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 15 毫秒
1.
Genital herpes, caused by herpes simplex virus type 2 (HSV-2), is one of the most prevalent sexually transmitted diseases worldwide and a risk factor for acquiring human immunodeficiency virus. Although many vaccine candidates have shown promising results in animal models, they have failed to be effective in human trials. In this study, a humanized mouse strain was evaluated as a potential preclinical model for studying human immune responses to HSV-2 infection and vaccination. Immunodeficient mouse strains were examined for their abilities to develop human innate and adaptive immune cells after transplantation of human umbilical cord stem cells. A RAG2−/− γc−/− mouse strain with a BALB/c background was chosen as the most appropriate model and was then examined for its ability to mount innate and adaptive immune responses to intravaginal HSV-2 infection and immunization. After primary infection, human cells in the lymph nodes were able to generate a protective innate immune response and produce gamma interferon (IFN-γ). After intravaginal immunization and infection, human T cells and NK cells were found in the genital tract and iliac lymph nodes. In addition, human T cells in the spleen, lymph nodes, and vaginal tract were able to respond to stimulation with HSV-2 antigens by replicating and producing IFN-γ. Human B cells were also able to produce HSV-2-specific immunoglobulin G. These adaptive responses were also shown to be protective and reduce local viral replication in the genital tract. This approach provides a means for studying human immune responses in vivo using a small-animal model and may become an important preclinical tool.Genital herpes, caused primarily by herpes simplex virus type 2 (HSV-2), is one of the most prevalent sexually transmitted diseases in the world and is associated with substantial morbidity (13). After initial infection of the genital tract, the virus establishes latency within the nervous system and thus maintains lifelong infection in humans. Latent virus can reactivate and cause recurrent symptoms, including genital lesions; however, subclinical infection and asymptomatic viral shedding also occur (11, 35, 40, 53). HSV-2 has gained increasing interest in the light of evidence that it is a major risk factor for human immunodeficiency virus type 1 (HIV-1) acquisition and transmission and for the progression of HIV-1 infection (8, 9, 17, 25, 37, 55, 56). In addition, there is evidence that anti-HSV therapy can reduce the amount of infectious HIV-1 in the genital tracts of women (9, 45). Although antiviral treatment is available and can reduce the severity of the infection, compliance problems, as well as difficulty in diagnosing infection in patients, have hampered efforts to control the disease. A vaccine would provide a more effective way of preventing or limiting infection and would therefore greatly reduce the social and economic burdens caused by HSV-2 infection.Several vaccine candidates exist; however, they have proven to be less successful in clinical trials than anticipated, and new strategies may need to be developed (24, 61). A key concern is that preclinical vaccine strategies have been evaluated largely by using studies performed with mouse models of HSV-2 infection and, thus, the immune responses observed were mediated by murine cells. As a consequence, the results of these studies may not accurately represent the human immune response to infection. In order to develop an effective vaccine and/or treatment, it is necessary to understand which immune mechanisms provide protection against infection at the site of viral entry, the vaginal tract, and how these immune responses can be induced in humans.Innate and adaptive immune responses are both important for controlling HSV-2 infection. Innate immune cells such as NK and NKT cells are required for protection against genital HSV-2 infection in mice (1) and in humans; NK cells accumulate at sites of HSV-2 infection and can lyse HSV-infected cells (30, 67). Adaptive immune responses to HSV-2 include the cellular response mediated by CD4+ and CD8+ T cells and the humoral response mediated by B cells and antibodies. There is much evidence that T cells play a crucial role in protection against HSV-2 in mice and humans (28). T cells are present in herpes lesions, and depletion of T cells in mice greatly reduces protection (16, 27, 29, 30, 44, 51, 70). Gamma interferon (IFN-γ), which is produced early after infection by NK cells and later by CD4+ T cells, has been shown to be a crucial cytokine for the control of HSV (43, 52, 58, 63). Although HSV-2-specific antibodies are produced in response to infection and vaccination, a correlation with protection in humans has not been established (2, 3, 7, 10, 11, 48). In mice, a role for antibodies early after infection has been shown; however, if B cells are knocked out, mice are still able to eventually clear the virus (16, 50). Although we do not have a complete understanding of the components that are necessary for protection, it appears that both innate and adaptive immune responses will be required and that it will be important to elicit these responses at the site of infection in the genital tract.The lack of an effective vaccine and accurate translation of results obtained with mice to humans indicates a need for a more relevant preclinical model to study human immune responses and disease. Substantial improvements in the development of humanized mice have made them a novel tool for the study of human diseases (69). Human CD34+ stem cells have been injected into several immunodeficient mouse strains, such as NOD/SCID/γc−/− and RAG2−/− γc−/− mice, in which superior engraftment has resulted in multilineage differentiation of the human cells (23, 64). These novel humanized mice have been shown to develop human immune responses to pathogens such as Epstein-Barr virus, dengue virus, and influenza virus and to immunization with cholera toxin (33, 64, 66, 68). In addition, humanized mice can support infection with HIV after systemic or mucosal challenge in the vaginal tract and rectum (4-6, 62, 65). HSV-2 infection in humanized mice has not been examined, and mucosal immunization that can provide protection from infection with wild-type virus has also not been demonstrated. In addition, although it is clear that adaptive immune responses can be generated in humanized mice, innate responses to viral infection have not been extensively examined.In this study, we evaluated three immunodeficient mouse strains for their abilities to engraft human umbilical cord-derived stem cells and support the differentiation of these cells into important innate and adaptive immune cells. The most appropriate model was then used to examine mucosal immune responses following primary HSV-2 infection, immunization, and secondary HSV-2 challenge. We show for the first time that the humanized mice can mount protective human NK cell-mediated innate immune responses to primary mucosal infection with HSV-2. In addition, mucosal immunization and infection can induce HSV-2-specific antibody production and, to a greater extent, T-cell-mediated responses both systemically and locally in the genital tracts of humanized mice. We further show that mucosal immunization can provide protection against a lethal intravaginal (IVAG) challenge with HSV-2.  相似文献   

