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.
Herpesviruses can enter host cells using pH-dependent endocytosis pathways in a cell-specific manner. Envelope glycoprotein B (gB) is conserved among all herpesviruses and is a critical component of the complex that mediates membrane fusion and entry. Here we demonstrate that mildly acidic pH triggers specific conformational changes in herpes simplex virus (HSV) gB. The antigenic structure of gB was specifically altered by exposure to low pH both in vitro and during entry into host cells. The oligomeric conformation of gB was altered at a similar pH range. Exposure to acid pH appeared to convert virion gB into a lower-order oligomer. The detected conformational changes were reversible, similar to those in other class III fusion proteins. Exposure of purified, recombinant gB to mildly acidic pH resulted in similar changes in conformation and caused gB to become more hydrophobic, suggesting that low pH directly affects gB. We propose that intracellular low pH induces alterations in gB conformation that, together with additional triggers such as receptor binding, are essential for virion-cell fusion during herpesviral entry by endocytosis.Herpes simplex virus (HSV) is an important human pathogen, causing significant morbidity and mortality worldwide. HSV enters host cells by fusion of the viral envelope with either an endosomal membrane (38) or the plasma membrane (63). The entry pathway taken is thought to be determined by both virus (17, 45) and host cell (4, 17, 35, 39, 45) factors. Based on experiments with lysosomotropic agents, which elevate the normally low pH of endosomes, acidic pH has been implicated in the endocytic entry of HSV into several cell types, including human epithelial cells (37). Low pH has also recently been implicated in cell infection by several other human and veterinary herpesviruses (1, 21, 26, 47). The mechanistic role of endosomal pH in herpesvirus entry into cells is not known.Herpesviruses are a paradigm for membrane fusion mediated by a complex of several glycoproteins. We have proposed that HSV likely encodes machinery to mediate both pH-dependent and pH-independent membrane fusion reactions. Envelope glycoproteins glycoprotein B (gB) and gD and the heterodimer gH-gL are required for both pH-independent and pH-dependent entry pathways (11, 22, 30, 39, 46). Interaction of gD with one of its cognate receptors is an essential trigger for membrane fusion and entry (13, 52), regardless of the cellular pathway. However, engagement of a gD receptor is not sufficient for fusion, and at least one additional unknown trigger involving gB or gH-gL is likely necessary. gB is conserved among all herpesviruses, and in all cases studied to date, it plays roles in viral entry, including receptor binding and membrane fusion. The crystal structure of an ectodomain fragment of HSV type 1 (HSV-1) gB is an elongated, rod-like structure containing hydrophobic internal fusion loops (28). This structure bears striking architectural homology to the low pH, postfusion form of G glycoprotein from vesicular stomatitis virus (VSV-G) (43). Both the gB and G structures have features of class I and class II fusion proteins and are thus designated class III proteins (57).During entry of the majority of virus families, low pH acts directly on glycoproteins to induce membrane fusion (60). In some cases, the low pH trigger is not sufficient, or it plays an indirect role. For example, host cell proteases, such as cathepsins D and L, require intravesicular low pH to cleave Ebola virus and severe acute respiratory syndrome (SARS) glycoproteins to trigger fusion (14, 51).We investigated the role of low pH in the molecular mechanism of herpesviral entry. The results suggest that mildly acidic pH, similar to that found within endosomes, triggers a conformational change in gB. We propose that, together with other cellular cues such as receptor interaction, intracellular low pH can play a direct activating role in HSV membrane fusion and entry.  相似文献   

