首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 46 毫秒
1.
2.
A key question in pandemic influenza is the relative roles of innate immunity and target cell depletion in limiting primary infection and modulating pathology. Here, we model these interactions using detailed data from equine influenza virus infection, combining viral and immune (type I interferon) kinetics with estimates of cell depletion. The resulting dynamics indicate a powerful role for innate immunity in controlling the rapid peak in virus shedding. As a corollary, cells are much less depleted than suggested by a model of human influenza based only on virus-shedding data. We then explore how differences in the influence of viral proteins on interferon kinetics can account for the observed spectrum of virus shedding, immune response, and influenza pathology. In particular, induction of high levels of interferon (“cytokine storms”), coupled with evasion of its effects, could lead to severe pathology, as hypothesized for some fatal cases of influenza.Influenza A virus causes an acute respiratory disease in humans and other mammals; in humans, it is particularly important because of the rapidity with which epidemics develop, its widespread morbidity, and the seriousness of complications. Every year, an estimated 500,000 deaths worldwide, primarily of young children and the elderly, are attributed to seasonal influenza virus infections (49). Influenza pandemics may occur when an influenza virus with new surface proteins emerges, against which the majority of the population has no preexisting immunity. Both the emergence of H5N1 virus (34) and the current H1N1 virus pandemic (43) underline the importance of understanding the dynamics of infection and disease. A key question is, what regulates virus abundance in an individual host, causing the characteristic rapid decline in virus shedding following its initial peak? The main contenders in primary influenza virus infection are depletion of susceptible target cells and the impact of the host''s innate immune response (2, 20).On infection, the influenza virus elicits an immune response, including a rapid innate response that is correlated with the observed decline in the virus load after the first 2 days of infection (1). The slower adaptive response, including both humoral and cell-mediated components, takes several days to consolidate but is important for complete virus clearance and establishment of protective immunity. During infection of an immunologically naïve host, the innate immune response is particularly important as the first line of defense against infection. The innate immune response is regulated by chemokines and cytokines, chemical messengers produced by virus-infected epithelial cells and leukocytes (23), and natural interferon-producing cells, such as plasmacytoid dendritic cells (13). Among the key cytokines induced by epithelial cells infected with influenza A virus are type I interferons (IFNs) (IFN-α/β) (23), which directly contribute to the antiviral effect on infected and neighboring cells (38).Like other viruses, influenza A viruses have evolved strategies to limit the induction of innate immune responses (38). The NS1 protein plays a dominant role, and without it, the virus is unable to grow well or to cause pathology in an immunocompetent host (14). NS1 is multifunctional and counteracts both the induction of IFN expression and the function of IFN-activated antiviral effectors via multiple mechanisms (12, 17). Individual strains of influenza A virus possess these activities to various degrees (15, 21, 22, 26), and accordingly, NS1 has been implicated as a virulence factor (3, 17). A striking effect of the failure to control the innate response to virus infection is seen as a “cytokine storm,” which causes severe pathology (8).While there is an extensive literature on modeling influenza virus spread at the population level, the individual-host scale has received much less attention (2, 4, 5, 18, 19, 20, 27, 28). In a recent important paper, Baccam et al. modeled the kinetics of influenza A virus (2). The innate dynamics were included in the form of an IFN response that delayed and reduced virus production but did not prevent it; thus, the infection was resolved primarily through near-total depletion of epithelial cells. Their model was fitted to virus titers from human volunteers exposed to H1N1 influenza virus, but no data were available on the innate immune response or epithelial cell pathology. This has been a general difficulty in developing and validating more refined within-host models; there is a lack of detailed biological data from natural host systems, in particular, measures of immune kinetics and patterns of cellular depletion.The model presented here explicitly includes the ability of IFN to induce a fully antiviral state in order to explore the relative regulatory role of innate immunity and target cell depletion. Data from experimental infections of immunologically naïve horses with an equine influenza virus (36) allowed us to calibrate our model, not only to viral kinetics, but also to IFN dynamics and cell depletion in the context of infection of a naïve natural mammalian host. With our fitted model, we then investigate modulation of the immune response.  相似文献   

3.
To escape immune recognition, viruses acquire amino acid substitutions in class I human leukocyte antigen (HLA)-presented cytotoxic T-lymphocyte (CTL) epitopes. Such viral escape mutations may (i) prevent peptide processing, (ii) diminish class I HLA binding, or (iii) alter T-cell recognition. Because residues 418 to 426 of the hypervariable influenza A virus nucleoprotein (NP418-426) epitope are consistently bound by class I HLA and presented to CTL, we assessed the impact that intraepitope sequence variability has upon T-cell recognition. CTL elicited by intranasal influenza virus infection were tested for their cross-recognition of 20 natural NP418-426 epitope variants. Six of the variant epitopes, of both H1N1 and H3N2 origin, were cross-recognized by CTL while the remaining NP418-426 epitope variants escaped targeting. A pattern emerged whereby variability at position 5 (P5) within the epitope reduced T-cell recognition, changes at P4 or P6 enabled CTL escape, and a mutation at P8 enhanced T-cell recognition. These data demonstrate that substitutions at P4 and/or P6 facilitate influenza virus escape from T-cell recognition and provide a model for the number, nature, and location of viral mutations that influence T-cell cross-recognition.Cytotoxic T-lymphocytes (CTL) kill virus-infected cells and release antiviral cytokines upon recognition of short viral peptides displayed on the cell surface by the class I HLA molecule (36). Virus-derived peptides are processed in the cytoplasm by proteasome degradation of viral proteins (25), shuttled into the lumen of the endoplasmic reticulum (ER) by the transporter-associated protein, and loaded into the basket-like groove of the class I molecule. Class I HLA molecules await peptide loading in the ER and demonstrate specificity for viral peptides with particular anchor residues representing a good fit for the class I HLA binding groove. Once stable class I HLA-peptide complexes are formed, the class I molecule and its peptide cargo are transported via the Golgi apparatus to the cell surface, where the complex is anchored to the plasma membrane (21, 36-38). CTL then survey class I HLA-presented peptides on the cell surface. Viral peptides must therefore be processed, specifically bound by class I HLA, and presented at the plasma membrane for CTL to distinguish infected cells from uninfected tissue.A high mutation rate is one of many mechanisms utilized by viruses to escape detection by the immune system. Mutations within the genome allow viruses to accumulate and select for amino acid substitutions that (i) inhibit proteasome processing and viral peptide generation (2, 23), (ii) alter anchor residues within viral peptides to diminish class I HLA binding specificity (3, 14, 24, 32), or (iii) reduce immune recognition of the class I HLA-peptide complex by varying amino acids that come in contact with the T-cell receptor (6, 10, 27, 30, 35). While viral mutations might be advantageous for escaping immune detection, such flexibility can cost the virus in terms of replicative fitness. In order to maintain reproductive fitness and structural integrity, viruses must temper their use of genetic flexibility as a means of immune escape.Influenza viruses have the well-documented ability to escape detection by various immune epitopes (3, 10, 27). A priori, investigators often assume that variable regions of the virus represent poor immune targets because such regions will not be consistently processed, presented, or recognized (15, 20). However, we along with others continue to find that a hypervariable stretch of the influenza virus nucleoprotein consisting of residues 418 to 426 (NP418-426) is presented to CTL by different HLA-B alleles (B*0702 and B*3501) in spite of extensive viral variability within this epitope (8, 10, 27, 34). Moreover, NP418-426 is a dominant immune epitope (8, 10, 27, 34). The consistent processing and presentation of NP418-426 by class I HLA can be explained by the finding that different influenza virus isolates cannot mutate the proline located at position 2 (P2) within the epitope because elimination of this proline reduces viral fitness (4, 5). Little to no variability is found at the methionine P9 anchor as well. These facts lead to the unique observation that strain-to-strain variability does not abrogate class I HLA presentation of the influenza virus NP418-426 epitope and that CTL respond to this consistently presented viral epitope in an immunodominant fashion.In this study we took advantage of the anchor residue conservation that prompts the NP418-426 epitope to be consistently presented to CTL by investigating the functional impact that influenza virus intraepitope variability has on CTL recognition. The amino acid alignment of human influenza A (H1N1 and H3N2) virus nucleoprotein molecules identifies 20 unique NP418-426 peptide sequences which demonstrate amino acid diversity between the anchors. We infected HLA-transgenic mice intranasally with influenza virus and tested CTL from these animals for their ability to recognize each of the 20 NP418-426 variants. These 20 NP418-426 sequences represent a natural “recombinant library” of viral epitopes that the immune system has and will face. The resulting data demonstrate a gradient of viral substitutions whereby CTL recognition diminishes depending upon the number of viral substitutions and their location within the epitope. Understanding how intraepitope variability impacts CTL recognition is discussed in terms of eliciting immune responses to variants of influenza.  相似文献   

4.