2.
Glycoprotein B (gB) of herpes simplex virus (HSV) is one of four glycoproteins essential for viral entry and cell fusion. Recently, paired immunoglobulin-like type 2 receptor (PILRα) was identified as a receptor for HSV type 1 (HSV-1) gB. Both PILRα and a gD receptor were shown to participate in HSV-1 entry into certain cell types. The purpose of this study was to determine whether insertional mutations in gB had differential effects on its function with PILRα and the gD receptor, nectin-1. Previously described gB mutants and additional newly characterized mutants were used in this study. We found that insertional mutations near the N terminus and C terminus of gB and especially in the central region of the ectodomain reduced cell fusion activity when PILRα was overexpressed much more than when nectin-1 was overexpressed. Most of the insertions reduced the binding of gB to PILRα, for at least some forms of gB, but this reduction did not necessarily correlate with the selective reduction in cell fusion activity with PILRα. These results suggest that the regions targeted by the relevant mutations are critical for functional activity with PILRα. They also suggest that, although both the binding of gB to a gB receptor and the binding of gD to a gD receptor may be required for HSV-induced cell fusion, the two receptor-binding activities may have unequal weights in triggering fusogenic activity, depending on the ratios of gB and gD receptors or other factors.Manifestations of disease caused by herpes simplex virus (HSV) include recurrent mucocutaneous lesions in the mouth or on the face or genitalia and, more rarely, meningitis or encephalitis. The infection of host cells occurs by the fusion of the virion envelope with a cell membrane to deliver the nucleocapsid containing the viral genome into the host cell. This entry process and virus-induced cell fusion require glycoprotein B (gB), along with gD, gH, and gL. The membrane-fusing activity of HSV depends in part on the binding of gD to one of its receptors, herpesvirus entry mediator (HVEM), nectin-1, nectin-2, or 3-O-sulfated heparan sulfate (18). HVEM is a member of the tumor necrosis factor receptor family and is expressed by cells of the immune system, as well as many other cell types, such as epithelial, stromal, and endothelial cells (23). Nectin-1 and nectin-2 are cell adhesion molecules belonging to the immunoglobulin superfamily and are widely expressed by a variety of cell types, including epithelial cells and neurons (20). Specific sites in heparan sulfate generated by particular 3-O-sulfotransferases can serve as gD-binding entry receptors (17). This binding of gD to a receptor is associated with conformational changes in gD that are thought to enable gD to interact with gB and/or the heterodimer gH-gL to trigger fusogenic activity (8, 12). Both gB and gH have properties of fusogenic viral proteins (1, 7). Although evidence has been presented that gD and gH-gL are sufficient for hemifusion and that gB, in addition, is required for fusion pore formation (19), the specific roles each plays in HSV-induced membrane fusion have not been fully defined.gB was recently discovered to bind to paired immunoglobulin-like type 2 receptor (PILRα) in an interaction that can mediate viral entry and cell fusion, provided that gD also binds to one of its receptors (14). For cells such as CD14+ monocytes, antibodies specific for either HVEM or PILRα were shown to block HSV entry. Also, entry requires the presence of both gD and gB in the virion. Although the overexpression of either a gD receptor or a gB receptor can enhance the susceptibility of cells to HSV entry and HSV-induced cell fusion, there are very few, if any, cell types that do not express at least low levels of endogenous receptors. Thus, the possibility exists that these endogenous receptors are cooperating with the introduced receptors to render the cells susceptible to HSV-induced membrane fusion.PILRα belongs to the paired-receptor families, which consist of activating and inhibitory receptors (4, 11, 19). They are conserved among mammals (24). The inhibitory form PILRα has an immunoreceptor tyrosine-based inhibition motif in its cytoplasmic domain and transduces inhibitory signals (4). On the other hand, the activating form PILRβ associates with the immunoreceptor tyrosine-based activation motif-bearing DAP12 adaptor molecule and delivers activating signals (16). Both PILRα and PILRβ are expressed on cells of the immune system, especially monocytes, dendritic cells, and NK cells (4, 11, 19), and also in neurons (14). CD99 has been identified as a natural ligand for both PILRα and PILRβ (16). The binding of either PILRα or PILRβ to CD99 depends on the presence of sialyated O-linked glycans on CD99 (22).In addition to binding to PILRα, gB can bind to heparin and heparan sulfate and may contribute, along with gC, to the binding of HSV to cell surface heparan sulfate (17). Also, gB and gC can bind to DC-SIGN, which serves as a binding receptor for the infection of dendritic cells (2). An X-ray structure of the HSV-1 gB ectodomain reveals a homotrimeric conformation with structural homology to vesicular stomatitis virus (VSV) G glycoprotein, the single glycoprotein responsible for the entry of VSV. Both HSV-1 gB and VSV G glycoprotein have features of class 1 and class 2 viral fusion proteins and have been designated class 3 fusion proteins (7, 14, 15). The heparan sulfate-binding determinant of gB has been localized to a lysine-rich domain in the N terminus and shown to be dispensable for viral entry (9). It lies within a region that is probably disordered and was not included in the defined coordinates of the X-ray structure. The binding of DC-SIGN to gB probably depends on high-mannose N-glycans of gB (6).In a previous study (10), 81 insertion mutants of HSV-1 gB were characterized to assess the effects of the insertions on protein processing and function in cell fusion with gD receptors, in relation to structural domains of gB identified in an X-ray structure (7). Only 27 mutants were found to be processed into mature glycosylated forms and transported to the cell surface. Only 11 of these retained fusion activity toward target cells expressing nectin-1 or HVEM. For the present study, we used 25 previously described gB insertion mutants shown to be expressed on cell surfaces and also identified an additional 10 such mutants.The present study was designed to determine whether the effects of insertions in gB on cell fusion activity would be dependent on whether a gD receptor (nectin-1) or a gB receptor (PILRα) was overexpressed in target cells that also expressed unidentified weak endogenous receptors. In addition, we assessed the abilities of the gB mutants to bind to PILRα. Our results showed that some insertions inhibited cell fusion activity when PILRα was overexpressed significantly more than when nectin-1 was overexpressed, but without necessarily preventing the binding of PILRα to gB, at least to some stable oligomeric forms of gB. The results indicate that, although both a gB receptor and a gD receptor may be required for cell fusion activity, the two receptor-binding activities have unequal weights in triggering fusogenic activity, depending on the ratios of gB and gD receptors or other factors.  相似文献   