2.
Using a liposome-binding assay, we investigated the requirements for activation of herpes simplex virus (HSV) into a state capable of membrane interaction. Virions were mixed with liposomes along with the ectodomain of one of three gD receptors (HVEMt, nectin-1t, or nectin-2t) and incubated under different pH and temperature conditions. Virions failed to associate with liposomes in the presence of nectin-1 or nectin-2 at any temperature or pH tested. In contrast, HVEMt triggered association of HSV with liposomes at pH 5.3 or 5.0 when incubated at 37 degrees C, suggesting that HVEM binding and mildly acidic pH at a physiological temperature provide coactivation signals, allowing virus association with membranes. Virions incubated with HVEMt at 37 degrees C without liposomes rapidly lost infectivity upon exposure to pH 5.0, suggesting that these conditions lead to irreversible virus inactivation in the absence of target membranes. Consistent with the idea that soluble receptor molecules provide a trigger for HSV entry, HVEMt promoted virus entry into receptor-deficient CHO K1 cells. However, in B78H1 cells, HVEMt promoted virus entry with markedly lower efficiency. Interestingly, HSV entry into receptor-bearing CHO K1 cells has been shown to proceed via a pH-dependent manner, whereas HSV entry into receptor-bearing B78H1 cells is pH independent. Based on these observations, we propose that the changes triggered by HVEM and mildly acidic pH that allow liposome association are similar or identical to changes that occur during pH-dependent HSV entry.  相似文献   

3.

Background

Herpes simplex virus (HSV) can utilize multiple pathways to enter host cells. The factors that determine which route is taken are not clear. Chinese hamster ovary (CHO) cells that express glycoprotein D (gD)-binding receptors are model cells that support a pH-dependent, endocytic entry pathway for all HSV strains tested to date. Fusion-from-without (FFWO) is the induction of target cell fusion by addition of intact virions to cell monolayers in the absence of viral protein expression. The receptor requirements for HSV-induced FFWO are not known. We used the syncytial HSV-1 strain ANG path as a tool to evaluate the complex interplay between receptor usage, membrane fusion, and selection of entry pathway.

Results

Inhibitors of endocytosis and endosome acidification blocked ANG path entry into CHO cells expressing nectin-1 receptors, but not CHO-nectin-2 cells. Thus, under these conditions, nectin-2 mediates pH-independent entry at the plasma membrane. In addition, CHO-nectin-2 cells supported pH-dependent, endocytic entry of different strains of HSV-1, including rid1 and HFEM. The kinetics of ANG path entry was rapid (t1/2 of 5–10 min) regardless of entry route. However, HSV-1 ANG path entry by fusion with the CHO-nectin-2 cell plasma membrane was more efficient and resulted in larger syncytia. ANG path virions added to the surface of CHO-nectin-2 cells, but not receptor-negative CHO cells or CHO-nectin-1 cells, induced rapid FFWO.

Conclusion

HSV-1 ANG path can enter CHO cells by either endocytic or non-endocytic pathways depending on whether nectin-1 or nectin-2 is present. In addition to these cellular receptors, one or more viral determinants is important for the selection of entry pathway. HSV-induced FFWO depends on the presence of an appropriate gD-receptor in the target membrane. Nectin-1 and nectin-2 target ANG path to divergent cellular pathways, and these receptors may have different roles in triggering viral membrane fusion.  相似文献   

4.
5.
During infection by herpes simplex virus type‐1 (HSV‐1) the host cell undergoes widespread changes in gene expression and morphology in response to viral replication and release. However, relatively little is known about the specific proteome changes that occur during the early stages of HSV‐1 replication prior to the global damaging effects of virion maturation and egress. To investigate pathways that may be activated or utilised during the early stages of HSV‐1 replication, 2‐DE and LC‐MS/MS were used to identify cellular proteome changes at 6 h post infection. Comparative analysis of multiple gels representing whole cell extracts from mock‐ and HSV‐1‐infected HEp‐2 cells revealed a total of 103 protein spot changes. Of these, 63 were up‐regulated and 40 down‐regulated in response to infection. Changes in selected candidate proteins were verified by Western blot analysis and their respective cellular localisations analysed by confocal microscopy. We have identified differential regulation and modification of proteins with key roles in diverse cellular pathways, including DNA replication, chromatin remodelling, mRNA stability and the ER stress response. This work represents the first global comparative analysis of HSV‐1 infected cells and provides an important insight into host cell proteome changes during the early stages of HSV‐1 infection.  相似文献   

