, Division of Molecular and Life Science, Pohang University of Science and Technology, San 31, Hyoja-dong, Nam-gu, Pohang, Kyungbuk 790-784, South Korea
| | | | | | | | | | | |
Collapse
|
96
|
Malkin AJ, McPherson A, Gershon PD. Structure of intracellular mature vaccinia virus visualized by in situ atomic force microscopy. J Virol 2003; 77:6332-40. [PMID: 12743290 PMCID: PMC155008 DOI: 10.1128/jvi.77.11.6332-6340.2003] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2002] [Accepted: 03/04/2003] [Indexed: 11/20/2022] Open
Abstract
Vaccinia virus, the basis of the smallpox vaccine, is one of the largest viruses to replicate in humans. We have used in situ atomic force microscopy (AFM) to directly visualize fully hydrated, intact intracellular mature vaccinia virus (IMV) virions and chemical and enzymatic treatment products thereof. The latter included virion cores, core-enveloping coats, and core substructures. The isolated coats appeared to be composed of a highly cross-linked protein array. AFM imaging of core substructures indicated association of the linear viral DNA genome with a segmented protein sheath forming an extended approximately 16-nm-diameter filament with helical surface topography; enclosure of this filament within a 30- to 40-nm-diameter tubule which also shows helical topography; and enclosure of the folded, condensed 30- to 40-nm-diameter tubule within the core by a wall covered with peg-like projections. Proteins observed attached to the 30- to 40-nm-diameter tubules may mediate folding and/or compaction of the tubules and/or represent vestiges of the core wall and/or pegs. An accessory "satellite domain" was observed protruding from the intact core. This corresponded in size to isolated 70- to 100-nm-diameter particles that were imaged independently and might represent detached accessory domains. AFM imaging of intact virions indicated that IMV underwent a reversible shrinkage upon dehydration (as much as 2.2- to 2.5-fold in the height dimension), accompanied by topological and topographical changes, including protrusion of the satellite domain. As shown here, the chemical and enzymatic dissection of large, asymmetrical virus particles in combination with in situ AFM provides an informative complement to other structure determination techniques.
Collapse
Affiliation(s)
- A J Malkin
- BioSecurity and NanoSciences Laboratory, Department of Chemistry and Materials Science, Lawrence Livermore National Laboratory, California 94551, USA.
| | | | | |
Collapse
|
97
|
Abstract
Prostate cancer is the second leading cause of cancer death in the US, largely because of the limitations of our current therapeutic options, especially once the cancer has metastasized. Investigators have long sought new therapeutic modalities such as angiogenesis inhibitors, vaccines, and gene therapy, among others. It appears that a combination approach will be required to cure the majority of malignancies. Immunotherapy for prostate cancer appears feasible and a likely therapeutic modality in the armamentarium. Unfortunately, further research in basic immunology and the interaction of the immune system with other forms of therapy is needed. Many obstacles exist in immunotherapy, including vector design, tumouricidal specificity, and tumor evasion, which will have to be overcome in order to realize the maximum therapeutic benefit from this treatment modality.