3.
The herpes simplex virus 1 (HSV-1) glycoprotein K (gK)/UL20 protein complex is incorporated into virion envelopes and cellular membranes and functions during virus entry and cell-to-cell spread. To investigate the role of gK/UL20 in the context of a highly neurovirulent virus strain, the HSV-1(McKrae) genome was cloned into a bacterial artificial chromosome plasmid (McKbac) and utilized to construct the mutant virus McK(gKΔ31-68), carrying a 37-amino-acid deletion within the gK amino terminus. The McKbac virus entered efficiently into Chinese hamster ovary (CHO) cells constitutively expressing HSV-1 human receptors, nectin-1, herpesvirus entry mediator (HVEM), or paired immunoglobulin-like type-2 receptor alpha (PILRα). In contrast, the McK(gKΔ31-68) virus failed to enter into CHO-PILRα cells, while it entered CHO cells expressing HVEM and nectin-1 more efficiently than the McKbac virus. Both McKbac and McK(gKΔ31-68) viruses entered all CHO cells expressing HSV-1 receptors via a pH-independent pathway. The HSV-1(F) gBΔ28syn mutant virus, encoding a carboxyl-terminal truncated gB, causes extensive cell fusion. Previously, we showed that the gKΔ31-68 amino acid deletion abrogated gBΔ28syn virus-induced cell fusion, indicating that the amino terminus of gK is required for gB-mediated virus-induced cell fusion (V. N. Chouljenko, A. V. Iyer, S. Chowdhury, D. V. Chouljenko, and K. G. J. Kousoulas, Virology 83:12301–12313, 2009). Surprisingly, the gKΔ31-68/gBΔ28syn virus caused extensive fusion of CHO-nectin-1 cells but limited cell fusion of CHO-PILRα cells. Coimmunoprecipitation experiments revealed that both gK and PILRα bound gB in infected cells. Collectively, these results indicate that the amino terminus of gK is functionally and physically associated with the gB-PILRα protein complex and regulates membrane fusion of the viral envelope with cellular membranes during virus entry as well as virus-induced cell-to-cell fusion.  相似文献   