6.
The virological synapse (VS) is a specialized molecular structure that facilitates the transfer of certain lymphotropic viruses into uninfected T cells. However, the role of the VS in the transfer of nonlymphotropic viruses into T cells is unknown. Herpes simplex virus (HSV) has been shown in vitro to infect T cells and modulate T-cell receptor function, thereby suppressing T-cell antiviral function. However, whether such infection of T cells occurs in vivo is unknown. Here, we examined whether T-cell infection could be observed in human HSV disease and investigated the mechanism of HSV entry into T cells. We found that HSV-infected T cells were readily detectable during human disease, suggesting that infection and modulation of T-cell function plays a role in human immunopathology. HSV infection of both CD4+ and CD8+ T cells occurred much more efficiently via direct cell-to-cell spread from infected fibroblasts than by cell-free virus. Activation of T cells increased their permissivity to HSV infection. Cell-to-cell spread to T cells did not require HSV glycoproteins E and I (gE and gI), which are critical for cell-to-cell spread between epithelial cells. Transfer of HSV to T cells required gD, and the four known entry receptors appear to be contributing to viral entry, with a dominant role for the herpesvirus entry mediator and nectin-1. VS-like structures enriched in activated lymphocyte function-associated antigen 1 (LFA-1) were observed at the point of contact between HSV-infected fibroblasts and T cells. Consistent with spread occurring via the VS, transfer of HSV was increased by activation of LFA-1, and cell-to-cell spread could be inhibited by antibodies to LFA-1 or gD. Taken together, these results constitute the first demonstration of VS-dependent cell-to-cell spread for a predominantly nonlymphotropic virus. Furthermore, they support an important role for infection and immunomodulation of T cells in clinical human disease. Targeting of the VS might allow selective immunopotentiation during infections with HSV or other nonlymphotropic viruses.The virological synapse (VS) is a specialized molecular structure that facilitates the transfer of certain lymphotropic viruses, such as human immunodeficiency virus (HIV) and human T-cell leukemia virus type 1 (HTLV-1), into uninfected T cells (22, 28, 38). Entry and infection of T cells by HIV or HTLV-1 via the VS is far more efficient than infection by cell-free virus, and thus this structure plays a critical role in the pathogenesis of these viruses. The organization of the VS is in many respects similar to the immunological synapse (IS), in particular, to the immature IS. The VS is highly enriched in the adhesion molecule lymphocyte function-associated antigen 1 (LFA-1) and its ligands intercellular adhesion molecule 1 (ICAM-1) and ICAM-3 (29); however, it does not possess the CD3-enriched central region associated with the mature IS (28, 47). While the VS is critical to the pathogenesis of HIV and HTLV-1, it remains an unanswered question whether the VS is also involved in T-cell infection by other viruses, especially those not typically considered lymphotropic.Herpes simplex virus (HSV) is a remarkably successful human pathogen that establishes lifelong latency in neurons of the dorsal root ganglia. HSV can efficiently reactivate from the latent state and transmit to new hosts despite the presence of preformed immunity. HSV is thought to achieve this feat by employing a number of sophisticated immune evasion mechanisms (33), many of which are directed at the cellular arm of the immune response. In one such potential mechanism, HSV has evolved the ability to enter and infect T cells. Although T cells do not support efficient viral replication (25), infection by HSV profoundly modulates T-cell receptor (TCR) signaling, which prevents T-cell cytotoxic function (55) and alters cytokine production profiles toward an interleukin-10-dominated immunosuppressive phenotype (54). However, it is unknown whether and to what extent HSV infection of T cells occurs during human HSV disease. Furthermore, the dominant mechanisms by which HSV might gain access to lesion-infiltrating T cells have not been elucidated.Here, we evaluated T-cell infection during human HSV infections, the mechanisms by which HSV enters T cells, the relative involvement of cell-cell spread versus cell-free virus in T-cell infection, and the role of the VS in the infection of T cells by HSV. The demonstration of infection of T cells in human HSV disease and of a dominant role for the VS in entry of HSV into T cells suggests that the VS is important in the pathogenesis of nonlymphotropic as well as lymphotropic viruses. Thus, the VS may be a unique pharmacologic target to allow improved immune control of a wide variety of viral infections.  相似文献   