Influenza is an acute respiratory viral disease that is transmitted in the first few days of infection. Evasion of host innate immune defenses, including natural killer (NK) cells, is important for the virus''s success as a pathogen of humans and other animals. NK cells encounter influenza viruses within the microenvironment of infected cells and are important for host innate immunity during influenza virus infection. It is therefore important to investigate the direct effects of influenza virus on NK cells. In this study, we demonstrated for the first time that influenza virus directly infects and replicates in primary human NK cells. Viral entry into NK cells was mediated by both clathrin- and caveolin-dependent endocytosis rather than through macropinocytosis and was dependent on the sialic acids on cell surfaces. In addition, influenza virus infection induced a marked apoptosis of NK cells. Our findings suggest that influenza virus can directly target and kill NK cells, a potential novel strategy of influenza virus to evade the NK cell innate immune defense that is likely to facilitate viral transmission and may also contribute to virus pathogenesis.Influenza is an acute respiratory virus infection that continues to pose endemic, zoonotic, and pandemic threats to human health, with significant morbidity and mortality (17). At the early phase of viral infection, innate immunity plays important roles in host defense by limiting viral replication and helping to initiate an adaptive immune response. Natural killer (NK) cells are key effector cells in innate immunity and play a critical role in the first line of host defense against acute viral infections by directly destroying infected cells without the need for prior antigen stimulation (7, 20). As influenza illness and virus transmission usually occur in the first few days of infection, the virus has to devise strategies to evade host innate immune responses, including NK cell immunity (15, 21).NK cells can recognize and kill influenza virus-infected cells (2, 10, 23); to counteract this killing, however, influenza virus has developed an escape strategy that inhibits NK cell cytotoxicity by increasing the binding of two inhibitory receptors to the infected cells after infection (1). The individuals with complete NK cell deficiency developed life-threatening varicella zoster virus and cytomegalovirus infection, but no severe influenza virus infection occurred (30, 40). Indeed, the interaction between human NK cells and influenza virus remains poorly understood. After influenza virus infection, respiratory epithelial cells release inflammatory chemokines that recruit NK cells to the site of infection (12). As a lytic virus, numerous influenza virus particles are released from the infected epithelia and macrophages (5, 9, 33). In the infected microenvironment, NK cells undoubtedly encounter these infective virus particles. It is therefore important to investigate the direct interaction of NK cells with influenza virus. Patients with severe influenza virus infection were shown to have diminished NK cells in peripheral blood and an almost complete absence of pulmonary NK cells, together with marked apoptosis (13, 42). During influenza virus infection in mice, a transient increase of NK cytotoxicity is followed by a marked decrease in NK cell activity, with a virus dose-dependent effect (8, 28). These data suggest that influenza virus may directly target NK cells as part of its immunoevasion strategies. However, no reports of the direct effects of influenza virus on human NK cells have so far been available.In this study, we demonstrated that influenza virus infects and replicates in primary human NK cells. Viral infection was dependent on sialic acids on the cells. The entry was mediated by both clathrin- and caveolin-dependent endocytosis rather than macropinocytosis. Influenza virus infection induced a marked apoptosis of NK cells, which contributed to reduced NK cell cytotoxicity. This, to the best of our knowledge, is the first paper to demonstrate that influenza virus can directly infect NK cells and induce cell apoptosis. These findings suggest that influenza virus may have developed a novel strategy to evade NK cell innate immune defenses, which is likely to facilitate viral transmission and may also contribute to virus pathogenesis.  相似文献   

5.
6.
Prophylactic or therapeutic immunomodulation is an antigen-independent strategy that induces nonspecific immune system activation, thereby enhancing host defense to disease. In this study, we investigated the effect of prophylactic immunomodulation on the outcome of influenza virus infection using three bacterially derived immune-enhancing agents known for promoting distinct immunological profiles. BALB/c mice were treated nasally with either cholera toxin (CT), a mutant form of the CT-related Escherichia coli heat-labile enterotoxin designated LT(R192G), or CpG oligodeoxynucleotide. Mice were subsequently challenged with a lethal dose of influenza A/PR/8/34 virus 24 h after the last immunomodulation treatment and either monitored for survival or sacrificed postchallenge for viral and immunological analysis. Treatment with the three immunomodulators prevented or delayed mortality and weight loss, but only CT and LT(R192G) significantly reduced initial lung viral loads as measured by plaque assay. Analysis performed 4 days postinfection indicated that prophylactic treatments with CT, LT(R192G), or CpG resulted in significantly increased numbers of CD4 T cells, B cells, and dendritic cells and altered costimulatory marker expression in the airways of infected mice, coinciding with reduced expression of pulmonary chemokines and the appearance of inducible bronchus-associated lymphoid tissue-like structures in the lungs. Collectively, these results suggest that, despite different immunomodulatory mechanisms, CT, LT(R192G), and CpG induce an initial inflammatory process and enhance the immune response to primary influenza virus challenge while preventing potentially damaging chemokine expression. These studies provide insight into the immunological parameters and immune modulation strategies that have the potential to enhance the nonspecific host response to influenza virus infection.Influenza viruses cause acute, contagious respiratory disease. Despite the availability of vaccines and antiviral therapies, influenza virus infections cause considerable morbidity and mortality each year. It is estimated that during seasonal epidemics 10% of the world population is infected, resulting in 2 to 3 million severe cases and up to 500,000 deaths (1). The failure of conventional methods to prevent illness and death from influenza is attributed to the continuous antigenic variability of the virus due to mutations (antigen drift) and reassortments (antigenic shift). The inadequacy of current anti-influenza virus treatments is particularly concerning in the case of influenza pandemics with new viral strains for which effective vaccines would not be initially available. Thus, an antigen-independent prophylactic treatment that could nonspecifically enhance immune responses to negate or inhibit the progression of influenza virus infection would provide invaluable benefits.Several recent studies have explored the use of immunomodulation strategies as prophylaxis or therapeutic treatments to modify the immune response to influenza virus infection, thereby preventing or decreasing viral burden, disease symptoms, and mortality. These strategies have one of two distinct immunologic goals: either to increase immune system activation and/or Th1 responses specific against influenza virus, or alternatively, decrease inflammation and immunopathology. The first strategy has been demonstrated in animal models by administering host proteins/glycoproteins that function in immune defense, such as the pattern recognition receptor (PRR) mindin (28), milk-derived glycoproteins (61), and virally delivered interferon (IFN) cytokines (27). Immunomodifiers of microbial origin have also been used to enhance host response to infection, including the binding subunit of cholera toxin (CT-B) (49), Th1-promoting Toll-like receptor (TLR) agonists CpG oligodeoxynucleotides (ODN) (15, 82), poly(I:C) (81), 3 M-011 (23), and synthetic lipid A analogs (11). Immunomodulators used in the second strategy, with the aim to prevent detrimental inflammation, have been associated with improved infection outcomes and include enterotoxin mutant LT(S63K) (80) and anti-inflammatory COX-2 inhibitors (84). However, immunomodulation does not always result in beneficial responses to infection. Administration of Δ9-THC, an immunosuppressive compound, decreased cellular infiltration and increased viral load when given prior to and during influenza virus infection (7). Similarly, sphingosine 1-phosphate (S1P) analog, an immunotherapeutic agent, was found to suppress induction of T-cell responses to influenza virus (46). Lastly, fish oil-fed mice demonstrated reduced lung inflammation, cellular infiltration, and cytokine secretion but increased mortality during influenza virus infection (60).These studies highlight the need for experiments that clarify the consequences of various immunomodulation strategies on influenza virus infection and the particular requirements for generating a protective response. Furthermore, very little attention has been given to the