1
|
Kanai Y, Kawagishi T, Okamoto M, Sakai Y, Matsuura Y, Kobayashi T. Lethal murine infection model for human respiratory disease-associated Pteropine orthoreovirus. Virology 2018; 514:57-65. [PMID: 29128757 PMCID: PMC7173163 DOI: 10.1016/j.virol.2017.10.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 12/24/2022]
Abstract
Pteropine orthoreovirus (PRV) is an emerging bat-borne human pathogen causing severe respiratory illness. To date, however, the evaluation of PRV virulence has largely depended on the limited numbers of clinical cases owing to the lack of animal models. To develop an in vivo model of PRV infection, an inbred C3H mouse strain was infected intranasally with pathogenic PRV strain Miyazaki-Bali/2007. C3H mice suffered severe lung infection with significant body weight reduction and died within 7 days after intranasal infection. Infectious viruses were isolated mainly from the lungs and trachea. Histopathological examination revealed interstitial pneumonia with monocytes infiltration. Following repeated intranasal infection, mice developed antibodies to particular structural and non-structural proteins of PRV. The results of these immunological assays will help to develop laboratory protocols for sero-epidemiological studies. Our small rodent model of lethal respiratory infection will further allow investigation of the molecular mechanisms underlying the high pathogenicity of PRV. A lethal PRV strain Miyazaki-Bali/2007 murine infection model was established. Susceptibility of different mouse strains to PRV infection was investigated. Antibody responses to PRV proteins in C3H mice post intranasal infection were studied.
Collapse
Affiliation(s)
- Yuta Kanai
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takahiro Kawagishi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Minoru Okamoto
- Department of Veterinary Pathology, Rakuno Gakuen University, Hokkaido, Japan
| | - Yusuke Sakai
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yoshiharu Matsuura
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takeshi Kobayashi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
| |
Collapse
|
2
|
Martin-Murphy BV, You Q, Wang H, De La Houssaye BA, Reilly TP, Friedman JE, Ju C. Mice lacking natural killer T cells are more susceptible to metabolic alterations following high fat diet feeding. PLoS One 2014; 9:e80949. [PMID: 24465369 PMCID: PMC3896335 DOI: 10.1371/journal.pone.0080949] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 10/14/2013] [Indexed: 12/12/2022] Open
Abstract
Current estimates suggest that over one-third of the adult population has metabolic syndrome and three-fourths of the obese population has non-alcoholic fatty liver disease (NAFLD). Inflammation in metabolic tissues has emerged as a universal feature of obesity and its co-morbidities, including NAFLD. Natural Killer T (NKT) cells are a subset of innate immune cells that abundantly reside within the liver and are readily activated by lipid antigens. There is general consensus that NKT cells are pivotal regulators of inflammation; however, disagreement exists as to whether NKT cells exert pathogenic or suppressive functions in obesity. Here we demonstrate that CD1d−/− mice, which lack NKT cells, were more susceptible to weight gain and fatty liver following high fat diet (HFD) feeding. Compared with their WT counterparts, CD1d−/− mice displayed increased adiposity and greater induction of inflammatory genes in the liver suggestive of the precursors of NAFLD. Calorimetry studies revealed a significant increase in food intake and trends toward decreased metabolic rate and activity in CD1d−/− mice compared with WT mice. Based on these findings, our results suggest that NKT cells play a regulatory role that helps to prevent diet-induced obesity and metabolic dysfunction and may play an important role in mechanisms governing cross-talk between metabolism and the immune system to regulate energy balance and liver health.
Collapse
Affiliation(s)
- Brittany V. Martin-Murphy
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Qiang You
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Hong Wang
- Division of Endocrinology, Diabetes & Metabolism, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Becky A. De La Houssaye
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Timothy P. Reilly
- Drug Safety Evaluation, Research & Development, Bristol-Myers Squibb Company, Princeton, New Jersey, United States of America
| | - Jacob E. Friedman
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Cynthia Ju
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
3
|
Virus-heat shock protein interaction and a novel axis for innate antiviral immunity. Cells 2012; 1:646-66. [PMID: 24710494 PMCID: PMC3901102 DOI: 10.3390/cells1030646] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 08/24/2012] [Accepted: 08/27/2012] [Indexed: 12/20/2022] Open
Abstract
Virus infections induce heat shock proteins that in turn enhance virus gene expression, a phenomenon that is particularly well characterized for the major inducible 70 kDa heat shock protein (hsp70). However, hsp70 is also readily induced by fever, a phylogenetically conserved response to microbial infections, and when released from cells, hsp70 can stimulate innate immune responses through toll like receptors 2 and 4 (TLR2 and 4). This review examines how the virus-hsp70 relationship can lead to host protective innate antiviral immunity, and the importance of hsp70 dependent stimulation of virus gene expression in this host response. Beginning with the well-characterized measles virus-hsp70 relationship and the mouse model of neuronal infection in brain, we examine data indicating that the innate immune response is not driven by intracellular sensors of pathogen associated molecular patterns, but rather by extracellular ligands signaling through TLR2 and 4. Specifically, we address the relationship between virus gene expression, extracellular release of hsp70 (as a damage associated molecular pattern), and hsp70-mediated induction of antigen presentation and type 1 interferons in uninfected macrophages as a novel axis of antiviral immunity. New data are discussed that examines the more broad relevance of this protective mechanism using vesicular stomatitis virus, and a review of the literature is presented that supports the probable relevance to both RNA and DNA viruses and for infections both within and outside of the central nervous system.
Collapse
|
4
|
Foxp3+ regulatory T cells control persistence of viral CNS infection. PLoS One 2012; 7:e33989. [PMID: 22448284 PMCID: PMC3309005 DOI: 10.1371/journal.pone.0033989] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 02/22/2012] [Indexed: 11/19/2022] Open
Abstract
We earlier established a model of a persistent viral CNS infection using two week old immunologically normal (genetically unmodified) mice and recombinant measles virus (MV). Using this model infection we investigated the role of regulatory T cells (Tregs) as regulators of the immune response in the brain, and assessed whether the persistent CNS infection can be modulated by manipulation of Tregs in the periphery. CD4+ CD25+ Foxp3+ Tregs were expanded or depleted during the persistent phase of the CNS infection, and the consequences for the virus-specific immune response and the extent of persistent infection were analyzed. Virus-specific CD8+ T cells predominantly recognising the H-2Db-presented viral hemagglutinin epitope MV-H22–30 (RIVINREHL) were quantified in the brain by pentamer staining. Expansion of Tregs after intraperitoneal (i.p.) application of the superagonistic anti-CD28 antibody D665 inducing transient immunosuppression caused increased virus replication and spread in the CNS. In contrast, depletion of Tregs using diphtheria toxin (DT) in DEREG (depletion of regulatory T cells)-mice induced an increase of virus-specific CD8+ effector T cells in the brain and caused a reduction of the persistent infection. These data indicate that manipulation of Tregs in the periphery can be utilized to regulate virus persistence in the CNS.
Collapse
|
5
|
Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, Bettini ML, Gravano DM, Vogel P, Liu CL, Tangsombatvisit S, Grosso JF, Netto G, Smeltzer MP, Chaux A, Utz PJ, Workman CJ, Pardoll DM, Korman AJ, Drake CG, Vignali DAA. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res 2011; 72:917-27. [PMID: 22186141 DOI: 10.1158/0008-5472.can-11-1620] [Citation(s) in RCA: 1268] [Impact Index Per Article: 90.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inhibitory receptors on immune cells are pivotal regulators of immune escape in cancer. Among these inhibitory receptors, CTLA-4 (targeted clinically by ipilimumab) serves as a dominant off-switch while other receptors such as PD-1 and LAG-3 seem to serve more subtle rheostat functions. However, the extent of synergy and cooperative interactions between inhibitory pathways in cancer remain largely unexplored. Here, we reveal extensive coexpression of PD-1 and LAG-3 on tumor-infiltrating CD4(+) and CD8(+) T cells in three distinct transplantable tumors. Dual anti-LAG-3/anti-PD-1 antibody treatment cured most mice of established tumors that were largely resistant to single antibody treatment. Despite minimal immunopathologic sequelae in PD-1 and LAG-3 single knockout mice, dual knockout mice abrogated self-tolerance with resultant autoimmune infiltrates in multiple organs, leading to eventual lethality. However, Lag3(-/-)Pdcd1(-/-) mice showed markedly increased survival from and clearance of multiple transplantable tumors. Together, these results define a strong synergy between the PD-1 and LAG-3 inhibitory pathways in tolerance to both self and tumor antigens. In addition, they argue strongly that dual blockade of these molecules represents a promising combinatorial strategy for cancer.