7.

Background

Herpes viruses establish a life-long latency and can cause symptoms during both first-time infection and later reactivation. The aim of the present study was to describe the seroepidemiology of Herpes simplex type 1 (HSV1), Herpes simplex type 2 (HSV2), Cytomegalovirus (CMV), Varicella Zoster virus (VZV) and Human herpes virus type 6 (HHV6) in an adult Swedish population (35–95 years of age).

Methods

Presence of antibodies against the respective viruses in serum from individuals in the Betula study was determined with an enzyme-linked immunosorbent assay (ELISA). Singular samples from 535 persons (53.9% women, mean age at inclusion 62.7?±?14.4 years) collected 2003-2005 were analyzed for the five HHVs mentioned above. In addition, samples including follow-up samples collected 1988–2010 from 3,444 persons were analyzed for HSV.

Results

Prevalence of HSV1 was 79.4%, HSV2 12.9%, CMV 83.2%, VZV 97.9%, and HHV6 97.5%. Herpes virus infections were more common among women (p?=?0.010) and a lower age-adjusted HSV seroprevalence was found in later birth cohorts (p?<?0.001). The yearly incidence of HSV infection was estimated at 14.0/1000.

Conclusion

Women are more often seropositive for HHV, especially HSV2. Age-adjusted seroprevalence for HSV was lower in later birth cohorts indicating a decreasing childhood and adolescent risk of infection.
  相似文献   

8.
Herpes simplex virus (HSV) infection of many cultured cells, e.g., Vero cells, can be initiated by receptor binding and pH-neutral fusion with the cell surface. Here we report that a major pathway for HSV entry into the HeLa and CHO-K1 cell lines is dependent on endocytosis and exposure to a low pH. Enveloped virions were readily detected in HeLa or receptor-expressing CHO cell vesicles by electron microscopy at <30 min postinfection. As expected, images of virus fusion with the Vero cell surface were prevalent. Treatment with energy depletion or hypertonic medium, which inhibits endocytosis, prevented uptake of HSV from the HeLa and CHO cell surface relative to uptake from the Vero cell surface. Incubation of HeLa and CHO cells with the weak base ammonium chloride or the ionophore monensin, which elevate the low pH of organelles, blocked HSV entry in a dose-dependent manner. Noncytotoxic concentrations of these agents acted at an early step during infection by HSV type 1 and 2 strains. Entry mediated by the HSV receptor HveA, nectin-1, or nectin-2 was also blocked. As analyzed by fluorescence microscopy, lysosomotropic agents such as the vacuolar H(+)-ATPase inhibitor bafilomycin A1 blocked the delivery of virus capsids to the nuclei of the HeLa and CHO cell lines but had no effect on capsid transport in Vero cells. The results suggest that HSV can utilize two distinct entry pathways, depending on the type of cell encountered.  相似文献   