mechanisms by which different immunomodulators with unique effector functions modulate the host response when evaluated in the same infection model. To address these questions and increase our understanding of the consequences brought about by prophylactic immunomodulation in pulmonary disease, we chose to compare the effects of pulmonary delivery of three well-characterized vaccine adjuvants on the outcome of influenza virus infection in a murine model. The immunomodulators used in this study are CpG, a nontoxic protein designated LT(R192G) that was derived from the cholera-related heat-labile enterotoxin produced by Escherichia coli, and CT. These bacterially derived agents, known to promote distinct effector functions, are excellent immunomodulators, as they induce strong immune activation and have been previously evaluated as components of influenza vaccines (29, 42, 49, 53, 56, 58). CpG ODNs are synthetic unmethylated oligodeoxynucleotides containing CpG motifs that trigger a TLR9-dependent MyD88 signaling pathway. CpG treatment results in potent Th1 cytokine expression (IFNs and interleukin-12 [IL-12]), activation of dendritic cells (DCs), NK cells, and B cells, and induction of Th1 cells and a Th1 antibody profile (30, 35, 83). CpG has been extensively studied in animal models of systemic and pulmonary infectious diseases caused by influenza virus (15, 82) and other bacterial, fungal, and parasitic pathogens (3, 9, 15, 17, 25, 34, 51, 77).Bacterially derived ADP-ribosylating enterotoxins, including CT from Vibrio cholerae and LT from E. coli, are robust systemic and mucosal adjuvants. Both in vitro and in vivo studies have demonstrated that CT induces secretion of Th2 cytokines (IL-4, IL-5, IL-6, and IL-10) by immune system cells, maturation of DCs, generation of Th2 and T-regulatory cells, and active suppression of Th1 responses (2, 32, 38, 39, 47, 49, 53, 56). Studies in vivo have also shown that intranasal delivery of CT-B, the binding subunit of the enterotoxin, combined with minimal levels of CT holotoxin, induces protective effects in influenza virus-infected mice (49). In contrast to CpG and CT, LT and LT(R192G) induce a more balanced cytokine and antibody subclass profile indicative of a mixed Th1/Th2 immune response (16, 45, 73). LT(R192G) has yet to be evaluated as a prophylactic immunomodulator, but another LT mutant, LT(S63K), has demonstrated some protective effects against influenza virus, respiratory syncytial virus (RSV), and Cryptococcus neoformans infections (80). Although safety concerns limit the use of native enterotoxins for intranasal or intrapulmonary use in humans (54, 76), animal model studies are warranted because they enhance our understanding of the initial responses that can ultimately lead to protection of the host against infection. In addition, the use of these enterotoxins in laboratory research has the potential to be translated into clinical application by using mutated low-toxinogenic derivatives that retain their immunomodulatory properties.In this study we used a comprehensive approach to evaluate the effects of intrapulmonary delivery of three strong immunomodulators prior to influenza virus infection in a murine model. We hypothesized that the unique immunologic effects induced by prophylactic treatment with CT, LT(R192G), or CpG would differentially affect survival, viral loads, and immune responses of BALB/c mice to influenza A/PR/8/34 (H1N1) virus infection. The relevance of this study to influenza virus disease pathogenesis and infectious disease immunomodulation strategies is discussed.  相似文献   

7.
8.
The ectopic overexpression of Bcl-2 restricts both influenza A virus-induced apoptosis and influenza A virus replication in MDCK cells, thus suggesting a role for Bcl-2 family members during infection. Here we report that influenza A virus cannot establish an apoptotic response without functional Bax, a downstream target of Bcl-2, and that both Bax and Bak are directly involved in influenza A virus replication and virus-induced cell death. Bak is substantially downregulated during influenza A virus infection in MDCK cells, and the knockout of Bak in mouse embryonic fibroblasts yields a dramatic rise in the rate of apoptotic death and a corresponding increase in levels of virus replication, suggesting that Bak suppresses both apoptosis and the replication of virus and that the virus suppresses Bak. Bax, however, is activated and translocates from the cytosol to the mitochondria; this activation is required for the efficient induction of apoptosis and virus replication. The knockout of Bax in mouse embryonic fibroblasts blocks the induction of apoptosis, restricts the infection-mediated activation of executioner caspases, and inhibits virus propagation. Bax knockout cells still die but by an alternative death pathway displaying characteristics of autophagy, similarly to our previous observation that influenza A virus infection in the presence of a pancaspase inhibitor leads to an increase in levels of autophagy. The knockout of Bax causes a retention of influenza A virus NP within the nucleus. We conclude that the cell and virus struggle to control apoptosis and autophagy, as appropriately timed apoptosis is important for the replication of influenza A virus.The pathology of influenza A virus infection usually arises from acute lymphopenia and inflammation of the lungs and airway columnar epithelial cells (23, 38). Influenza A virus induces apoptotic death in infected epithelial, lymphocyte, and phagocytic cells, and apoptosis is a source of tissue damage during infection (3, 22, 33) and increased susceptibility to bacterial pathogens postinfection (31). While the induction of apoptosis by influenza A virus has been well documented (4, 19-21, 28, 33, 37), the mechanisms of this interaction are not well understood. Two viral proteins, NS1 and PB1-F2, have been associated with viral killing of cells. NS1, originally characterized as being proapoptotic (34), was later identified as being an interferon antagonist, inhibiting the activation of several key antiviral responses and restricting the apoptotic response to infection (1, 10, 15, 18, 35, 39, 46). In contrast, PB1-F2 induces apoptosis primarily by localizing to the outer mitochondrial membrane, promoting cytochrome c release, and triggering the apoptotic cascade (43). This effect, however, is typically restricted to infected monocytes, leading to the hypothesis that PB1-F2 induces apoptosis specifically to clear the landscape of immune responders (5, 44). Although PB1-F2 activity does not directly manipulate virus replication or virus-induced apoptosis, PB1-F2 localization to the mitochondrial membrane during infection potentiates the apoptotic response in epithelial and fibroblastic cells through tBID signaling with proapoptotic Bcl-2 family protein members Bax and Bak (22, 43, 44).The Bcl-2 protein family consists of both pro- and antiapoptotic members that regulate cytochrome c release during mitochondrion-mediated apoptosis through the formation of pore-like channels in the outer mitochondrial membrane (12, 16). During the initiation of mitochondrion-mediated apoptosis, cytoplasmic Bid is cleaved to form tBID. This, in turn, activates proapoptotic Bax and Bak (40), which drive cytochrome c release and subsequent caspase activation. Bak is constitutively associated with the mitochondrial membrane, whereas inactive Bax is primarily cytosolic, translocating to the outer mitochondrial membrane only after activation (6). The activation of Bax and Bak results in homo- and heterodimer formation at the outer mitochondrial membrane, generating pores that facilitate mitochondrial membrane permeabilization and cytochrome c release (14, 17), leading to caspase activation and the apoptotic cascade (8). Antiapoptotic members of the Bcl-2 protein family, including Bcl-2, inhibit the activation of proapoptotic Bax and Bak primarily by sequestering inactive Bax and Bak monomers via interactions between their BH3 homology domains (7).Bcl-2 expression has been linked to decreased viral replication rates (26). Bcl-2 overexpression inhibits influenza A virus-induced cell death and reduces the titer and spread of newly formed virions (29). The activation of caspase-3 in the absence of sufficient Bcl-2 is critical to the influenza A virus life cycle. Both Bcl-2 expression and the lack of caspase activation during infection lead to the nuclear accumulation of influenza virus ribonucleoprotein (RNP) complexes, thereby leading to the improper assembly of progeny virions and a marked reduction in titers of infectious virus (26, 41, 42, 45).Here we show that influenza A virus induces mitochondrion-mediated (intrinsic-pathway) apoptosis signaled specifically through Bax and that this Bax signaling is essential for the maximum efficiency of virus propagation. In contrast, Bak expression is strongly downregulated during infection. Cells lacking Bak (while expressing Bax) display a much more severe apoptotic phenotype in response to infection and produce infectious virions at a higher rate than the wild type (WT), suggesting that Bak, which can suppress viral replication, is potentially downregulated by the virus. Our results indicate essential and opposing roles for Bax and Bak in both the response of cells to influenza A virus infection and the ability of the virus to maximize its own replicative potential.  相似文献   

9.