4.
5.
6.
The Us2 gene encodes a tegument protein that is conserved in most members of the Alphaherpesvirinae. Previous studies on the pseudorabies virus (PRV) Us2 ortholog indicated that it is prenylated, associates with membranes, and spatially regulates the enzymatic activity of the MAP (mitogen-activated protein) kinase ERK (extracellular signal-related kinase) through direct binding and sequestration of ERK at the cytoplasmic face of the plasma membrane. Here we present an analysis of the herpes simplex virus 2 (HSV-2) Us2 ortholog and demonstrate that, like PRV Us2, HSV-2 Us2 is a virion component and that, unlike PRV Us2, it does not interact with ERK in yeast two-hybrid assays. HSV-2 Us2 lacks prenylation signals and other canonical membrane-targeting motifs yet is tightly associated with detergent-insoluble membranes and localizes predominantly to recycling endosomes. Experiments to identify cellular proteins that facilitate HSV-2 Us2 membrane association were inconclusive; however, these studies led to the identification of HSV-2 Us2 as a ubiquitin-interacting protein, providing new insight into the functions of HSV-2 Us2.  相似文献   

7.
Herpes simplex virus type 2 (HSV-2) is a sexually transmitted virus that is highly prevalent worldwide, causing a range of symptoms that result in significant healthcare costs and human suffering. ACAM529 is a replication-defective vaccine candidate prepared by growing the previously described dl5-29 on a cell line appropriate for GMP manufacturing. This vaccine, when administered subcutaneously, was previously shown to protect mice from a lethal vaginal HSV-2 challenge and to afford better protection than adjuvanted glycoprotein D (gD) in guinea pigs. Here we show that ACAM529 given via the intramuscular route affords significantly greater immunogenicity and protection in comparison with subcutaneous administration in the mouse vaginal HSV-2 challenge model. Further, we describe a side-by-side comparison of intramuscular ACAM529 with a gD vaccine across a range of challenge virus doses. While differences in protection against death are not significant, ACAM529 protects significantly better against mucosal infection, reducing peak challenge virus shedding at the highest challenge dose by over 500-fold versus 5-fold for gD. Over 27% (11/40) of ACAM529-immunized animals were protected from viral shedding while 2.5% (1/40) were protected by the gD vaccine. Similarly, 35% (7/20) of mice vaccinated with ACAM529 were protected from infection of their dorsal root ganglia while none of the gD-vaccinated mice were protected. These results indicate that measuring infection of the vaginal mucosa and of dorsal root ganglia over a range of challenge doses is more sensitive than evaluating survival at a single challenge dose as a means of directly comparing vaccine efficacy in the mouse vaginal challenge model. The data also support further investigation of ACAM529 for prophylaxis in human subjects.  相似文献   