9.
The phenomenon of antibody-dependent cell-mediated cytoxicity (ADCC) has been extended to include target cells acutely infected with herpes simplex type 1 virus (HSV-1) or herpes simplex type 2 virus (HSV-2) in an in vitro system that employs immune human serum and human blood mononuclear cells. The cytotoxic reaction was detectable after 1 hr of incubation and was complete between 4 and 8 hr. The amount of ADCC noted was directly proportional to the logarithm(10) of the effector: target cell ratio (E:T), and ADCC was noted at E:T as low as 1:1. The mononuclear effector cell was present in the blood of both HSV immune and non-immune individuals. The immune serum factor was demonstrated to be an antibody with specificity for HSV membrane antigen(s) and was reactive with target cells infected with either of the two HSV types. The antibody rendered the mononuclear cell cytotoxic by sensitization of the target cell rather than by direct attachment to or "arming" of the mononuclear cell. The physiochemical properties of the antibody as well as its presence in cord blood demonstrated that it is an immunoglobulin on the IgG class.  相似文献   

10.
Herpes simplex virus 1 (HSV1) is an enveloped virus that uses undefined transport carriers for trafficking of its glycoproteins to envelopment sites. Screening of an siRNA library against 60 Rab GTPases revealed Rab6 as the principal Rab involved in HSV1 infection, with its depletion preventing Golgi‐to‐plasma membrane transport of HSV1 glycoproteins in a pathway used by several integral membrane proteins but not the luminal secreted protein Gaussia luciferase. Knockdown of Rab6 reduced virus yield to 1% and inhibited capsid envelopment, revealing glycoprotein exocytosis as a prerequisite for morphogenesis. Rab6‐dependent virus production did not require the effectors myosin‐II, bicaudal‐D, dynactin‐1 or rabkinesin‐6, but was facilitated by ERC1, a factor involved in linking microtubules to the cell cortex. Tubulation and exocytosis of Rab6‐positive, glycoprotein‐containing membranes from the Golgi was substantially augmented by infection, resulting in enhanced and targeted delivery to cell tips. This reveals HSV1 morphogenesis as one of the first biological processes shown to be dependent on the exocytic activity of Rab6.   相似文献   

11.
Enveloped animal viruses enter host cells either by direct fusion at neutral pH or by endocytosis. Herpes simplex virus (HSV) is believed to fuse with the plasma membrane of cells at neutral pH, and the glycoproteins gB and gD have been implicated in virus entry and cell fusion. Using cloned gB or gD genes, we show that cells expressing HSV-1 glycoproteins gB or gD can undergo fusion to form polykaryons by exposure only to acidic pH. The low pH-induced cell fusion was blocked in the presence of monoclonal antibodies specific to the glycoproteins. Infection of cells expressing gB or gD glycoproteins with HSV-1 inhibited the low pH-induced cell fusion. The results suggest that although the glycoproteins gB and gD possess fusogenic activity at acidic pH, other HSV proteins may regulate it such that in the virus-infected cell, this fusion activity is blocked.  相似文献   

12.
Development of strategies to prevent herpes simplex virus (HSV) infection requires knowledge of cellular pathways harnessed by the virus for invasion. This study demonstrates that HSV induces rapid phosphorylation of focal adhesion kinase (FAK) in several human target cells and that phosphorylation is important for entry post-binding. Nuclear transport of the viral tegument protein VP16, transport of viral capsids to the nuclear pore, and downstream events (including expression of immediate-early genes and viral plaque formation) were substantially reduced in cells transfected with dominant-negative mutants of FAK or small interfering RNA designed to inhibit FAK expression. These observations were substantiated using mouse embryonic fibroblast cells derived from embryonic FAK-deficient mice. Infection was reduced by >90% in knockout cells relative to control cells and was further reduced if the knockout cells were transfected with small interfering RNA targeting proline-rich tyrosine kinase-2, which was also phosphorylated in response to HSV. The knockout cells were permissive for viral binding, and virus triggered an intracellular calcium response, but nuclear transport was inhibited. Together, these results support a novel model for invasion that implicates FAK phosphorylation as important for delivery of viral capsids to the nuclear pore.  相似文献   