10.
11.
12.
13.
The highly pathogenic H5N1 avian influenza virus emerged from China in 1996 and has spread across Eurasia and Africa, with a continuous stream of new cases of human infection appearing since the first large-scale outbreak among migratory birds at Qinghai Lake. The role of wild birds, which are the natural reservoirs for the virus, in the epidemiology of the H5N1 virus has raised great public health concern, but their role in the spread of the virus within the natural ecosystem of free-ranging terrestrial wild mammals remains unclear. In this study, we investigated H5N1 virus infection in wild pikas in an attempt to trace the circulation of the virus. Seroepidemiological surveys confirmed a natural H5N1 virus infection of wild pikas in their native environment. The hemagglutination gene of the H5N1 virus isolated from pikas reveals two distinct evolutionary clades, a mixed/Vietnam H5N1 virus sublineage (MV-like pika virus) and a wild bird Qinghai (QH)-like H5N1 virus sublineage (QH-like pika virus). The amino acid residue (glutamic acid) at position 627 encoded by the PB2 gene of the MV-like pika virus was different from that of the QH-like pika virus; the residue of the MV-like pika virus was the same as that of the goose H5N1 virus (A/GS/Guangdong [GD]/1/96). Further, we discovered that in contrast to the MV-like pika virus, which is nonpathogenic to mice, the QH-like pika virus is highly pathogenic. To mimic the virus infection of pikas, we intranasally inoculated rabbits, a species closely related to pikas, with the H5N1 virus of pika origin. Our findings first demonstrate that wild pikas are mammalian hosts exposed to H5N1 subtype avian influenza viruses in the natural ecosystem and also imply a potential transmission of highly pathogenic avian influenza virus from wild mammals into domestic mammalian hosts and humans.Highly pathogenic avian influenza (HPAI) is an extremely infectious, systemic viral disease that causes a high rate of mortality in birds. HPAI H5N1 viruses are now endemic in avian populations in Southeast Asia and have repeatedly been transmitted to humans (9, 14, 27). Since 2003, the H5N1 subtype has been reported in 391 human cases of influenza and has caused 247 human deaths in 15 countries, leading to greater than 60% mortality among infected individuals (38). Although currently incapable of sustained human-to-human transmission, H5N1 viruses undoubtedly pose a serious threat to public health, as well as to the global economy. Hence, preparedness for such a threat is a global priority (36).Wild birds are considered to be natural reservoirs for influenza A viruses (6, 18, 21, 35, 37). Of the 144 type A influenza virus hemagglutinin-neuraminidase (HA-NA) combinations, 103 have been found in wild birds (5, 7, 17, 37). Since the first HPAI outbreak among migratory wild birds appeared at Qinghai Lake in western China in May 2005 (3, 16, 25, 34, 41), HPAI viruses of the H5N1 subtype have been isolated from poultry throughout Eurasia and Africa. The continued occurrence of human cases has created a situation that could facilitate a pandemic emergence. There is heightened concern that wild birds are a reservoir for influenza A viruses that switch hosts and stably adapt to mammals, including horses, swine, and humans (11, 19, 22, 37).Despite the recent expansion of avian influenza virus (AIV) surveillance and genomic data (5, 17, 20, 21, 33, 40), fundamental questions remain concerning the ecology and evolution of these viruses. Little is known about how terrestrial wild mammals within their natural ecological systems affect HPAI H5N1 epidemiology or about the virus''s effects on public health, though there are many reports of natural and experimental H5N1 virus infection in animals belonging to the taxonomic orders Carnivora (12, 13, 15, 28, 29) and Artiodactyla (15). Herein, we provide the results of our investigation into H5N1 virus infection in wild pikas (Ochotona curzoniae of the order Lagomorpha) within their natural ecological setting. We describe our attempt to trace the circulation of H5N1 viruses and to characterize pika H5N1 influenza virus (PK virus).  相似文献   

14.