Collapse
Affiliation(s)
- Seng-Ryong Woo
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Lai D, Zhu J, Wang T, Hu-Li J, Terabe M, Berzofsky JA, Clayberger C, Krensky AM. KLF13 sustains thymic memory-like CD8(+) T cells in BALB/c mice by regulating IL-4-generating invariant natural killer T cells. J Exp Med 2011; 208:1093-103. [PMID: 21482696 PMCID: PMC3092346 DOI: 10.1084/jem.20101527] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 03/09/2011] [Indexed: 11/07/2022] Open
Abstract
"Memory-like T cells" are a subset of thymic cells that acquire effector function through the maturation process rather than interaction with specific antigen. Disruption of genes encoding T cell signaling proteins or transcription factors have provided insights into the differentiation of such cells. In this study, we show that in BALB/c, but not C57BL/6, mice, a large portion of thymic CD4(-)CD8(+) T cells exhibit a memory-like phenotype. In BALB/c mice, IL-4 secreted by invariant natural killer T (iNKT) cells is both essential and sufficient for the generation of memory-like T cells. In C57BL/6 mice, iNKT cells are less abundant, producing IL-4 that is insufficient to induce thymic memory-like CD8(+) T cells. BALB/c mice deficient in the transcription factor Kruppel-like factor (KLF) 13 have comparable numbers of iNKT cells to C57BL/6 mice and extremely low levels of thymic memory-like CD8(+) T cells. This work documents the impact of a small number of KLF13-dependent iNKT cells on the generation of memory-like CD8(+) T cells.
Collapse
Affiliation(s)
- Dazhi Lai
- Laboratory of Cellular and Molecular Biology and Vaccine Branch, National Cancer Institute, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jinfang Zhu
- Laboratory of Cellular and Molecular Biology and Vaccine Branch, National Cancer Institute, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Tianhong Wang
- Laboratory of Cellular and Molecular Biology and Vaccine Branch, National Cancer Institute, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jane Hu-Li
- Laboratory of Cellular and Molecular Biology and Vaccine Branch, National Cancer Institute, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Masaki Terabe
- Laboratory of Cellular and Molecular Biology and Vaccine Branch, National Cancer Institute, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jay A. Berzofsky
- Laboratory of Cellular and Molecular Biology and Vaccine Branch, National Cancer Institute, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Carol Clayberger
- Laboratory of Cellular and Molecular Biology and Vaccine Branch, National Cancer Institute, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Alan M. Krensky
- Laboratory of Cellular and Molecular Biology and Vaccine Branch, National Cancer Institute, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
7
|
Abstract
Measles virus is highly neuroinvasive, yet host immune responses are highly effective at limiting neurovirulence in humans. We know that neurons are an important target of infection and that both IFN-γ and -β expression are observed in the measles virus-infected human brain. Rodent models can be used to understand how this response is orchestrated. Constitutive expression of the major inducible 70-kDa heat-shock protein is a feature of primate tissues that is lacking in mice. This article examines the importance of addressing this difference when modeling outcomes of brain infection in mice, particularly in terms of understanding how infected neurons may activate uninfected brain macrophages to produce IFN-β and support T-cell production of IFN-γ, a mediator of noncytolytic viral clearance. New and historical data suggest that the virus heat-shock protein 70 relationship is key to a protective host immune response and has potential broad relevance.
Collapse
Affiliation(s)
- Michael Oglesbee
- Department of Veterinary Biosciences, Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| |
Collapse
|
8
|
Measles virus infection of the CNS: human disease, animal models, and approaches to therapy. Med Microbiol Immunol 2010; 199:261-71. [PMID: 20390298 DOI: 10.1007/s00430-010-0153-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Indexed: 01/13/2023]
Abstract
Viral infections of the central nervous system(CNS) mostly represent clinically important, often life-threatening complications of systemic viral infections. After acute measles, CNS complications may occur early (acute postinfectious measles encephalitis, APME) or after years of viral persistence (subacute sclerosing panencephalitis, SSPE). In spite of a presumably functional cell-mediated immunity and high antiviral antibody titers, an immunological control of the CNS infection is not achieved in patients suffering from SSPE. There is still no specific therapy for acute complications and persistent MV infections of the CNS. Hamsters, rats, and (genetically unmodified and modified) mice have been used as model systems to study mechanisms of MV-induced CNS infections. Functional CD4+ and CD8+ T cells together with IFN-gamma are required to overcome the infection. With the help of recombinant measles viruses and mice expressing endogenous or transgenic receptors, interesting aspects such as receptor-dependent viral spread and viral determinants of virulence have been investigated. However, many questions concerning the lack of efficient immune control in the CNS are still open. Recent research opened new perspectives using specific antivirals such as short interfering RNA (siRNA) or small molecule inhibitors. Inspite of obvious hurdles, these treatments are the most promising approaches to future therapies.
Collapse
|
9
|
Major histocompatibility complex haplotype determines hsp70-dependent protection against measles virus neurovirulence. J Virol 2009; 83:5544-55. [PMID: 19321604 DOI: 10.1128/jvi.02673-08] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In vitro studies show that hsp70 promotes gene expression for multiple viral families, although there are few reports on the in vivo significance of virus-hsp70 interaction. Previously we showed that hsp70-dependent stimulation of Edmonston measles virus (Ed MeV) transcription caused an increased cytopathic effect and mortality in transgenic hsp70-overexpressing C57BL/6 mice (H-2(b)). The response to MeV infection is influenced by the major histocompatibility complex haplotype; H-2(d) mice are resistant to brain infection due to robust antiviral immune responses, whereas H-2(b) mice are susceptible due to deficiencies in this response. We therefore tested the hypothesis that the outcome of MeV-hsp70 interaction may be dependent upon the host H-2 haplotype. The impact of selective neuronal hsp70 overexpression on Ed MeV brain infection was tested with congenic C57BL/10 H-2(d) neonatal mice. In this context, hsp70 overexpression conferred complete protection against virus-induced mortality, compared to >30% mortality in nontransgenic mice. Selective depletion of T-cell populations showed that transgenic mice exhibit a diminished reliance on T cells for protection. Brain transcript analysis indicated enhanced innate immune activation and signaling through Toll-like receptors 2 and 4 at early times postinfection for transgenic infected mice relative to those for nontransgenic infected mice. Collectively, results suggest that hsp70 can enhance innate antiviral immunity through Toll-like receptor signaling, supporting a protective role for physiological responses that enhance tissue levels of hsp70 (e.g., fever), and that the H-2 haplotype determines the effectiveness of this response.
Collapse
|
10
|
Buccellato MA, Carsillo T, Traylor Z, Oglesbee M. Heat shock protein expression in brain: a protective role spanning intrinsic thermal resistance and defense against neurotropic viruses. PROGRESS IN BRAIN RESEARCH 2007; 162:395-415. [PMID: 17645929 DOI: 10.1016/s0079-6123(06)62019-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Heat shock proteins (HSPs) play an important role in the maintenance of cellular homeostasis, particularly in response to stressful conditions that adversely affect normal cellular structure and function, such as hyperthermia. A remarkable intrinsic resistance of brain to hyperthermia reflects protection mediated by constitutive and induced expression of HSPs in both neurons and glia. Induced expression underlies the phenomenon of hyperthermic pre-reconditioning, where transient, low-intensity heating induces HSPs that protect brain from subsequent insult, reflecting the prolonged half-life of HSPs. The expression and activity of HSPs that is characteristic of nervous tissue plays a role not just in the maintenance and defense of cellular viability, but also in the preservation of neuron-specific luxury functions, particularly those that support synaptic activity. In response to hyperthermia, HSPs mediate preservation or rapid recovery of synaptic function up to the point where damage in other organ systems becomes evident and life threatening. Given the ability of HSPs to enhance gene expression by neurotropic viruses, the constitutive and inducible HSP expression profiles would seem to place nervous tissues at risk. However, we present evidence that the virus-HSP relationship can promote viral clearance in animals capable of mounting effective virus-specific cell-mediated immune responses, potentially reflecting HSP-dependent increases in viral antigenic burden, immune adjuvant effects and cross-presentation of viral antigen. Thus, the protective functions of HSPs span the well-characterized intracellular roles as chaperones to those that may directly or indirectly promote immune function.