8.
9.
10.
Kyasanur Forest disease virus (KFDV) and Alkhumra hemorrhagic fever virus (AHFV) are genetically closely-related, tick-borne flaviviruses that cause severe, often fatal disease in humans. Flaviviruses in the tick-borne encephalitis (TBE) complex typically cause neurological disease in humans whereas patients infected with KFDV and AHFV predominately present with hemorrhagic fever. A small animal model for KFDV and AHFV to study the pathogenesis and evaluate countermeasures has been lacking mostly due to the need of a high biocontainment laboratory to work with the viruses. To evaluate the utility of an existing mouse model for tick-borne flavivirus pathogenesis, we performed serial sacrifice studies in BALB/c mice infected with either KFDV strain P9605 or AHFV strain Zaki-1. Strikingly, infection with KFDV was completely lethal in mice, while AHFV caused no clinical signs of disease and no animals succumbed to infection. KFDV and high levels of pro-inflammatory cytokines were detected in the brain at later time points, but no virus was found in visceral organs; conversely, AHFV Zaki-1 and elevated levels of cytokines were found in the visceral organs at earlier time points, but were not detected in the brain. While infection with either virus caused a generalized leukopenia, only AHFV Zaki-1 induced hematologic abnormalities in infected animals. Our data suggest that KFDV P9605 may have lost its ability to cause hemorrhagic disease as the result of multiple passages in suckling mouse brains. However, likely by virtue of fewer mouse passages, AHFV Zaki-1 has retained the ability to replicate in visceral organs, cause hematologic abnormalities, and induce pro-inflammatory cytokines without causing overt disease. Given these striking differences, the use of inbred mice and the virus passage history need to be carefully considered in the interpretation of animal studies using these viruses.  相似文献   

11.
To facilitate detection of glycoprotein K (gK) specified by herpes simplex virus, a 12-amino-acid epitope tag was inserted within gK domain III. Recombinant virus gKprotC-DIII, expressing the tagged gK, was isolated. This virus formed wild-type plaques and replicated as efficiently as the wild-type KOS virus in Vero cells. Anti-protein C MAb detected high-mannose and Golgi complex-dependent glycosylated gK within cells as well as on purified virions. The gK-null virus DeltagK (gK(-/-)) entered Vero cells substantially more slowly than the wild-type KOS (gK(+/+)), while DeltagK virus grown in complementing VK302 cells (gK(-/+)) entered with entry kinetics similar to those of the KOS virus.  相似文献   

12.
Intravaginal (IVAG) inoculation of wild-type herpes simplex virus type 2 (HSV-2) in mice causes epithelial infection followed by lethal neurological illness, while IVAG inoculation of attenuated HSV-2 causes epithelial infection followed by development of protective immunity against subsequent IVAG challenge with wild-type virus. The role of T cells in this immunity was studied by in vivo depletion of these cells with monoclonal antibodies. Three groups of mice were used for each experiment: nonimmune/challenged mice, immune/challenged mice, and immune depleted mice [immune mice depleted of a T-cell subset(s) shortly before challenge with HSV-2]. Mice were assessed for epithelial infection 24 h after challenge, virus protein in the vaginal lumen 3 days after challenge, and neurological illness 8 to 14 days after challenge. Monoclonal antibodies to CD4, CD8, or Thy-1 markedly reduced T cells in blood, spleen, and vagina, but major histocompatibility complex class II antigens were still partially upregulated in the vaginal epithelium after virus challenge, indicating that virus-specific memory T-cell function was not entirely eliminated from the vagina. Nevertheless, immune mice depleted of CD4+ and CD8+ T cells, Thy-1+ T cells, or CD8+ T cells alone had greater viral infection in the vaginal epithelium than nondepleted immune mice, indicating that T cells contribute to immunity against vaginal HSV-2 infection. All immune depleted mice retained substantial immunity to epithelial infection and were immune to neurological illness, suggesting that other immune mechanisms such as virus-specific antibody may also contribute to immunity.