Influenza virus-like particles (VLPs) are a promising cell culture-based vaccine, and the skin is considered an attractive immunization site. In this study, we examined the immunogenicity and protective efficacy of influenza VLPs (H1N1 A/PR/8/34) after skin vaccination using vaccine dried on solid microneedle arrays. Coating of microneedles with influenza VLPs using an unstabilized formulation was found to decrease hemagglutinin (HA) activity, whereas inclusion of trehalose disaccharide preserved the HA activity of influenza VLP vaccines after microneedles were coated. Microneedle vaccination of mice in the skin with a single dose of stabilized influenza VLPs induced 100% protection against challenge infection with a high lethal dose. In contrast, unstabilized influenza VLPs, as well as intramuscularly injected vaccines, provided inferior immunity and only partial protection (≤40%). The stabilized microneedle vaccination group showed IgG2a levels that were 1 order of magnitude higher than those of other groups and had the lowest lung viral titers after challenge. Also, levels of recall immune responses, including hemagglutination inhibition titers, neutralizing antibodies, and antibody-secreting plasma cells, were significantly higher after skin vaccination with stabilized formulations. Therefore, our results indicate that HA stabilization, combined with vaccination via the skin using a vaccine formulated as a solid microneedle patch, confers protection superior to that with intramuscular injection and enables potential dose-sparing effects which are reflected by pronounced increases in rapid recall immune responses against influenza virus.Influenza is a major health threat among infectious diseases, posing a significant burden for public health worldwide. Over 200,000 hospitalizations and approximately 36,000 deaths are estimated to occur annually in the United States alone (48, 49). Vaccination is the most cost-effective measure for controlling influenza. However, the influenza vaccine needs to be updated and manufactured every year due to changes in circulating viral strains. Current influenza vaccines rely on egg substrate-based production, a lengthy process with limited capacity that can cause shortages in available vaccine supplies. The recent 2009 outbreak of H1N1 influenza virus is a good example of the urgent need to develop a more effective vaccine platform and vaccination method (38).Influenza virus-like particles (VLPs) have been suggested as a promising alternative candidate to current influenza vaccines. Influenza VLPs are noninfectious particles that mimic the virus in structure and morphology, can be produced using an egg-free cell culture system, and have been shown to be highly immunogenic, inducing protective immunity (9, 15, 19, 27, 35, 41, 42, 44). Most current vaccines are administered intramuscularly to humans in liquid formulations using hypodermic needles or syringes. Another strategy to meet the potential need for mass vaccination would be to develop an effective method for vaccine delivery to the skin (4, 8, 32, 50, 52). The skin is considered an important peripheral immune organ rich in potent immune-inducing cells, including Langerhans cells (LCs), dermal dendritic cells (DCs), and keratinocytes (5, 13, 14, 22). LCs and DCs residing in the epidermal and dermal layers of the skin have been shown to play an important role in antigen processing and presentation following skin immunization (1, 13, 14, 22). Intradermal (ID) vaccination delivering antigens to the dermal layer of the skin has been performed in many clinical studies and have demonstrated dose-sparing effects in some cases (4, 28, 29). Particularly, ID delivery of vaccines might be more effective in the elderly population (50), the highest risk group for influenza epidemics (49). However, ID delivery of vaccines using hypodermic needles is painful and needs highly trained medical personnel. In addition, more frequent local reactions at the injection site were observed after ID delivery. Therefore, a simple and effective approach for vaccination without using hypodermic needles would be highly desirable.To overcome the skin barrier of the outer layer of stratum corneum, solid microneedles were previously coated with inactivated influenza viruses and used to successfully deliver vaccines to the skin, which provided protection comparable to that with conventional intramuscular immunizations (32, 52). Other vaccines have also been delivered using microneedles (17, 17a), but VLPs have never been used this way before. Delivery of a powdered form of inactivated influenza vaccines to the skin has also been demonstrated using a high-speed jet delivery device (10). These previous studies used high doses of vaccines, possibly due to the instability of vaccines in dry formulations.Influenza hemagglutinin (HA) is responsible for attachment of the virus to sialic acid-containing receptors on target cells. However, it is not well understood how functional activity of HA affects the immunogenicity of influenza VLP vaccines. For the first time in this study, we investigated the effect of HA stability, immune responses, and protective efficacies of solid-microneedle VLP vaccines containing H1 HA as a major influenza viral component after delivery to the skin in comparison to results with intramuscular immunization. We found that the functional integrity of HA in influenza VLPs significantly influenced the immunological and protective outcomes for both microneedle and intramuscular vaccination. In addition, we have observed differential outcomes contributing to the protective immunity by the delivery of HA-stabilized VLPs to the skin in terms of the types of immune responses, recall antibody responses, and viral clearance at an early time point after challenge compared to those induced by intramuscular immunization.  相似文献   

15.
Highly pathogenic avian influenza A viruses of the H5N1 subtype continue to circulate in poultry, and zoonotic transmissions are reported frequently. Since a pandemic caused by these highly pathogenic viruses is still feared, there is interest in the development of influenza A/H5N1 virus vaccines that can protect humans against infection, preferably after a single vaccination with a low dose of antigen. Here we describe the induction of humoral and cellular immune responses in ferrets after vaccination with a cell culture-derived whole inactivated influenza A virus vaccine in combination with the novel adjuvant CoVaccine HT. The addition of CoVaccine HT to the influenza A virus vaccine increased antibody responses to homologous and heterologous influenza A/H5N1 viruses and increased virus-specific cell-mediated immune responses. Ferrets vaccinated once with a whole-virus equivalent of 3.8 μg hemagglutinin (HA) and CoVaccine HT were protected against homologous challenge infection with influenza virus A/VN/1194/04. Furthermore, ferrets vaccinated once with the same vaccine/adjuvant combination were partially protected against infection with a heterologous virus derived from clade 2.1 of H5N1 influenza viruses. Thus, the use of the novel adjuvant CoVaccine HT with cell culture-derived inactivated influenza A/H5N1 virus antigen is a promising and dose-sparing vaccine approach warranting further clinical evaluation.Since the first human case of infection with a highly pathogenic avian influenza A virus of the H5N1 subtype in 1997 (9, 10, 37), hundreds of zoonotic transmissions have been reported, with a high case-fatality rate (10, 44). Since these viruses continue to circulate among domestic birds and human cases are regularly reported, it is feared that they will adapt to their new host or exchange gene segments with other influenza A viruses, become transmissible from human to human, and cause a new pandemic. Recently, a novel influenza A virus of the H1N1 subtype emerged. This virus, which originated from pigs, was transmitted between humans efficiently, resulting in the first influenza pandemic of the 21st century (8, 45). Although millions of people have been inoculated with the (H1N1)2009 virus, the case-fatality rate was relatively low compared to that for infections with the H5N1 viruses (11, 31). However, the unexpected pandemic caused by influenza A/H1N1(2009) viruses has further highlighted the importance of rapid availability of safe and effective pandemic influenza virus vaccines. Other key issues for the development of pandemic influenza A virus vaccines include optimal use of the existing (limited) capacity for production of viral antigen and effectiveness against viruses that are antigenically distinct. Ideally, a single administration of a low dose of antigen would be sufficient to induce protective immunity against the homologous strain and heterologous antigenic variant strains. However, since the population at large will be immunologically naïve to a newly introduced virus, high doses of antigen are required to induce protective immunity in unprimed subjects (23, 36). The use of safe and effective adjuvants in pandemic influenza virus vaccines is considered a dose-sparing strategy. Clinical trials evaluating candidate inactivated influenza A/H5N1 virus vaccines showed that the use of adjuvants can increase their immunogenicity and broaden the specificity of the induced antibody responses (2, 7, 19, 23, 27, 36, 41). These research efforts have resulted in the licensing of adjuvanted vaccines against seasonal and pandemic influenza viruses (17). The protective efficacy of immune responses induced with candidate influenza A/H5N1 virus vaccines was demonstrated in ferrets after two immunizations (1, 22, 24, 25) or after a single immunization. The latter was achieved with a low dose of antigen in combination with the adjuvant Iscomatrix (26).Recently, a novel adjuvant that consists of a sucrose fatty acid sulfate ester (SFASE) immobilized on the oil droplets of a submicrometer emulsion of squalane in water has been developed (4). It has been demonstrated that the addition of this novel adjuvant, called CoVaccine HT, to multiple antigens increased the immune response to these antigens in pigs and horses and was well tolerated in both species (4, 16, 40). Furthermore, it was shown that the use of CoVaccine HT increased the virus-specific antibody responses in mice and ferrets after vaccination with a cell culture-derived whole inactivated influenza A/H5N1 virus vaccine (5, 13). One of the mode of actions of CoVaccine HT is the activation of antigen-presenting cells such as dendritic cells, most likely through Toll-like receptor 4 (TLR4) signaling (5).In the present study, we evaluated the protective potential of CoVaccine HT-adjuvanted cell culture-derived whole inactivated influenza A/H5N1 virus (WIV) vaccine in the ferret model, which is considered the most suitable animal model for the evaluation of candidate influenza virus vaccines (6, 14, 15). To this end, ferrets were vaccinated once or twice with various antigen doses with or without the adjuvant to test whether dose sparing could be achieved. The use of CoVaccine HT increased virus-specific antibody responses and T cell responses. A single administration of 3.8 μg hemagglutinin (HA) of WIV NIBRG-14 vaccine preparation in combination with CoVaccine HT conferred protection against challenge infection with the homologous highly pathogenic A/H5N1 virus strain A/VN/1194/04 and partial protection against infection with a heterologous, antigenically distinct strain, A/IND/5/05. Therefore, it was concluded that the use of CoVaccine HT in inactivated influenza virus vaccines induced protective virus-specific humoral and cell-mediated immune responses and that it could be suitable as adjuvant in (pre)pandemic A/H5N1 virus vaccines. Further clinical testing of these candidate vaccines seems to be warranted.  相似文献   

16.