Collapse
Affiliation(s)
- Matthew A Buccellato
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Rd., Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
11
|
Carsillo T, Traylor Z, Choi C, Niewiesk S, Oglesbee M. hsp72, a host determinant of measles virus neurovirulence. J Virol 2006; 80:11031-9. [PMID: 16971451 PMCID: PMC1642166 DOI: 10.1128/jvi.01438-06] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transient hyperthermia such as that experienced during febrile episodes increases expression of the major inducible 70-kDa heat shock protein (hsp72). Despite the relevance of febrile episodes to viral pathogenesis and the multiple in vitro roles of heat shock proteins in viral replication and gene expression, the in vivo significance of virus-heat shock protein interactions is unknown. The present work determined the in vivo relationship between hsp72 levels and neurovirulence of an hsp72-responsive virus using the mouse model of measles virus (MV) encephalitis. Transgenic C57BL/6 mice were created to constitutively overexpress hsp72 in neurons, and these mice were inoculated intracranially with Edmonston MV (Ed MV) at 42 h of age. The mean viral RNA burden in brain was approximately 2 orders of magnitude higher in transgenic animals than in nontransgenic animals 2 to 4 weeks postinfection, and this increased burden was associated with a fivefold increase in mortality. Mice were also challenged with an Ed MV variant exhibiting an attenuated in vitro response to hsp72-dependent stimulation of viral transcription (Ed N-522D). This virus exhibited an attenuated neuropathogenicity in transgenic mice, where mortality and viral RNA burdens were not significantly different from nontransgenic mice infected with either Ed N-522D or parent Ed MV. Collectively, these results indicate that hsp72 levels can serve as a host determinant of viral neurovirulence in C57BL/6 mice, reflecting the direct influence of hsp72 on viral gene expression.
Collapse
Affiliation(s)
- Thomas Carsillo
- Department of Veterinary Biosciences, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
12
|
Schubert S, Möller-Ehrlich K, Singethan K, Wiese S, Duprex WP, Rima BK, Niewiesk S, Schneider-Schaulies J. A mouse model of persistent brain infection with recombinant Measles virus. J Gen Virol 2006; 87:2011-2019. [PMID: 16760404 DOI: 10.1099/vir.0.81838-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Measles virus (MV) nucleocapsids are present abundantly in brain cells of patients with subacute sclerosing panencephalitis (SSPE). This invariably lethal brain disease develops years after acute measles as result of a persistent MV infection. Various rodent models for MV infection of the central nervous system (CNS) have been described in the past, in which the detection of viral antigens is based on histological staining procedures of paraffin embedded brains. Here, the usage of a recombinant MV (MV-EGFP-CAMH) expressing the haemagglutinin (H) of the rodent-adapted MV-strain CAM/RB and the enhanced green fluorescent protein (EGFP) is described. In newborn rodents the virus infects neurons and causes an acute lethal encephalitis. From 2 weeks on, when the immune system of the genetically unmodified animal is maturating, intracerebral (i.c.) infection is overcome subclinically, however, a focal persistent infection in groups of neurons remains. The complete brain can be analysed in 50 or 100 microm slices, and infected autofluorescent cells are readily detected. Seven and 28 days post-infection (p.i.) 86 and 81% of mice are infected, respectively, and virus persists for more than 50 days p.i. Intraperitoneal immunization with MV 1 week before infection, but not after infection, protects and prevents persistence. The high percentage of persistence demonstrates that this is a reliable and useful model of a persistent CNS infection in fully immunocompetent mice, which allows the investigation of determinants of the immune system.
Collapse
Affiliation(s)
- S Schubert
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Straße 7, D-97078 Würzburg, Germany
| | - K Möller-Ehrlich
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Straße 7, D-97078 Würzburg, Germany
| | - K Singethan
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Straße 7, D-97078 Würzburg, Germany
| | - S Wiese
- Institute for Clinical Neurobiology, University of Würzburg, D-97078 Würzburg, Germany
| | - W P Duprex
- School of Biomedical Sciences, The Queen's University of Belfast, Belfast BT9 7BL, UK
| | - B K Rima
- School of Biomedical Sciences, The Queen's University of Belfast, Belfast BT9 7BL, UK
| | - S Niewiesk
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210-1093, USA
| | - J Schneider-Schaulies
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Straße 7, D-97078 Würzburg, Germany
| |
Collapse
|
13
|
Rulli NE, Suhrbier A, Hueston L, Heise MT, Tupanceska D, Zaid A, Wilmes A, Gilmore K, Lidbury BA, Mahalingam S. Ross River virus: Molecular and cellular aspects of disease pathogenesis. Pharmacol Ther 2005; 107:329-42. [PMID: 15923040 DOI: 10.1016/j.pharmthera.2005.03.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2005] [Indexed: 11/16/2022]
Abstract
Ross River virus (RRV) is a mosquito-borne alphavirus indigenous to Australia and the Western Pacific region and is responsible for several thousand cases of human RRV disease (RRVD) per annum. The disease primarily involves polyarthritis/arthralgia, with many patients also presenting with rash, myalgia, fever, and/or lethargy. The symptoms can be debilitating at onset, but they usually resolve within 3-6 months. Recent insights into the RRV-host relationship, associated pathology, and molecular biology of infection have generated a number of potential avenues for improved treatment. Although vaccine development has been proposed, the small market size and potential for antibody-dependent enhancement (ADE) of disease make this approach unattractive. Recent insights into the molecular basis of RRV-ADE and the virus's ability to manipulate host inflammatory and immune responses create potential new opportunities for therapeutic invention. Such interventions should overcome virus-induced dysregulation of protective host responses to promote viral clearance and/or ameliorate inflammatory immunopathology.
Collapse
Affiliation(s)
- Nestor E Rulli
- School of Health Sciences, University of Canberra, Kirinari Street, Canberra ACT 2601, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Ritchey JW, Payton ME, Eberle R. Clinicopathological characterization of monkey B virus (Cercopithecine herpesvirus 1) infection in mice. J Comp Pathol 2005; 132:202-17. [PMID: 15737347 DOI: 10.1016/j.jcpa.2004.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2004] [Accepted: 10/12/2004] [Indexed: 11/24/2022]
Abstract
The purpose of this study was to establish a small animal model for monkey B virus (BV) infection. Mice were inoculated intramuscularly with several BV isolates. Comparisons were based upon the doses required to produce infection (ID50), non-central nervous system (CNS) clinical disease (CS50), CNS disease (CNSD50) and lethal effect (LD50). Strains differed in respect of the dose required to produce clinical disease in BALB/c mice. C57BL/6 mice were more resistant than BALB/c mice to CNS disease. Skin lesions at the inoculation site consisted of epidermal necrosis, ulceration, serocellular crusts and underlying dermatitis. CNS lesions included marked inflammation in the ipsilateral dorsal root ganglion and lumbar spinal cord (point of viral entry). The distribution of the lumbar spinal cord lesions suggested viral entry via sensory afferent neurons, ventral motor tracts, or both. The lesions in the more cranial spinal cord segments suggested ascension to the brain via bilateral spinothalamic and spinoreticular tracts. Brain lesions included encephalitis with neuronal necrosis and white matter destruction located consistently at the base of the brainstem, the reticular system, and rostrally to the thalamus and hypothalamus. Viral antigen was detected immunohistochemically in the lesions. The results indicated an ascending encephalomyelitis syndrome similar to that produced by BV in man.
Collapse
Affiliation(s)
- J W Ritchey
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078-2007, USA
| | | | | |
Collapse
|
15
|
Abstract
High vaccination coverage rates and the administration of a second dose of measles vaccine have resulted in a significant decline in the incidence of measles and neurologic diseases due to measles in many countries. However, intermittent outbreaks of measles still occur even in countries with excellent vaccination coverage, suggesting the existence of high rates of measles virus introduction from endemic regions and/or waning of vaccine-induced immunity. Strategies to sustain high levels of global immunity to measles virus by increasing vaccine coverage with routine and supplementary vaccination campaigns must be supported.