Herpes simplex virus type 2 (HSV-2) is a sexually transmitted pathogen that infects the human genital tract. The prevalence of this infection is increasing worldwide, and at present over 20% of the adult U.S. population is infected with the virus (12). The virus spreads from the genital tract to the nervous system, and latent virus can persist in infected ganglia for long periods after primary infection is resolved. Activation of latent virus causes recurrent lesions in the genital tract and adjacent tissues (3). Infections are particularly severe in immunocompromised individuals and in infants who are infected during delivery through an infected birth canal. Oral treatment with acyclovir can reduce the severity of infections, but vaccination to prevent or control HSV-2 infections is highly desirable. Development of an effective vaccine to prevent genital HSV-2 infection in women is problematic at present because we do not clearly understand how to elicit strong protective immunity in the mucosa of the female genital tract. Investigations of immunity to genital HSV-2 infection in animal models are likely to play an important part in the development of a vaccine for human use. An added advantage of such investigations is that the basic information so obtained may be applicable to vaccines for other human sexually transmitted diseases.Experimental studies of host resistance to genital herpes have been carried out by using a mouse model (79). In this model, intravaginal (IVAG) inoculation of wild-type, thymidine kinase-expressing HSV-2 (TK+HSV-2) into young BALB/c mice caused epithelial infection followed by lethal neurological illness. The investigators also constructed an attenuated strain of the virus, ΔTKHSV-2, that contained a partial deletion of the thymidine kinase gene (9). Unlike its wild-type counterpart, the attenuated virus inoculated IVAG caused mild inflammation in the vagina and was incapable of lethal neurological spread. Importantly, IVAG inoculation of BALB/c mice with ΔTKHSV-2 induced a protective immunity to subsequent lethal challenge with TK+HSV-2 (9).Studies of immunity to vaginal HSV-2 infection in the young-mouse model were constrained by the relationship between vaginal infection and age (9, 21). Approximately 100% of weaned mice were susceptible to vaginal HSV-2 infection, but infection declined exponentially with increasing host age; fewer than 10% of mice were susceptible to HSV-2 at 14 to 16 weeks of age (9). However, several studies have shown that adult female mice treated with progesterone or sequentially with estradiol and Depo-Provera (E/DP-treated mice) became uniformly susceptible to vaginal HSV-2 infection (1, 13, 16). Vaginal infection of E/DP-treated mice with attenuated HSV-2 produced immunity that protected the mice against later infection by wild-type virus (16). Interestingly, 35 of 36 nonimmune mice showed immunostaining of virus proteins in the vaginal epithelium 24 h after IVAG inoculation of HSV-2, while only 1 of 9 immune mice challenged with the virus showed epithelial infection at this time (16). This indicates that virus infection or replication in the vaginal epithelium was rapidly and severely inhibited in the immune mice and suggests that local immune mechanisms in the vaginal mucosa were important in protection against challenge infection.One local immune mechanism that could prevent infection of the vaginal epithelium is neutralization of challenge virus by secreted antibody in the vaginal lumen. McDermott et al. (7) and Milligan and Bernstein (11) demonstrated immunoglobulin G (IgG) antibodies specific for HSV-2 in vaginal secretions of young immune mice; antiviral IgA either was not detected or was detected only at very low titers in vaginal fluids in these mice. More recently, Parr et al. (14) found IgG viral antibody in vaginal secretions of adult immune mice at a mean titer of 6,200, whereas the mean titer of viral secretory IgA (S-IgA) in the same secretions was only 1.9. The protective role of IgG and S-IgA in vaginal secretions of adult immune mice has also been studied (15). Unfractionated vaginal antibodies from immune and nonimmune mice and affinity-purified IgG and S-IgA from immune vaginal secretions were adjusted to their in vivo concentrations in the vagina. Neutralization of HSV-2 was studied by incubating the virus in the antibody preparations in vitro, followed by inoculation into vaginas of nonimmune test mice. Virus was neutralized by unfractionated immune antibody and by purified immune IgG but not by unfractionated nonimmune antibody or by purified immune S-IgA. To determine whether immune IgG alone could protect against vaginal HSV-2 infection in vivo, purified serum IgG from immune and nonimmune donors was passively transferred to nonimmune recipients 72 h prior to virus challenge in the vagina. Passively transferred immune IgG reduced virus infection of vaginal epithelium, shed virus protein concentrations in the vaginal lumen, and illness scores, even though the viral antibody titers in serum and vaginal secretions of recipient mice were only 29 and 8%, respectively, of those in standards prepared from actively immunized mice. Collectively, the data indicated that IgG viral antibody in vaginal secretions of immune mice provided early protection against vaginal challenge infection, probably by neutralizing virus in the vaginal lumen before it could infect the epithelium. In contrast, viral S-IgA antibody contributed relatively little to immune protection of the vagina in this model.Another immune mechanism that might reduce infection of the vaginal epithelium after viral challenge is T-cell-mediated immunity. Adoptive transfer of lymphocytes from the genital lymph nodes of immune mice protected nonimmune mice against neurological illness after vaginal challenge with wild-type HSV-2 (8). This observation indicates that virus-specific T cells, if present in sufficient numbers, can protect against neurological illness, but it remains unknown whether the T cells that are actually present in immune mice protect against either vaginal epithelial infection or neurological illness. Few T cells were present in the vaginas of normal mice (17), but the numbers of CD4+, CD8+, and Thy-1.2+ T cells increased markedly in the vaginas of immune mice after challenge with wild-type virus (16). Similarly, we have shown that T cells with the memory phenotype continuously recirculate through the vaginal epithelium and that the number of recirculating memory cells was markedly increased when immune mice were challenged in the vagina with HSV-2 (5). The presence of specific HSV-2 memory T cells in the vaginal epithelium of immune mice is also indicated by the rapid (less than 24 h) upregulation of major histocompatibility complex (MHC) class II antigen expression in the epithelium after vaginal challenge with HSV-2. In comparison, upregulation of MHC class II antigens was not detected in the vaginal epithelium until 3 days after a primary vaginal HSV-2 infection in nonimmune mice (16). In the present study, we used the adult mouse model to examine the effects of acute in vivo depletion of T-cell subsets in immune mice on vaginal epithelial infection and neurological illness after vaginal challenge with wild-type HSV-2.  相似文献   