17.
H2 influenza viruses have not circulated in humans since 1968, and therefore a large segment of the population would likely be susceptible to infection should H2 influenza viruses reemerge. The development of an H2 pandemic influenza virus vaccine candidate should therefore be considered a priority in pandemic influenza preparedness planning. We selected a group of geographically and temporally diverse wild-type H2 influenza viruses and evaluated the kinetics of replication and compared the ability of these viruses to induce a broadly cross-reactive antibody response in mice and ferrets. In both mice and ferrets, A/Japan/305/1957 (H2N2), A/mallard/NY/1978 (H2N2), and A/swine/MO/2006 (H2N3) elicited the broadest cross-reactive antibody responses against heterologous H2 influenza viruses as measured by hemagglutination inhibition and microneutralization assays. These data suggested that these three viruses may be suitable candidates for development as live attenuated H2 pandemic influenza virus vaccines.Influenza pandemics occur when a novel influenza virus enters a population with little preexisting immunity (36). During the pandemics of the last century, novel influenza viruses were introduced either directly from an avian reservoir (34) or were the result of reassortment between contemporaneously circulating human, avian, and swine influenza viruses (5, 29, 36). Due to the lack of preexisting immunity to the novel virus, morbidity and mortality rates are typically higher than in epidemics caused by seasonal influenza viruses (4).Although pandemic preparedness planning has largely focused on the highly pathogenic H5 and H7 avian influenza virus subtypes, the recent emergence of the 2009 pandemic H1N1 viruses underscores the need to consider other influenza virus subtypes as well. Of the 16 hemagglutinin (HA) influenza A virus subtypes that have been identified to date, H1, H2, and H3 have been known to cause influenza pandemics (7, 27), suggesting that these viruses are capable of sustained transmission and can cause disease in humans. While the H1 and H3 subtypes have cocirculated in humans since 1977, H2 influenza viruses have not circulated in humans since 1968 (36) and therefore a large segment of the population would likely be susceptible to infection should H2 influenza viruses reemerge. The 1957 H2 pandemic virus was a reassortant that derived the HA, neuraminidase (NA), and PB1 genes from an avian virus and the remaining gene segments from the circulating H1N1 virus (15, 30). As H2 subtype viruses continue to circulate in avian reservoirs worldwide (12, 17, 18, 22, 33), they remain a potential pandemic threat. The development of an H2 influenza virus vaccine candidate should therefore be considered a priority in future pandemic influenza preparedness planning.Given the low likelihood that a previously selected vaccine virus will exactly match the pandemic virus, the ability to elicit a broadly cross-reactive antibody response to antigenically distinct viruses within a subtype is an important consideration in the selection of a pandemic influenza vaccine candidate. Previous studies have examined the ability of inactivated H2 influenza viruses to provide cross-protection against mouse-adapted variants of reassortant human viruses and an avian H2 influenza virus from 1978 (9, 14). Given the potential for live attenuated influenza virus vaccines to confer a great breadth of heterologous cross-protection (1, 2, 6, 35), we recently conducted a study evaluating cold-adapted A/Ann Arbor/6/1960 (AA CA), an H2 influenza virus used as the backbone of the seasonal live attenuated influenza A virus vaccine currently licensed in the United States (3). However, as H2 influenza virus continues to circulate widely and appear in migratory birds (10, 24, 26), in poultry markets (20), and in swine (21), with evidence of interregional gene transmission (19, 22), a more extensive evaluation of recent isolates may be warranted in the selection of a potential H2 pandemic vaccine candidate.H2 influenza viruses fall into three main lineages: a human lineage, a North American avian lineage, and a Eurasian avian lineage (29). In addition to viruses whose replicative ability in mammals has previously been established (11, 21, 23, 25), we selected a group of geographically and temporally diverse H2 influenza viruses from each lineage. We evaluated the kinetics of replication of each of these viruses in mice and ferrets and compared the abilities of these viruses to induce a broadly cross-reactive antibody response to determine which of these viruses would be suitable for further development as an H2 pandemic influenza vaccine candidate.  相似文献   

18.
Mature glycoprotein spikes are inserted in the Lassa virus envelope and consist of the distal subunit GP-1, the transmembrane-spanning subunit GP-2, and the signal peptide, which originate from the precursor glycoprotein pre-GP-C by proteolytic processing. In this study, we analyzed the oligomeric structure of the viral surface glycoprotein. Chemical cross-linking studies of mature glycoprotein spikes from purified virus revealed the formation of trimers. Interestingly, sucrose density gradient analysis of cellularly expressed glycoprotein showed that in contrast to trimeric mature glycoprotein complexes, the noncleaved glycoprotein forms monomers and oligomers spanning a wide size range, indicating that maturation cleavage of GP by the cellular subtilase SKI-1/S1P is critical for formation of the correct oligomeric state. To shed light on a potential relation between cholesterol and GP trimer stability, we performed cholesterol depletion experiments. Although depletion of cholesterol had no effect on trimerization of the glycoprotein spike complex, our studies revealed that the cholesterol content of the viral envelope is important for the infectivity of Lassa virus. Analyses of the distribution of viral proteins in cholesterol-rich detergent-resistant membrane areas showed that Lassa virus buds from membrane areas other than those responsible for impaired infectivity due to cholesterol depletion of lipid rafts. Thus, derivation of the viral envelope from cholesterol-rich membrane areas is not a prerequisite for the impact of cholesterol on virus infectivity.Lassa virus (LASV) is a member of the family Arenaviridae, of which Lymphocytic choriomeningitis virus (LCMV) is the prototype. Arenaviruses comprise more than 20 species, divided into the Old World and New World virus complexes (19). The Old World arenaviruses include the human pathogenic LASV strains, Lujo virus, which was first identified in late 2008 and is associated with an unprecedented high case fatality rate in humans, the nonhuman pathogenic Ippy, Mobala, and Mopeia viruses, and the recently described Kodoko virus (10, 30, 49). The New World virus complex contains, among others, the South American hemorrhagic fever-causing viruses Junín virus, Machupo virus, Guanarito virus, Sabiá virus, and the recently discovered Chapare virus (22).Arenaviruses contain a bisegmented single-stranded RNA genome encoding the polymerase L, matrix protein Z, nucleoprotein NP, and glycoprotein GP. The bipartite ribonucleoprotein of LASV is surrounded by a lipid envelope derived from the plasma membrane of the host cell. The matrix protein Z has been identified as a major budding factor, which lines the interior of the viral lipid membrane, in which GP spikes are inserted (61, 75). The glycoprotein is synthesized as precursor protein pre-GP-C and is cotranslationally cleaved by signal peptidase into GP-C and the signal peptide, which exhibits unusual length, stability, and topology (3, 27, 28, 33, 70, 87). Moreover, the arenaviral signal peptide functions as trans-acting maturation factor (2, 26, 33). After processing by signal peptidase, GP-C of both New World and Old World arenaviruses is cleaved by the cellular subtilase subtilisin kexin isozyme-1/site-1 protease (SKI-1/S1P) into the distal subunit GP-1 and the membrane-anchored subunit GP-2 within the secretory pathway (5, 52, 63). For LCMV, it has been shown that GP-1 subunits are linked to each other by disulfide bonds and are noncovalently connected to GP-2 subunits (14, 24, 31). GP-1 is responsible for binding to the host cell receptor, while GP-2 mediates fusion between the virus envelope and the endosomal membrane at low pH due to a bipartite fusion peptide near the amino terminus (24, 36, 44). Sequence analysis of the LCMV GP-2 ectodomain revealed two heptad repeats that most likely form amphipathic helices important for this process (34, 86).In general, viral class I fusion proteins have triplets of α-helical structures in common, which contain heptad repeats (47, 73). In contrast, class II fusion proteins are characterized by β-sheets that form dimers in the prefusion status and trimers in the postfusion status (43). The class III fusion proteins are trimers that, unlike class I fusion proteins, were not proteolytically processed N-terminally of the fusion peptide, resulting in a fusion-active membrane-anchored subunit (39, 62). Previous studies with LCMV described a tetrameric organization