Collapse
Affiliation(s)
- Martin O Ota
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA.
| | | | | |
Collapse
|
16
|
Pütz MM, Bouche FB, de Swart RL, Muller CP. Experimental vaccines against measles in a world of changing epidemiology. Int J Parasitol 2003; 33:525-45. [PMID: 12782053 DOI: 10.1016/s0020-7519(03)00062-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Vaccination with the current live attenuated measles vaccine is one of the most successful and cost-effective medical interventions. However, as a result of persisting maternal antibodies and immaturity of the infant immune system, this vaccine is poorly immunogenic in children <9 months old. Immunity against the live vaccine is less robust than natural immunity and protection less durable. There may also be some concern about (vaccine) virus spread during the final stage of an eventual measles eradication program. Opinions may differ with respect to the potential threat that some of these concerns may be to the World Health Organisation goal of measles elimination, but there is a consensus that the development of new measles vaccines cannot wait. Candidate vaccines are based on viral or bacterial vectors expressing recombinant viral proteins, naked DNA, immune stimulating complexes or synthetic peptides mimicking neutralising epitopes. While some of these candidate vaccines have proven their efficacy in monkey studies, aerosol formulated live attenuated measles vaccine are evaluated in clinical trials.
Collapse
Affiliation(s)
- Mike M Pütz
- Department of Immunology, Laboratoire National de Santé, 20A Rue Auguste Lumière, 1950 Luxembourg, Luxembourg
| | | | | | | |
Collapse
|
17
|
Abstract
Measles virus (MV) infection still belongs to the most important infectious diseases world wide. To identify the components of the immune system that combat MV infection, infection models in rodents have been established. In rats and mice, the immune response to experimental MV infection is governed by the major histocombatibility complex (MHC). According to the MHC haplotype, the functional composition of the T-cell subsets determines the degree of susceptibility to experimental measles virus infection. CD4+ T-cells are the most important T-cell subset in combating experimental MV infection in rodents. However, the mechanism of action still remains to be elucidated.
Collapse
Affiliation(s)
- Gerald Weidinger
- Department of Neuroimmunology, Max-Planck Institute of Neurobiology, Planegg-Martinsried, Germany.
| |
Collapse
|
18
|
Gundel I, Weidinger G, Ter Meulen V, Heesemann J, Rüssmann H, Niewiesk S. Oral immunization with recombinant Yersinia enterocolitica expressing a measles virus CD4 T cell epitope protects against measles virus-induced encephalitis. J Gen Virol 2003; 84:775-779. [PMID: 12655077 DOI: 10.1099/vir.0.18834-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Immunization via the oral route with an attenuated Yersinia enterocolitica strain expressing a fragment of the measles virus nucleocapsid protein (aa 79-161) via its type III protein secretion system induced a T helper type 1 response in immunized C3H mice, which conferred protection against measles virus-induced encephalitis in a time- and dose-dependent manner.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/immunology
- Disease Models, Animal
- Dose-Response Relationship, Immunologic
- Encephalitis, Viral/prevention & control
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Genetic Vectors
- Measles/immunology
- Measles/prevention & control
- Measles Vaccine/administration & dosage
- Measles Vaccine/genetics
- Mice
- Mice, Inbred C3H
- Morbillivirus/immunology
- Nucleocapsid Proteins/biosynthesis
- Nucleocapsid Proteins/genetics
- T-Lymphocytes, Helper-Inducer/immunology
- Time Factors
- Vaccination
- Vaccines, Synthetic/administration & dosage
- Yersinia enterocolitica/metabolism
Collapse
Affiliation(s)
- Iris Gundel
- Institut für Virologie und Immunbiologie, Versbacher Str. 7, 97078 Würzburg, Germany
| | - Gerald Weidinger
- Institut für Virologie und Immunbiologie, Versbacher Str. 7, 97078 Würzburg, Germany
| | - Volker Ter Meulen
- Institut für Virologie und Immunbiologie, Versbacher Str. 7, 97078 Würzburg, Germany
| | - Jürgen Heesemann
- Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, München, Germany
| | - Holger Rüssmann
- Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, München, Germany
| | - Stefan Niewiesk
- Institut für Virologie und Immunbiologie, Versbacher Str. 7, 97078 Würzburg, Germany
| |
Collapse
|
19
|
Schneider-Schaulies J, Meulen VT, Schneider-Schaulies S. Measles infection of the central nervous system. J Neurovirol 2003; 9:247-52. [PMID: 12707855 DOI: 10.1080/13550280390193993] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2002] [Revised: 10/22/2002] [Accepted: 11/13/2002] [Indexed: 10/20/2022]
Abstract
Central nervous system (CNS) complications occurring early and late after acute measles are serious and often fatal. In spite of functional cell-mediated immunity and high antiviral antibody titers, an immunological control of the CNS infection is not achieved in patients suffering from subacute sclerosing panencephalitis (SSPE). The known cellular receptors for measle virus (MV) in humans, CD46 and CD150 (signaling lymphocyte activation molecule, SLAM), are important components of the viral tropism by mediating binding and entry to peripheral cells. Because neural cells do not express SLAM and only sporadically CD46, virus entry to neural cells, and spread within the CNS, remain mechanistically unclear. Mice, hamsters, and rats have been used as model systems to study MV-induced CNS infections, and revealed interesting aspects of virulence, persistence, the immune response, and prerequisites of protection. With the help of recombinant MV and mice expressing transgenic receptors, questions such as receptor-dependent viral spread, or viral determinants of virulence, have been investigated. However, many questions concerning the human MV-induced CNS diseases are still open.
Collapse
|
20
|
Hirama K, Togashi KI, Wakasa C, Yoneda M, Nishi T, Endo Y, Miura R, Tsukiyama-Kohara K, Kai C. Cytotoxic T-lymphocyte activity specific for hemagglutinin (H) protein of canine distemper virus in dogs. J Vet Med Sci 2003; 65:109-12. [PMID: 12576714 DOI: 10.1292/jvms.65.109] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cytotoxic T-lymphocyte (CTL) responses to hemagglutinin (H) protein of canine distemper virus (CDV) were evaluated in dogs using the replication-deficient adenovirus protein expression system. Skin fibroblasts were isolated from two dogs and were infected with recombinant adenovirus bearing the CDV-H gene (Ade-CDVH). CTL assay was performed using fibroblasts expressing CDV-H protein as target cells and peripheral blood lymphocytes (PBL) collected from the same dogs one week after immunization of CDV as effector cells. Specific cytotoxic activity was observed against autologous but not heterologous fibroblasts expressing CDV-H protein. These results indicate that the CTL epitope(s) were localized in the H protein.
Collapse
Affiliation(s)
- Kyoko Hirama
- Laboratory Animal Research Center, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Chesler DA, Reiss CS. The role of IFN-gamma in immune responses to viral infections of the central nervous system. Cytokine Growth Factor Rev 2002; 13:441-54. [PMID: 12401479 DOI: 10.1016/s1359-6101(02)00044-8] [Citation(s) in RCA: 142] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Interferon (IFN)-gamma, is not only a marker of T(H)1 CD4, CD8 and natural killer (NK) cells, it is also a critical antiviral mediator which is central to the elimination of viruses from the CNS. In this review, we describe IFN-gamma, its receptor, signal transduction from receptor engagement, and antiviral downstream mediators. We demonstrate that although neurons are post-mitotic and non-renewing, they respond to IFN-gamma in a fashion similar to peripheral fibroblasts or lymphocytes. We have illustrated this review with details about studies on the role(s) of IFN-gamma in the pathogenesis of measles virus (MV), herpes simplex virus (HSV) type 1, and vesicular stomatitis virus (VSV) infections of the CNS. For VSV infection, IFN-gamma signals through Jaks 1 and 2 and STAT1 to activate (interferon regulatory factor) IRF-1; although viral protein synthesis is inhibited, PKR is not a critical mediator in the antiviral response to VSV in murine neurons. In contrast, induction of nitric oxide synthase (NOS) type 1 and its production of nitric oxide is essential in the elimination of viruses from neurons.