13.
Cell number in the mouse thymus increases steadily during the first two weeks after birth. It then plateaus and begins to decline by seven weeks after birth. The factors governing these dramatic changes in cell production are not well understood. The data herein correlate levels of High mobility group A 2 protein (Hmga2) expression with these temporal changes in thymopoiesis. Hmga2 is expressed at high levels in murine fetal and neonatal early T cell progenitors (ETP), which are the most immature intrathymic precursors, and becomes almost undetectable in these progenitors after 5 weeks of age. Hmga2 expression is critical for the initial, exponential expansion of thymopoiesis, as Hmga2 deficient mice have a deficit of ETPs within days after birth, and total thymocyte number is repressed compared to wild type littermates. Finally, our data raise the possibility that similar events occur in humans, because Hmga2 expression is high in human fetal thymic progenitors and falls in these cells during early infancy.  相似文献   

14.
15.
Several vaccines have been investigated experimentally in the herpes simplex virus type 2 (HSV-2) model system. While it is believed that CD4+-T-cell responses are important for protection in general, the correlates of protection from HSV-2 infection are still under investigation. Recently, the use of molecular adjuvants to drive vaccine responses induced by DNA vaccines has been reported in a number of experimental systems. We sought to take advantage of this immunization model to gain insight into the correlates of immune protection in the HSV-2 mouse model system and to further explore DNA vaccine technology. To investigate whether the Th1- or Th2-type immune responses are more important for protection from HSV-2 infection, we codelivered the DNA expression construct encoding the HSV-2 gD protein with the gene plasmids encoding the Th1-type (interleukin-2 [IL-2], IL-12, IL-15, and IL-18) and Th2-type (IL-4 and IL-10) cytokines in an effort to drive immunity induced by vaccination. We then analyzed the modulatory effects of the vaccine on the resulting immune phenotype and on the mortality and the morbidity of the immunized animals following a lethal challenge with HSV-2. We observed that Th1 cytokine gene coadministration not only enhanced the survival rate but also reduced the frequency and severity of herpetic lesions following intravaginal HSV challenge. On the other hand, coinjection with Th2 cytokine genes increased the rate of mortality and morbidity of the challenged mice. Moreover, of the Th1-type cytokine genes tested, IL-12 was a particularly potent adjuvant for the gD DNA vaccination.  相似文献   

16.
为了构建单纯疱疹病毒2型(HSV-2)感染细胞多肽27(ICP27)真核表达质粒,应用PCR技术从HSV-2 333株的基因组中扩增ICP27基因,并连接至真核表达载体pEGFPC2,对阳性克隆进行菌落PCR、酶切和测序鉴定后,成功构建了重组质粒pEGFPC2-ICP27。用X fect转染试剂盒将重组质粒pEGFPC2-ICP27转染至Vero细胞中,并用RT-PCR及W estern b lot-ting检测其表达情况。结果显示,ICP27基因在Vero细胞中得到正确表达。真核表达质粒pEGFPC2-ICP27的构建成功,为进一步研究ICP27对宿主细胞的影响奠定了基础。  相似文献   