of the glycoprotein spikes (14), while more recent data using a bacterially expressed truncated ectodomain of the LCMV GP-2 subunit pointed toward a trimeric spike structure (31). Due to these conflicting data regarding the oligomerization status of LCMV GP, it remains unclear to which class of fusion proteins the arenaviral glycoproteins belong.The state of oligomerization and the correct conformation of viral glycoproteins are crucial for membrane fusion during virus entry. The early steps of infection have been shown for several viruses to be dependent on the cholesterol content of the participating membranes (i.e., either the virus envelope or the host cell membrane) (4, 9, 15, 20, 21, 23, 40, 42, 53, 56, 76, 78, 79). In fact, it has been shown previously that entry of both LASV and LCMV is susceptible to cholesterol depletion of the target host cell membrane using methyl-β-cyclodextrin (MβCD) treatment (64, 71). Moreover, cholesterol not only plays an important role in the early steps during entry in the viral life cycle but also is critical in the virus assembly and release process. Several viruses of various families, including influenza virus, human immunodeficiency virus type 1 (HIV-1), measles virus, and Ebola virus, use the ordered environment of lipid raft microdomains. Due to their high levels of glycosphingolipids and cholesterol, these domains are characterized by insolubility in nonionic detergents under cold conditions (60, 72). Recent observations have suggested that budding of the New World arenavirus Junin virus occurs from detergent-soluble membrane areas (1). Assembly and release from distinct membrane microdomains that are detergent soluble have also been described for vesicular stomatitis virus (VSV) (12, 38, 68). At present, however, it is not known whether LASV requires cholesterol in its viral envelope for successful virus entry or whether specific membrane microdomains are important for LASV assembly and release.In this study, we first investigated the oligomeric state of the premature and mature LASV glycoprotein complexes. Since it has been shown for several membrane proteins that the oligomerization and conformation are dependent on cholesterol (58, 59, 76, 78), we further analyzed the dependence of the cholesterol content of the virus envelope on glycoprotein oligomerization and virus infectivity. Finally, we characterized the lipid membrane areas from which LASV is released.  相似文献   

19.
Poxviruses produce complement regulatory proteins to subvert the host''s immune response. Similar to the human pathogen variola virus, ectromelia virus has a limited host range and provides a mouse model where the virus and the host''s immune response have coevolved. We previously demonstrated that multiple components (C3, C4, and factor B) of the classical and alternative pathways are required to survive ectromelia virus infection. Complement''s role in the innate and adaptive immune responses likely drove the evolution of a virus-encoded virulence factor that regulates complement activation. In this study, we characterized the ectromelia virus inhibitor of complement enzymes (EMICE). Recombinant EMICE regulated complement activation on the surface of CHO cells, and it protected complement-sensitive intracellular mature virions (IMV) from neutralization in vitro. It accomplished this by serving as a cofactor for the inactivation of C3b and C4b and by dissociating the catalytic domain of the classical pathway C3 convertase. Infected murine cells initiated synthesis of EMICE within 4 to 6 h postinoculation. The levels were sufficient in the supernatant to protect the IMV, upon release, from complement-mediated neutralization. EMICE on the surface of infected murine cells also reduced complement activation by the alternative pathway. In contrast, classical pathway activation by high-titer antibody overwhelmed EMICE''s regulatory capacity. These results suggest that EMICE''s role is early during infection when it counteracts the innate immune response. In summary, ectromelia virus produced EMICE within a few hours of an infection, and EMICE in turn decreased complement activation on IMV and infected cells.Poxviruses encode in their large double-stranded DNA genomes many factors that modify the immune system (30, 56). The analysis of these molecules has revealed a delicate balance between viral pathogenesis and the host''s immune response (2, 21, 31, 61). Variola, vaccinia, monkeypox, cowpox, and ectromelia (ECTV) viruses each produce an orthologous complement regulatory protein (poxviral inhibitor of complement enzymes [PICE]) that has structural and functional homology to host proteins (14, 29, 34, 38, 41, 45, 54). The loss of the regulatory protein resulted in smaller local lesions with vaccinia virus lacking the vaccinia virus complement control protein (VCP) (29) and in a greater local inflammatory response in the case of cowpox lacking the inflammation-modulatory protein (IMP; the cowpox virus PICE) (35, 45, 46). Additionally, the complete loss of the monkeypox virus inhibitor of complement enzymes (MOPICE) may account for part of the reduced mortality observed in the West African compared to Congo basin strains of monkeypox virus (12).The complement system consists of proteins on the cell surface and in blood that recognize and destroy invading pathogens and infected host cells (36, 52). Viruses protect themselves from the antiviral effects of complement activation in a variety of ways, including hijacking the host''s complement regulatory proteins or producing their own inhibitors (7, 8, 15, 20, 23). Another effective strategy is to incorporate the host''s complement regulators in the outermost viral membrane, which then protects the virus from complement attack (62). The extracellular enveloped virus (EEV) produced by poxviruses acquires a unique outer membrane derived from the Golgi complex or early endosomes that contain the protective host complement regulators (58, 62). Poxviruses have multiple infectious forms, and the most abundant, intracellular mature virions (IMV), are released when infected cells lyse (58). The IMV lacks the outermost membrane found on EEV and is sensitive to complement-mediated neutralization. The multiple strategies viruses have evolved to evade the complement system underscore its importance to innate and adaptive immunity (15, 36).The most well-characterized PICE is VCP (24-29, 34, 49, 50, 53, 55, 59, 60). Originally described as a secreted complement inhibitor (34), VCP also attaches to the surface of infected cells through an interaction with the viral membrane protein A56 that requires an unpaired N-terminal cysteine (26). This extra cysteine also adds to the potency of the inhibitor by forming function-enhancing dimers (41). VCP and the smallpox virus inhibitor of complement enzymes (SPICE) bind heparin in vitro, and this may facilitate cell surface interactions (24, 38, 50, 59). The coevolution of variola virus with its only natural host, humans, likely explains the enhanced activity against human complement observed with SPICE compared to the other PICEs (54, 64).Our recent work with ECTV, the causative agent of mousepox infection, demonstrated that the classical and alternative pathways of the complement system are required for host survival (48). The mouse-specific pathogen ECTV causes severe disease in most strains and has coevolved with its natural host, analogous to variola virus in humans (9). This close host-virus relationship is particularly important for evaluating the role of the complement system, given the species specificity of many complement proteins, receptors, and regulators (10, 47, 62). Additionally, the availability of complement-deficient mice permits dissection of the complement activation pathways involved. Naïve C57BL/6 mouse serum neutralizes the IMV of ECTV in vitro, predominately through opsonization (48). Maximal neutralization requires natural antibody, classical-pathway activation, and amplification by the alternative pathway. C3 deficiency in the normally resistant C57BL/6 strain results in acute mortality, similar to immunodeficiencies in important elements of the antiviral immune response, including CD8+ T cells (19, 32), natural killer cells (18, 51), and gamma interferon (33). During ECTV infection, the complement system acts in the first few hours and days to delay the spread of infection, resulting in lower levels of viremia and viral burden in tissues (48).This study characterized the PICE produced by ECTV, ectromelia virus inhibitor of complement enzymes (EMICE), and assessed its complement regulatory activity. Recombinant EMICE (rEMICE) decreased activation of both human and mouse complement. Murine cells produced EMICE at 4 to 6 h postinfection prior to the release of the majority of the complement-sensitive IMV from infected cells. rEMICE protected ECTV IMV from complement-mediated neutralization. Further, EMICE produced during natural infection inhibited complement deposition on infected cells by the alternative pathway. ECTV likely produces this abundance of EMICE to protect both the IMV and infected cells.  相似文献   

20.