Collapse
Affiliation(s)
- David A Chesler
- Department of Biology, New York University, 1009 Main Building, 100 Washington Square East, New York, NY 10003, USA
| | | |
Collapse
|
22
|
Galbraith SE, McQuaid S, Hamill L, Pullen L, Barrett T, Cosby SL. Rinderpest and peste des petits ruminants viruses exhibit neurovirulence in mice. J Neurovirol 2002; 8:45-52. [PMID: 11847591 DOI: 10.1080/135502802317247802] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Members of the morbillivirus genus, canine distemper (CDV), phocine distemper virus (PDV), and the cetacean viruses of dolphins and porpoises exhibit high levels of CNS infection in their natural hosts. CNS complications are rare for measles virus (MV) and are not associated with rinderpest virus (RPV) and peste des petits ruminants virus (PPRV) infection. However, it is possible that all morbilliviruses infect the CNS but in some hosts are rapidly cleared by the immune response. In this study, we assessed whether RPV and PPRV have the potential to be neurovirulent. We describe the outcome of infection, of selected mouse strains, with isolates of RPV, PPRV, PDV, porpoise morbillivirus (PMV), dolphin morbillivirus (DMV), and a wild-type strain of MV. In the case of RPV virus, strains with different passage histories have been examined. The results of experiments with these viruses were compared with those using neuroadapted and vaccine strains of MV, which acted as positive and negative controls respectively. Intracerebral inoculation with RPV (Saudi/81) and PPRV (Nigeria75/1) strains produced infection in Balb/C and Cd1, but not C57 suckling mice, whereas the CAM/RB rodent-adapted strain of MV infected all three strains of mice. Weanling mice were only infected by CAM/RB. Intranasal and intraperitoneal inoculation failed to produce infection with any virus strains. We have shown that, both RPV and PPRV, in common with other morbilliviruses are neurovirulent in a permissive system. Transient infection of the CNS of cattle and goats with RPV and PPRV, respectively, remains a possibility, which could provide relevant models for the initial stages of MV infection in humans.
Collapse
Affiliation(s)
- Sareen E Galbraith
- School of Biology and Biochemistry, The Queen's University of Belfast, Belfast, Northern Ireland, United Kingdom
| | | | | | | | | | | |
Collapse
|
23
|
Liebert UG. Slow and persistent virus infections of neurones--a compromise for neuronal survival. Curr Top Microbiol Immunol 2001; 253:35-60. [PMID: 11417139 DOI: 10.1007/978-3-662-10356-2_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Affiliation(s)
- U G Liebert
- Institute of Virology, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| |
Collapse
|
24
|
Weidinger G, Ohlmann M, Schlereth B, Sutter G, Niewiesk S. Vaccination with recombinant modified vaccinia virus Ankara protects against measles virus infection in the mouse and cotton rat model. Vaccine 2001; 19:2764-8. [PMID: 11282186 DOI: 10.1016/s0264-410x(00)00531-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Modified vaccinia virus Ankara (MVA) has been used as an experimental vaccine vector against respiratory infections. We have tested the safety and immunogenicity of a recombinant virus expressing the hemagglutinin of measles virus (MVA-MV-H) using the mouse model of measles virus induced encephalitis and the cotton rat model for respiratory infection. MVA-MV-H proved to induce a TH1 response, neutralizing antibodies and conferred protection against both encephalitis and lung infection. The cotton rat is very sensitive to infection with replication competent vaccinia virus. In these animals MVA-MV-H proved to be a very well tolerated vaccine. However, the efficiency in the presence of MV specific maternal antibodies was low (even using a prime-boost strategy) and therefore might have to be improved.
Collapse
Affiliation(s)
- G Weidinger
- Institute of Virology and Immunobiology, University of Wuerzburg, Versbacher Str. 7, 97078 Wurzburg, Germany
| | | | | | | | | |
Collapse
|
25
|
Weidinger G, Henning G, ter Meulen V, Niewiesk S. Inhibition of major histocompatibility complex class II-dependent antigen presentation by neutralization of gamma interferon leads to breakdown of resistance against measles virus-induced encephalitis. J Virol 2001; 75:3059-65. [PMID: 11238832 PMCID: PMC114099 DOI: 10.1128/jvi.75.7.3059-3065.2001] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
BALB/c mice are resistant to measles virus (MV)-induced encephalitis due to their strong MV-specific CD4(+) T-cell response. Resistance is broken by neutralization of gamma interferon with monoclonal antibodies, indicating an important role for this pleiotropic cytokine. Here, we demonstrate that mouse gamma interferon has no direct antiviral effect in vitro and in vivo. The breakdown of resistance is due neither to a switch in the T-helper response nor to an impaired migration of CD4(+) T cells. Neutralization of gamma interferon interferes with the major histocompatibility complex class II-dependent antigen presentation and subsequent proliferation of CD4(+) T cells in vitro and in vivo. In consequence, the reduction in numbers of CD4(+) T cells below a protective threshold leads to susceptibility to MV-induced encephalitis.
Collapse
Affiliation(s)
- G Weidinger
- Institute of Virology and Immunobiology, University of Wuerzburg, 97078 Wuerzburg, Germany
| | | | | | | |
Collapse
|
26
|
Neumeister C, Nanan R, Cornu TI, Lüder CGK, Ter Meulen V, Naim H, Niewiesk S. Measles virus and canine distemper virus target proteins into a TAP-independent MHC class I-restricted antigen-processing pathway. J Gen Virol 2001; 82:441-447. [PMID: 11161284 DOI: 10.1099/0022-1317-82-2-441] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
After infection of CEM174.T2 cells [deficient for the transporter of antigen presentation (TAP)] with measles virus (MV) the nucleocapsid protein is recognized by L(d)-restricted cytotoxic T cells in a TAP-independent, chloroquine-sensitive fashion. Presentation via the TAP-independent pathway requires virus replication. During MV infection of the cell the nucleocapsid as well as the matrix protein enter the endolysosomal compartment as indicated by colocalization with the lysosomal-associated membrane protein 1 (LAMP-1). Similarly, the nucleocapsid protein of canine distemper virus (CDV) is recognized in a TAP-independent fashion. In addition, a recombinant MV expressing bacterial beta-galactosidase protein is able to introduce the recombinant antigen into the TAP-independent pathway whereas a vaccinia virus expressing beta-galactosidase is not. These data and a report about TAP-independent recognition of parainfluenza virus type 1 suggest that members of the Paramyxoviridae family regularly introduce viral proteins into the TAP-independent antigen-processing pathway.
Collapse
Affiliation(s)
- Claudia Neumeister
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078 Würzburg, Germany1
| | - Ralph Nanan
- Children Hospital, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany3
| | - Tatjana I Cornu
- Institute of Molecular Biology, University of Zürich, Winterthurer Str. 190, 8057 Zürich, Switzerland2
| | - Carsten G K Lüder
- Institute of Hygiene and Microbiology, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany4
| | - Volker Ter Meulen
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078 Würzburg, Germany1
| | - Hussein Naim
- Institute of Molecular Biology, University of Zürich, Winterthurer Str. 190, 8057 Zürich, Switzerland2
| | - Stefan Niewiesk
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078 Würzburg, Germany1
| |
Collapse
|
27
|
Weidinger G, Czub S, Neumeister C, Harriott P, Ter Meulen V, Niewiesk S. Role of CD4(+) and CD8(+) T cells in the prevention of measles virus-induced encephalitis in mice. J Gen Virol 2000; 81:2707-2713. [PMID: 11038383 DOI: 10.1099/0022-1317-81-11-2707] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Depending on their major histocompatibility complex (MHC) haplotype, inbred mouse strains are either resistant (H2-d, BALB/c), susceptible (H2-k, C3H) or partially resistant (H2-dxk, BaCF1) to intracerebral infection with the neurotropic rodent-adapted measles virus (MV) strain CAM/RBH. Here, mortality is demonstrated to be correlated directly with virus spread and virus replication in the CNS and to be inversely correlated with the activation of MV-specific T cells. Previously, it has been shown that primary CD4(+) T cells alone are protective in the resistant background. In the susceptible background, CD4(+) T cells acquire protective capacity after immunization with a newly defined CD4(+) T cell epitope peptide. In the partially resistant mice, CD4(+) T cells provide help for CD8(+) T cells and protect in cooperation with them. It seems that the lytic capacity of CD8(+) T cells is crucial in providing protection, as MV-specific L(d)-restricted CD8(+) T cells, which are highly lytic in vitro after transfer, protect naive animals against MV-induced encephalitis (MVE). In contrast, K(k)-restricted CD8(+) T cells with low lytic capacity do not protect. In the MVE model, CD4(+) T cells are able to protect either alone (resistant mice), through cooperation with CD8(+) T cells (intermediate susceptible) or after immunization as secondary T cells (susceptible mice). CD8(+) T cells are able to protect alone after immunization if they are cytolytic. Thus, susceptibility and resistance depend upon the functional composition of CD4(+) and CD8(+) T cells governed by the MHC haplotype.