Collapse
Affiliation(s)
- Joseph M Kaminski
- Department of Radiology, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|
98
|
Zhong B, Jiang K, Gilvary DL, Epling-Burnette PK, Ritchey C, Liu J, Jackson RJ, Hong-Geller E, Wei S. Human neutrophils utilize a Rac/Cdc42-dependent MAPK pathway to direct intracellular granule mobilization toward ingested microbial pathogens. Blood 2003; 101:3240-8. [PMID: 12511425 DOI: 10.1182/blood-2001-12-0180] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Elevated levels of mitogen-activated protein kinase/extracellular regulatory kinase (MAPK/ERK) activity are frequently found in some cancer cells. In efforts to reduce tumor growth, attempts have been made to develop cancer therapeutic agents targeting the MAPK. Here, by use of biologic, biochemical, and gene manipulation methods in human polymorphonuclear neutrophils (PMNs), we have identified a key pathway important in normal cell function involving MAPK/ERK in PMNs for growth inhibition of Candida albicans. Contact with C albicans triggered MAPK/ERK activation in PMNs within 5 minutes, and blocking of MAPK/ERK activation, either by the pharmacologic reagent PD098059 or by dominant-negative MAPK kinase (MEK) expression via vaccinia viral delivery, suppressed antimicrobial activity. Rac and Cdc42, but not Ras or Rho, were responsible for this MAPK/ERK activation. Expression of dominant-negative Rac (N17Rac) or Cdc42 (N17Cdc42) eliminated not only C albicans- mediated ERK phosphorylation but also phagocytosis and granule migration toward the ingested microbes, whereas dominant-negative Ras (N17Ras) and Rho (N19Rho) did not. PAK1 (p21-activated kinase 1) activation is induced by C albicans, suggesting that PAK1 may also be involved in the Rac1 activation of MAPK/ERK. We conclude from these data that Rac/Cdc42-dependent activation of MAPK/ERK is a critical event in the immediate phagocytic response of PMNs to microbial challenge. Therefore, use of MAPK pharmacologic inhibitors for the treatment of cancer may result in the interruption of normal neutrophil function. A balance between therapeutic outcome and undesirable side effects must be attained to achieve successful and safe anticancer therapy.
Collapse
Affiliation(s)
- Bin Zhong
- Immunology Program, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida College of Medicine, Department of Interdisciplinary Oncology, Tampa, 33612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Abstract
The incidence of malignant melanoma is increasing faster than that of any other malignancy in the United States, and therefore this disease represents a significant health threat now and in the future. The impact of conventional systemic therapy for metastatic melanoma is minimal, with best response rates for conventional therapy nearing only 30% and cure rates well below 10%. This justifies the development of immunotherapy as a new treatment modality for patients with melanoma. This review summarizes the most recent findings and the newest approaches in the design of vaccines for melanoma. The antigens associated with melanoma and their uses in the vaccine development are described. Possible clinical applications of the new vaccines for melanoma and future directions for their development are also discussed.
Collapse
Affiliation(s)
- Boris R Minev
- Cancer Center, University of California-San Diego, La Jolla, CA 92093-0063, USA
| |
Collapse
|
100
|
Kidokoro M, Aoki A, Horiuchi K, Shida H. Large-scale preparation of biologically active measles virus haemagglutinin expressed by attenuated vaccinia virus vectors. Microbes Infect 2002; 4:1035-44. [PMID: 12191653 DOI: 10.1016/s1286-4579(02)01627-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
A procedure described here allows the efficient and rapid purification of histidine-tagged measles virus haemagglutinin that is synthesized under the control of powerful promoters (PSFJ1-10 and PSFJ2-16) of the highly attenuated vaccinia virus (VV) strain LC16mO. A single affinity chromatography step purifies recombinant haemagglutinin proteins from the lysates of cells infected with the recombinant VVs. The recovery and purity are both very high (a yield of 0.5-2.8 mg/10(8) cells and purity of >94-98%), indicating that this procedure is approximately 400 times more efficient than the conventional methods used to prepare haemagglutinin. The haemagglutinins are correctly transported to the cell surface and have haemadsorption activity. Moreover, the recombinant haemagglutinin proteins cooperate with the measles virus fusion protein to elicit cell fusion activity. In addition, the antibody titres against measles virus, as measured by enzyme-linked immunosorbent assay using the purified haemagglutinin as the capture antigen, correlated closely with neutralization test titres (R(2) = 0.84, p < 0.05), indicating the preservation of immunologically relevant antigenicity. Such recombinant haemagglutinin preparations will be useful in diagnostic tests that measure functional anti-measles immunity and investigate the biological functions and structure of the haemagglutinin.
Collapse
Affiliation(s)
- Minoru Kidokoro
- Department of Research and Development, Chiba Serum Institute, 2-6-1 Kohnodai, Ichikawa, Chiba 272-0827, Japan.
| | | | | | | |
Collapse
|