17.
Anthrax toxin receptor 1/tumor endothelial marker 8 (Antxr1 or TEM8) is up-regulated in tumor vasculature and serves as a receptor for anthrax toxin, but its physiologic function is unclear. The objective of this study was to evaluate the role of Antxr1 in arteriogenesis. The role of Antxr1 in arteriogenesis was tested by measuring gene expression and immunohistochemistry in a mouse model of hindlimb ischemia using wild-type and ANTXR1-/- mice. Additional tests were performed by measuring gene expression in in vitro models of fluid shear stress and hypoxia, as well as in human muscle tissues obtained from patients having peripheral artery disease. We observed that Antxr1 expression transiently increased in ischemic tissues following femoral artery ligation and that its expression was necessary for arteriogenesis. In the absence of Antxr1, the mean arterial lumen area in ischemic tissues decreased. Antxr1 mRNA and protein expression was positively regulated by fluid shear stress, but not by hypoxia. Furthermore, Antxr1 expression was elevated in human peripheral artery disease requiring lower extremity bypass surgery. These findings demonstrate an essential physiologic role for Antxr1 in arteriogenesis and peripheral artery disease, with important implications for managing ischemia and other arteriogenesis-dependent vascular diseases.  相似文献   

18.
Severe acute respiratory syndrome coronavirus (SARS-CoV) infection often caused severe end stage lung disease and organizing phase diffuse alveolar damage, especially in the elderly. The virus-host interactions that governed development of these acute end stage lung diseases and death are unknown. To address this question, we evaluated the role of innate immune signaling in protection from human (Urbani) and a recombinant mouse adapted SARS-CoV, designated rMA15. In contrast to most models of viral pathogenesis, infection of type I, type II or type III interferon knockout mice (129 background) with either Urbani or MA15 viruses resulted in clinical disease outcomes, including transient weight loss, denuding bronchiolitis and alveolar inflammation and recovery, identical to that seen in infection of wildtype mice. This suggests that type I, II and III interferon signaling play minor roles in regulating SARS pathogenesis in mouse models. In contrast, infection of STAT1−/− mice resulted in severe disease, high virus titer, extensive pulmonary lesions and 100% mortality by day 9 and 30 post-infection with rMA15 or Urbani viruses, respectively. Non-lethal in BALB/c mice, Urbani SARS-CoV infection in STAT1−/− mice caused disseminated infection involving the liver, spleen and other tissues after day 9. These findings demonstrated that SARS-CoV pathogenesis is regulated by a STAT1 dependent but type I, II and III interferon receptor independent, mechanism. In contrast to a well documented role in innate immunity, we propose that STAT1 also protects mice via its role as an antagonist of unrestrained cell proliferation.  相似文献   

19.
Human metapneumovirus (hMPV) infection causes acute respiratory tract infections (RTI) which can result in hospitalization of both children and adults. To date, no antiviral or vaccine is available for this common viral infection. Immunomodulators could represent an interesting strategy for the treatment of severe viral infection. Recently, the role of protease-activated receptors (PAR) in inflammation, coagulation and infection processes has been of growing interest. Herein, the effects of a PAR1 agonist and a PAR1 antagonist on hMPV infection were investigated in BALB/c mice. Intranasal administration of the PAR1 agonist resulted in increased weight loss and mortality of infected mice. Conversely, the PAR1 antagonist was beneficial to hMPV infection by decreasing weight loss and clinical signs and by significantly reducing pulmonary inflammation, pro-inflammatory cytokine levels (including IL-6, KC and MCP-1) and recruitment of immune cells to the lungs. In addition, a significant reduction in pulmonary viral titers was also observed in the lungs of PAR1 antagonist-treated mice. Despite no apparent direct effect on virus replication during in vitro experiments, an important role for PAR1 in the regulation of furin expression in the lungs was shown for the first time. Further experiments indicated that the hMPV fusion protein can be cleaved by furin thus suggesting that PAR1 could have an effect on viral infectivity in addition to its immunomodulatory properties. Thus, inhibition of PAR1 by selected antagonists could represent an interesting strategy for decreasing the severity of paramyxovirus infections.  相似文献   

20.
The infant immune response to respiratory syncytial virus (RSV) remains incompletely understood. Here we review the use of a neonatal mouse model of RSV infection to mimic severe infection in human infants. We describe numerous age-specific responses, organized by cell type, observed in RSV-infected neonatal mice and draw comparisons (when possible) to human infants.  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