Collapse
Affiliation(s)
- Gerald Weidinger
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078 Würzburg, Germany1
| | - Stefanie Czub
- Institute of Pathology, University of Würzburg, Würzburg, Germany2
| | - Claudia Neumeister
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078 Würzburg, Germany1
| | - Pat Harriott
- Centre for Peptide and Protein Engineering, Queen's University of Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK3
| | - Volker Ter Meulen
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078 Würzburg, Germany1
| | - Stefan Niewiesk
- Institute of Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, 97078 Würzburg, Germany1
| |
Collapse
|
28
|
Spreng S, Gentschev I, Goebel W, Weidinger G, ter Meulen V, Niewiesk S. Salmonella vaccines secreting measles virus epitopes induce protective immune responses against measles virus encephalitis. Microbes Infect 2000; 2:1687-92. [PMID: 11137042 DOI: 10.1016/s1286-4579(00)01325-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In the present study we describe a live vaccine against measles virus (MV) infection on the basis of attenuated Salmonella typhimurium aroA secreting MV antigens via the Escherichia coli alpha-hemolysin secretion system. Two well-characterized MV epitopes, a B-cell epitope of the MV fusion protein (amino acids 404-414) and a T-cell epitope of the MV nucleocapsid protein (amino acids 79-99) were fused as single or repeating units to the C-terminal secretion signal of the E. coli hemolysin and expressed in secreted form by the attenuated S. typhimurium aroA SL7207. Immunization of MV-susceptible C3H mice revealed that S. typhimurium SL7207 secreting these antigens provoked a humoral and a cellular MV-specific immune response, respectively. Mice vaccinated orally with a combination of both recombinant S. typhimurium strains showed partial protection against a lethal MV encephalitis after intracerebral challenge with a rodent-adapted, neurotropic MV strain.
Collapse
Affiliation(s)
- S Spreng
- Lehrstuhl für Mikrobiologie, Theodor-Boveri-Institut für Biowissenschaften, Am Hubland, D-97074, Würzburg, Germany.
| | | | | | | | | | | |
Collapse
|
29
|
Ohishi K, Kamata H, Yamanouchi K, Barrett T. Identification of T-helper cell epitopes in the hypervariable region of the nucleocapsid (N) protein of rinderpest virus (RPV) in cattle. Vaccine 2000; 18:3077-81. [PMID: 10856786 DOI: 10.1016/s0264-410x(99)00538-1] [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] [Indexed: 10/17/2022]
Abstract
The proliferative responses to synthetic peptides by lymphocytes derived from rinderpest virus (RPV)-infected cattle, the natural host for RPV, were assayed by determining [3H]thymidine incorporation into the DNA. In eight out of twelve cattle tested, significant responses were detected to peptides representing amino acids 452-501 in the C-terminal hypervariable region of the virus nucleocapsid (N) protein. It appears that helper T-cell epitope(s) for cattle which can be broadly recognized within an MHC diverse population, exists in this region of the protein.
Collapse
Affiliation(s)
- K Ohishi
- Institute for Animal Health, Pirbright Laboratory, Ash Road, Surrey, GU24 0NF, Woking, UK
| | | | | | | |
Collapse
|
30
|
Schadeck EB, Partidos CD, Fooks AR, Obeid OE, Wilkinson GW, Stephenson JR, Steward MW. CTL epitopes identified with a defective recombinant adenovirus expressing measles virus nucleoprotein and evaluation of their protective capacity in mice. Virus Res 1999; 65:75-86. [PMID: 10564754 DOI: 10.1016/s0168-1702(99)00103-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cytotoxic T-lymphocyte (CTL) responses to measles virus (MV) play an important role in recovery from infection, with one of the major target proteins for CTL activity being the nucleoprotein (Np). In this report, a replication-deficient adenovirus-5 recombinant, expressing for MV Np (Rad68) was tested for in vivo priming of MV Np-specific CTL responses in BALB/c and CBA mice. In both strains of mice strong Np-specific CTL responses were induced and these responses were shown to be MHC class I restricted. Using overlapping 15mer peptides spanning residues 1-505 of MV Np a single epitope comprising residues 281-295 was identified in BALB/c mice whereas, in CBA mice two epitopes comprising residues 51-65 and 81-95, were identified. These epitopes were found to contain class I motifs for H-2L(d) and H-2K(k) MHC molecules, respectively. Immunization of BALB/c and CBA mice with the respective CTL epitopes resulted in the in vivo induction of peptide-and MV Np-specific CTL responses. In addition, the identified H-2K(k) restricted CTL epitopes conferred some protection against encephalitis induced following intracerebral challenge with a lethal dose of canine distemper virus (the Np of which shares 70% sequence homology with MV Np). These findings highlight the potential of using well-defined CTL epitopes to control virus infection.
Collapse
Affiliation(s)
- E B Schadeck
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, UK
| | | | | | | | | | | | | |
Collapse
|
31
|
Duprex WP, Duffy I, McQuaid S, Hamill L, Cosby SL, Billeter MA, Schneider-Schaulies J, ter Meulen V, Rima BK. The H gene of rodent brain-adapted measles virus confers neurovirulence to the Edmonston vaccine strain. J Virol 1999; 73:6916-22. [PMID: 10400789 PMCID: PMC112776 DOI: 10.1128/jvi.73.8.6916-6922.1999] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/1999] [Accepted: 05/03/1999] [Indexed: 12/31/2022] Open
Abstract
Molecular determinants of neuropathogenesis have been shown to be present in the hemagglutinin (H) protein of measles virus (MV). An H gene insertion vector has been generated from the Edmonston B vaccine full-length infectious clone of MV. Using this vector, it is possible to insert complete H open reading frames into the parental (Edtag) background. The H gene from a rodent brain-adapted MV strain (CAM/RB) was inserted into this vector, and a recombinant virus (EdtagCAMH) was rescued by using a modified vaccinia virus which expresses T7 RNA polymerase (MVA-T7). The recombinant virus grew at an equivalent rate and to similar titers as the CAM/RB and Edtag parental viruses. Neurovirulence was assayed in a mouse model for MV encephalitis. Viruses were injected intracerebrally into the right cortex of C57/BL/6 suckling mice. After infection mice inoculated with the CAM/RB strain developed hind limb paralysis and ataxia. Clinical symptoms were never observed with an equivalent dose of Edtag virus or in sham infections. Immunohistochemistry (IHC) was used to detect viral antigen in formalin-fixed brain sections. Measles antigen was observed in neurons and neuronal processes of the hippocampus, frontal, temporal, and olfactory cortices and neostriatum on both sides of symmetrical structures. Viral antigen was not detected in mice infected with Edtag virus. Mice infected with the recombinant virus, EdtagCAMH, became clinically ill, and virus was detected by IHC in regions of the brain similar to those in which it was detected in animals infected with CAM/RB. The EdtagCAMH infection had, however, progressed much less than the CAM/RB virus at 4 days postinfection. It therefore appears that additional determinants are encoded in other regions of the MV genome which are required for full neurovirulence equivalent to CAM/RB. Nevertheless, replacement of the H gene alone is sufficient to cause neuropathology.
Collapse
Affiliation(s)
- W P Duprex
- School of Biology and Biochemistry, The Queen's University of Belfast, Belfast BT9 7BL, Northern Ireland, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Manchester M, Eto DS, Oldstone MB. Characterization of the inflammatory response during acute measles encephalitis in NSE-CD46 transgenic mice. J Neuroimmunol 1999; 96:207-17. [PMID: 10337919 DOI: 10.1016/s0165-5728(99)00036-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Expression of the human measles virus receptor, CD46, in the murine central nervous system allows infection and replication by wild-type human measles virus (MV) strains (Rall, G.F., Manchester, M., Daniels L.R., Callahan, E., Belman, A., Oldstone, M.B.A., 1997. A transgenic mouse model for measles virus infection of the brain. Proc. Natl. Acad. Sci. U.S.A. 94, 2243-2248). MV replicates in neurons in focal lesions of the cortex, hippocampus and thalamus, leading to death of the animals. In MV-infected CD46 transgenic mice, infiltration of CD4+ and CD8+ T-lymphocytes, B-lymphocytes and macrophages was seen. Upregulation of MHC class I and class II molecules was observed, along with reactive astrocytosis and microgliosis. Increased chemokine mRNAs, especially RANTES and IP-10, and cytokine RNAs IL-6, TNF-alpha, and IL1-beta were observed. Apoptosis of neurons also was increased. No MV replication or inflammation was seen in similarly inoculated nontransgenic littermates. These results further characterize the MV-induced encephalitis in CD46 transgenic mice and highlight similarities to MV infection of the human CNS.