The specificity of the CD4 T-cell immune response to influenza virus is influenced by the genetic complexity of the virus and periodic encounters with variant subtypes and strains. In order to understand what controls CD4 T-cell reactivity to influenza virus proteins and how the influenza virus-specific memory compartment is shaped over time, it is first necessary to understand the diversity of the primary CD4 T-cell response. In the study reported here, we have used an unbiased approach to evaluate the peptide specificity of CD4 T cells elicited after live influenza virus infection. We have focused on four viral proteins that have distinct intracellular distributions in infected cells, hemagglutinin (HA), neuraminidase (NA), nucleoprotein, and the NS1 protein, which is expressed in infected cells but excluded from virion particles. Our studies revealed an extensive diversity of influenza virus-specific CD4 T cells that includes T cells for each viral protein and for the unexpected immunogenicity of the NS1 protein. Due to the recent concern about pandemic avian influenza virus and because CD4 T cells specific for HA and NA may be particularly useful for promoting the production of neutralizing antibody to influenza virus, we have also evaluated the ability of HA- and NA-specific CD4 T cells elicited by a circulating H1N1 strain to cross-react with related sequences found in an avian H5N1 virus and find substantial cross-reactivity, suggesting that seasonal vaccines may help promote protection against avian influenza virus.In recent decades, investigators studying both murine and human T-cell responses to influenza virus have succeeded in identifying peptide epitopes from immunized or vaccinated individuals that are the targets of CD4 T cells. These studies suggest a considerable diversity in CD4 responses. Epitopes derived from hemagglutinin (HA), neuraminidase (NA), nuclear protein (NP), polymerase (PB1 and PB2), matrix (M1), and nonstructural protein (NS1) have all been identified (9, 19, 25-28, 32, 61, 64, 85, 86). Our own laboratory previously analyzed the peptide specificity of CD4 T cells in the primary response of HLA-DR1 transgenic mice toward a human isolate of influenza virus and found that the CD4 T-cell repertoire specific for HA alone was diverse and encompassed at least 30 different peptide epitopes (63). In general, studies with humans have been much less systematic than those with the mouse because of the difficulty in obtaining lymphocyte samples from recently infected individuals and because of the complexity of major histocompatibility complex (MHC) molecules expressed in humans. However, recent studies with MHC class II tetramer reagents (19, 61, 64, 72, 86) have permitted the visualization of CD4 T cells specific for influenza virus directly ex vivo or after a brief (10- to 14-day) in vitro expansion. Those studies have led to the conclusion that the repertoire of CD4 T cells is more diverse than that of CD8 T cells and that CD4 T cells that are specific for most influenza virus proteins can be detected.We have focused on the identification of the peptide specificity of CD4 T cells during the primary response to influenza virus infection using HLA-DR1 transgenic mice with several goals in mind. First, we seek to understand the intracellular events within influenza virus-infected antigen-presenting cells (APC) that shape the repertoire of the peptide:class II complexes expressed, because these events will play a pivotal role in determining the specificity of the anti-influenza virus CD4 T-cell response. Second, we expect these studies to provide significant new insight into the CD4 T-cell antigen repertoire that becomes established upon natural infection of humans with influenza virus. Finally, because HLA-DR1 is widely expressed in human populations, the results of our experiments and the corresponding peptide epitopes identified can immediately be utilized for analyses of human immune responses to influenza viruses and vaccines.Our work (45, 57, 60, 68, 69) and the works of others (1, 18, 51, 58, 65, 71, 73, 75) regarding CD4 T-cell immunodominance in response to exogenous antigens indicate that CD4 T cells tend to focus on a limited number of peptides. Typical protein antigens that are taken up as a “pulse” by peripheral APC lead to CD4 T-cell priming that is very narrow in specificity, limited to usually only a few (less than five) epitopes. Our mechanistic studies (44, 68, 69) further indicate that immunodominant peptides characteristically display high-stability interactions with the MHC class II molecule. This selectivity in CD4 T-cell responses is at least in part due to DM editing within APC, where DM apparently removes the peptides that have low-stability interactions with class II molecules (44). Therefore, only a limited subset of antigenic peptides arrives at the cell surface at a sufficient density to recruit CD4 T cells.The characteristics of influenza virus infection suggest that the immunodominance hierarchy might not follow the “rules” established for exogenous protein antigens. Because influenza virus is typically not a systemic infection, virus replication is normally restricted to the lung (3, 29, 33, 59). Therefore, the primary source of viral antigens available for CD4 T-cell priming may not be free virus particles but, rather, may be dendritic cells that become infected with influenza virus while in the lung and then migrate to the draining lymph node (4, 5, 33, 35, 48, 52). If so, then one might predict that the specificity of CD4 T cells could more closely resemble the repertoire that is elicited by “endogenous” antigens synthesized within the APC (21). Endogenous antigens that have ready access to the endosomally localized MHC class II molecules, because they are either membrane associated or secreted, are most efficiently presented by class II molecules (46, 53, 67, 84). For the influenza virus-infected dendritic cell, these preferences in antigen access would favor the presentation of peptides derived from HA and NA, leading to the selective priming of CD4 T cells that are reactive to these viral proteins.Several critical questions remain with regard to the specificity of CD4 T cells that are elicited in response to influenza virus infection. The first question is how diverse the repertoire is, with regard to both peptide and protein specificities. The second issue is how the CD4 T-cell repertoire changes over time with repeated encounters with different strains of influenza virus, a common occurrence in humans. A final, very important question is whether CD4 T cells elicited during the primary response have equivalent potentials to promote protection against subsequent infection or if this potential is dependent on their antigen specificities. It is thought that the primary contribution of CD4 T cells to protective immunity is their role in facilitating the production of high-affinity neutralizing antibodies to HA and NA (38, 79). Recent studies by Sette and coworkers (74) suggest that for complex viral pathogens, the delivery of CD4 T-cell help for the production of high-affinity antibodies by B cells may require that the CD4 T cells share viral antigen specificity with the B cells. For influenza virus, the most useful CD4 T cells may therefore be those that are specific for the membrane glycoproteins HA and NA.In the study reported here, we use an unbiased and comprehensive approach to evaluate the peptide specificity of CD4 T cells elicited after live influenza virus infection. We have focused on four viral proteins that have distinct intracellular distributions in infected cells: HA and NA, expressed at the plasma membrane of infected cells and on the exterior of the virion membrane; NP, expressed in the cytoplasm and nucleus of infected cells; and, finally, the NS1 protein, with a distribution similar to that of NP in infected cells but which is excluded from the virion particles. Our studies lead to the conclusion that influenza virus-specific CD4 T cells elicited during the primary response are distributed across all proteins studied and that the NS1 protein is particularly immunogenic. Because of the recent concern about pandemic avian influenza virus and because CD4 T cells specific for HA and NA may be particularly useful for promoting the production of neutralizing antibody, we have also evaluated the ability of HA- and NA-specific CD4 T cells elicited against a circulating H1N1 strain of influenza virus to cross-react with related sequences found in an H5N1 avian virus. We find that priming with an H1N1 virus elicits CD4 T cells that display a significant degree of cross-reactivity with influenza virus epitopes derived from avian viruses.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号