Collapse
Affiliation(s)
- M Manchester
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
33
|
Lawrence DM, Vaughn MM, Belman AR, Cole JS, Rall GF. Immune response-mediated protection of adult but not neonatal mice from neuron-restricted measles virus infection and central nervous system disease. J Virol 1999; 73:1795-801. [PMID: 9971756 PMCID: PMC104418 DOI: 10.1128/jvi.73.3.1795-1801.1999] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/1998] [Accepted: 11/11/1998] [Indexed: 12/20/2022] Open
Abstract
In many cases of neurological disease associated with viral infection, such as measles virus (MV)-induced subacute sclerosing panencephalitis in children, it is unclear whether the virus or the antiviral immune response within the brain is the cause of disease. MV inoculation of transgenic mice expressing the human MV receptor, CD46, exclusively in neurons resulted in neuronal infection and fatal encephalitis within 2 weeks in neonates, while mice older than 3 weeks of age were resistant to both infection and disease. At all ages, T lymphocytes infiltrated the brain in response to inoculation. To determine the role of lymphocytes in disease progression, CD46(+) mice were back-crossed to T- and B-cell-deficient RAG-2 knockout mice. The lymphocyte deficiency did not affect the outcome of disease in neonates, but adult CD46(+) RAG-2(-) mice were much more susceptible to both neuronal infection and central nervous system disease than their immunocompetent littermates. These results indicate that CD46-dependent MV infection of neurons, rather than the antiviral immune response in the brain, produces neurological disease in this model system and that immunocompetent adult mice, but not immunologically compromised or immature mice, are protected from infection.
Collapse
Affiliation(s)
- D M Lawrence
- The Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | | | | | |
Collapse
|
34
|
Mrkic B, Pavlovic J, Rülicke T, Volpe P, Buchholz CJ, Hourcade D, Atkinson JP, Aguzzi A, Cattaneo R. Measles virus spread and pathogenesis in genetically modified mice. J Virol 1998; 72:7420-7. [PMID: 9696838 PMCID: PMC109970 DOI: 10.1128/jvi.72.9.7420-7427.1998] [Citation(s) in RCA: 241] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/1998] [Accepted: 06/08/1998] [Indexed: 12/11/2022] Open
Abstract
Attenuated Edmonston measles virus (MV-Edm) is not pathogenic in standard mice. We show here that MV-Edm inoculated via the natural respiratory route has a limited propagation in the lungs of mice with a targeted mutation inactivating the alpha/beta interferon receptor. A high dose of MV-Edm administered intracerebrally is lethal for about half of these mice. To study the consequences of the availability of a high-affinity receptor for MV propagation, we generated alpha/beta interferon-defective mice expressing human CD46 with human-like tissue specificity. Intranasal infection of these mice with MV-Edm resulted in enhanced spread to the lungs and more prominent inflammatory response. Virus replication was also detected in peripheral blood mononuclear cells, the spleen, and the liver. Moreover, intracerebral inoculation of adult animals with low MV-Edm doses caused encephalitis with almost inevitably lethal outcome. We conclude that in mice alpha/beta interferon controls MV infection and that a high-affinity receptor facilitates, but is not strictly required for, MV spread and pathogenesis.
Collapse
Affiliation(s)
- B Mrkic
- Institut für Molekularbiologie Abt. I, Universität Zürich, Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Linn ML, Mateo L, Gardner J, Suhrbier A. Alphavirus-specific cytotoxic T lymphocytes recognize a cross-reactive epitope from the capsid protein and can eliminate virus from persistently infected macrophages. J Virol 1998; 72:5146-53. [PMID: 9573286 PMCID: PMC110085 DOI: 10.1128/jvi.72.6.5146-5153.1998] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Persistent alphavirus infections in synovial and neural tissues are believed to be associated with chronic arthritis and encephalitis, respectively, and represent likely targets for CD8+ alphabeta cytotoxic T lymphocytes (CTL). Here we show that the capsid protein is a dominant target for alphavirus-specific CTL in BALB/c mice and that capsid-specific CTL from these mice recognize an H-2Kd restricted epitope, QYSGGRFTI. This epitope lies in the highly conserved region of the capsid protein, and QYSGGRFTI-specific CTL were cross reactive across a range of Old World alphaviruses. In vivo the acute primary viraemia of these highly cytopathic viruses was unaffected by QYSGGRFTI-specific CTL. However, in vitro these CTL were able to completely clear virus from macrophages persistently and productively infected with the arthrogenic alphavirus Ross River virus.
Collapse
Affiliation(s)
- M L Linn
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | | | | | | |
Collapse
|
36
|
Nakamura K, Ohishi K, Ohkubo S, Kamata H, Yamanouchi K, Fujiwara K, Kai C. Immunizing effect of vaccinia virus expressing the nucleoprotein of rinderpest virus on systemic rinderpest virus infection in rabbits. Comp Immunol Microbiol Infect Dis 1998; 21:91-9. [PMID: 9611679 DOI: 10.1016/s0147-9571(97)00022-2] [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] [Indexed: 02/07/2023]
Abstract
A recombinant vaccinia virus (RVV) expressing the nucleoprotein (NP) of rinderpest virus (RPV) was examined in rabbits for the involvement of the NP protein in protection from the RPV infection. Despite their production of anti-NP antibody, the RVV-immunized rabbits succumbed to the RPV challenge, although there was a slight delay in the onset of disease after the low-dose challenge. On the other hand, the animals immunized with RVV expressing the hemagglutinin (H) protein of the RPV were completely protected. These results indicate that the NP protein might be not so effective as the H protein for the protection against viremic and systemic infection with RPV.
Collapse
Affiliation(s)
- K Nakamura
- Department of Veterinary Microbiology, University of Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Niewiesk S, Eisenhuth I, Fooks A, Clegg JC, Schnorr JJ, Schneider-Schaulies S, ter Meulen V. Measles virus-induced immune suppression in the cotton rat (Sigmodon hispidus) model depends on viral glycoproteins. J Virol 1997; 71:7214-9. [PMID: 9311794 PMCID: PMC192061 DOI: 10.1128/jvi.71.10.7214-7219.1997] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Immune suppression during measles accounts for most of the morbidity and mortality associated with the virus infection. Experimental study of this phenomenon has been hampered by the lack of a suitable animal model. We have used the cotton rat to demonstrate that mitogen-induced proliferation of spleen cells from measles virus-infected animals is impaired. Proliferation inhibition is seen in all lymphocyte subsets and is not dependent on viral replication. Cells which express the viral glycoproteins (hemagglutinin and fusion protein) transiently by transfection induce proliferation inhibition after intraperitoneal inoculation, whereas application of a recombinant measles virus in which measles virus glycoproteins are replaced with the vesicular stomatitis virus G protein does not have an antiproliferative effect. Therefore, in vivo expression of measles virus glycoproteins is sufficient and necessary to induce inhibition of lymphocyte proliferation.
Collapse
Affiliation(s)
- S Niewiesk
- Institute of Virology and Immunobiology, University of Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
38
|
Niewiesk S, Schneider-Schaulies J, Ohnimus H, Jassoy C, Schneider-Schaulies S, Diamond L, Logan JS, ter Meulen V. CD46 expression does not overcome the intracellular block of measles virus replication in transgenic rats. J Virol 1997; 71:7969-73. [PMID: 9311889 PMCID: PMC192156 DOI: 10.1128/jvi.71.10.7969-7973.1997] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The study of measles pathogenesis and the testing of improved vaccine candidates is hampered by the lack of a small animal model which is susceptible to infection by the intranasal route. With the identification of CD46 as a measles virus (MV) receptor, it was feasible to generate transgenic rats to overcome this problem. Although there was widespread expression of CD46 in the transgenic Sprague-Dawley rats, no measles-like disease could be induced after various routes of infection. The expressed transgenic protein was functionally intact since it mediated MV fusion and was downregulated by contact with MV hemagglutinin. In vitro studies revealed that CD46-expressing rat fibroblasts take up MV but do not allow viral replication, which explains the nonpermissiveness of the transgenic rats for in vivo infection.
Collapse
Affiliation(s)
- S Niewiesk
- Institut für Virologie und Immunbiologie, Universität Würzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Several features of HLA-G's sequence and expression pattern distinguish HLA-G from its classical counterparts. These features, including HLA-G's limited polymorphism and its expression at the maternal-fetal interface, have been used as a basis for suggesting a distinct functional role for this nonclassical class I HLA molecule. On the other hand, published data do demonstrate that HLA-G has much in common with its classical counterparts. It associates with beta 2-microglobulin and cytosolic peptides, it binds to CD8, and its presence can inhibit NK-cell-mediated lysis of HLA-G-bearing target cells. To develop a model in which HLA-G's function could be more thoroughly studied, we produced several HLA-G-expressing transgenic mouse strains. We report here the results of skin graft experiments which show that nontransgenic mice reject HLA-G-expressing transgenic murine skin as foreign and that this rejection is associated with the presence in the recipient of lymphocytes capable of specifically lysing HLA-G-expressing cells. In addition, experiments are described which demonstrate that HLA-G transgenic mice recognize HLA-G as a "self" molecule. Together the reported data demonstrate that HLA-G is capable of stimulating an HLA-G-restricted CTL response, that HLA-G molecules can serve as target molecules in lytic interactions with CTLs, and that HLA-G is involved in education of the lymphocytic repertoire of HLA-G transgenic mice.
Collapse
Affiliation(s)
- C M Schmidt
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis 55455, USA
| | | | | |
Collapse
|
40
|
Müller N, Riedel M, Schwarz M, Gruber R, Ackenheil M. Immunomodulatory Effects of Neuroleptics to the Cytokine System and the Cellular Immune System in Schizophrenia. CURRENT UPDATE IN PSYCHOIMMUNOLOGY 1997. [DOI: 10.1007/978-3-7091-6870-7_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
41
|
Fooks AR, Jeevarajah D, Warnes A, Wilkinson GW, Clegg JC. Immunization of mice with plasmid DNA expressing the measles virus nucleoprotein gene. Viral Immunol 1996; 9:65-71. [PMID: 8822623 DOI: 10.1089/vim.1996.9.65] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The measles virus (MV) nucleocapsid (N) protein gene has been inserted into a plasmid vector so as to place the gene under the control of the strong constitutive human cytomegalovirus major immediate early promoter. On intramuscular injection of pMV64 DNA into C3H/He mice, seroconversion with increasing titers of N-specific serum IgG antibodies was observed over a period of 3 months. However, when 3-week-old mice were immunized by intramuscular injection of pMV64 in a two-dose schedule, and challenged intracranially with a rodent-adapted measles virus strain (CAM/RB) at 5 weeks of age, no significant protective response was seen. The lack of effective protection evoked by DNA immunization in this model, where MV challenge must take place before 8 weeks of age, may be due to inefficient induction of cell-mediated immunity resulting from expression in muscle tissue, compounded by a relatively slow rise in immune response compared with that seen with the recombinant adenovirus.
Collapse
Affiliation(s)
- A R Fooks
- Centre for Applied Microbiology and Research, Salisbury, U.K
| | | | | | | | | |
Collapse
|
42
|
Finke D, Brinckmann UG, ter Meulen V, Liebert UG. Gamma interferon is a major mediator of antiviral defense in experimental measles virus-induced encephalitis. J Virol 1995; 69:5469-74. [PMID: 7636992 PMCID: PMC189396 DOI: 10.1128/jvi.69.9.5469-5474.1995] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Measles virus infection of the central nervous system in the murine model of experimental measles virus-induced encephalitis is successfully controlled by virus-specific T-helper lymphocytes. T cells from BALB/c mice that are resistant to measles virus encephalitis proliferate well against measles virus in vitro, and bulk cultures recognize viral nucleocapsid and hemagglutinin as well as fusion proteins. The measles virus-specific T cells secrete large amounts of interleukin 2 (IL-2), gamma interferon (IFN-gamma), and tumor necrosis factor alpha (TNF-alpha) but no IL-4, IL-6, or IL-10, and hence the cytokine pattern is consistent with that of subtype 1 T-helper lymphocytes. In contrast, cells obtained from measles virus-infected susceptible C3H mice recognize measles virus proteins only weakly and secrete little IFN-gamma and TNF-alpha. Treatment of infected mice with anti-TNF-alpha antibodies has no effect on survival or virus clearance from the brain. Upon neutralization of IFN-gamma in vivo, the phenotype of measles virus-specific T-helper cells isolatable from BALB/c mice is reversed from subtype 1 to subtype 2-like. Anti-IFN-gamma antibody-treated BALB/c mice are susceptible to measles virus encephalitis, and viral clearance from the central nervous system is impaired. These results indicate that IFN-gamma plays a significant role in the control of measles virus infection of the central nervous system.
Collapse
Affiliation(s)
- D Finke
- Institut für Virologie und Immunbiologie, Universität Würzburg, Germany
| | | | | | | |
Collapse
|
43
|
van Binnendijk RS, van der Heijden RW, Osterhaus AD. Monkeys in measles research. Curr Top Microbiol Immunol 1995; 191:135-48. [PMID: 7789157 DOI: 10.1007/978-3-642-78621-1_9] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
44
|
Affiliation(s)
- U G Liebert
- Institut for Virology and Immunobiology, University of Würzburg, Germany
| | | |
Collapse
|
45
|
Affiliation(s)
- E Norrby
- MTC, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
46
|
Sakae H, Yoshikura H, Yanagi Y. L cell clone developing plaques upon infection with measles virus (Edmonston strain). Arch Virol 1994; 139:427-30. [PMID: 7832647 DOI: 10.1007/bf01310803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
An L cell clone developing cytopathic plaques upon infection with measles virus (Edmonston strain) was obtained. The sensitivity as measured by the newly devised UV-Vero assay was not significantly different between plaque-forming and non-plaque-forming L cell clones. Cytopathogenicity and sensitivity to the virus infection appear to be under different host cell regulations.
Collapse
Affiliation(s)
- H Sakae
- Department of Bacteriology, Faculty of Medicine, University of Tokyo, Japan
| | | | | |
Collapse
|
47
|
Yanagi Y, Hu HL, Seya T, Yoshikura H. Measles virus infects mouse fibroblast cell lines, but its multiplication is severely restricted in the absence of CD46. Arch Virol 1994; 138:39-53. [PMID: 7980010 DOI: 10.1007/bf01310037] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Mouse cell lines (L, NIH3T3, and RMA cells) infected with the Edmonston strain of measles virus (MV) did not exhibit cytopathic effects (CPE), consistent with the finding that mice are not susceptible to MV. Northern blot analysis, however, revealed that MV genes were transcribed in infected L and NIH3T3 cells, although expression levels were much lower than those in lytically infected Vero cells. Expression of MV genes was not detected in infected RMA cells. L and NIH3T3 cells were found to synthesize viral proteins and produce infectious virions after infection. These cell lines did not express on the surface the molecule detectable by antibodies directed against human CD46, the recently identified MV receptor. L cell transfectants expressing human CD46 exhibited CPE after MV infection, and produced 50 times more viral transcripts and 20 times more infectious virions than the parental L cells. The lowest titer of MV that induced viral multiplication in L cells as detected by cocultivation with Vero cells was larger than that in CD46+ L cells by two orders of magnitude. Our results indicate that MV can infect some mouse cells in the absence of CD46, yet the presence of CD46 facilitates multiplication and cytopathogenicity of MV in mouse cells.
Collapse
Affiliation(s)
- Y Yanagi
- Department of Bacteriology, Faculty of Medicine, University of Tokyo, Japan
| | | | | | | |
Collapse
|
48
|
UytdeHaag FG, van Binnendijk RS, Kenter MJ, Osterhaus AD. Cytotoxic T lymphocyte responses against measles virus. Curr Top Microbiol Immunol 1994; 189:151-67. [PMID: 7924435 DOI: 10.1007/978-3-642-78530-6_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- F G UytdeHaag
- Department of Virology, Erasmus University Rotterdam, The Netherlands
| | | | | | | |
Collapse
|