13.
Herpes simplex virus (HSV) enters cells by fusion at plasma membranes or endosomes. Cellular factors route the virus to different pathways. αVβ3-integrin directs HSV to a lipid raft and acidic endosome pathway. We report that infection mediated by nectin1 plus αVβ3-integrin exhibits the same characteristics as entry mediated by raft-located forms of nectin. αVβ3-integrin relocalizes nectin1 to lipid rafts, independently of virus. Thus, HSV routing to the lipid raft-dependent pathway is consequent to the integrin-induced relocalization of nectin1. Inhibition by the Na+/H+ exchanger 5-(N-ethyl-N-isopropyl)amirolide suggests that αVβ3-integrin overexpression favors HSV macropinocytic uptake in some cells but not in others.  相似文献   

14.
The use of herpes simplex virus (HSV) vectors for in vivo gene therapy will require the targeting of vector infection to specific cell types in certain in vivo applications. Because HSV glycoprotein D (gD) imparts a broad host range for viral infection through recognition of ubiquitous host cell receptors, vector targeting will require the manipulation of gD to provide new cell recognition specificities in a manner designed to preserve gD's essential role in virus entry. In this study, we have determined whether an entry-incompetent HSV mutant with deletions of all Us glycoproteins, including gD, can be complemented by a foreign attachment/entry protein with a different receptor-binding specificity, the vesicular stomatitis virus glycoprotein G (VSV-G). The results showed that transiently expressed VSV-G was incorporated into gD-deficient HSV envelopes and that the resulting pseudotyped virus formed plaques on gD-expressing VD60 cells, albeit at a 50-fold-reduced level compared to that of wild-type gD. This reduction may be related to differences in the entry pathways used by VSV and HSV or to the observed lower rate of incorporation of VSV-G into virus envelopes than that of gD. The rate of VSV-G incorporation was greatly improved by using recombinant molecules in which the transmembrane domain of HSV glycoprotein B or D was substituted for that of VSV-G, but these recombinant molecules failed to promote virus entry. These results show that foreign glycoproteins can be incorporated into the HSV envelope during replication and that gD can be dispensed with on the condition that a suitable attachment/entry function is provided.  相似文献   

15.
Herpes simplex virus type 1 (HSV1) is widely used as a gene delivery vector in a variety of laboratory animals. In a recent study, a thymidine-kinase–inactive (replication-conditional) HSV1 used as a delivery vector was lethal in naked mole rats, whereas mice infected with the identical virus showed no adverse effects. This result prompted us to undertake a controlled comparative histologic study of the effect of HSV1 infection on naked mole rats and mice. Replication-competent and replication-conditional HSV1 caused widespread inflammation and necrosis in multiple organ systems of naked mole rats but not mice; naked mole rats infected with replication-defective virus showed no adverse effects. We conclude that the lethality of HSV1 for naked mole rats is likely the result of overwhelming infection, possibly in part due to this species’ natural lack of proinflammatory neuropeptides at the initial site of infection.Abbreviations: HSV1, herpes simplex virus type 1Herpes simplex virus type 1 (HSV1) belongs to the Simplexvirus genus of the Alphaherpesvirineae subfamily and is an important human pathogen.21 Similar to other herpesviruses, HSV1 is well adapted to its natural host. Fatal HSV1 infections of immunocompetent humans are relatively rare. In most cases, human HSV1 infections lead to lifelong latent infection that is interrupted by episodes of viral reactivation.32 Experimental infection of mice, rabbits, rats, and guinea pigs has been used widely to study HSV1 pathogenesis.33 The pathogenesis of HSV1 in these animals shows close resemblance to infections seen in humans. Infection of peripheral tissues leads to local viral replication and brief viremia. The virus also spreads by neural pathways to the peripheral and central nervous systems, where virus again may replicate, this time in neurons and nonneuronal cells, and may cause encephalitis. Animals surviving the acute phase of infection do not demonstrate signs of encephalitis, and infectious virus is no longer detectable in their nervous system or other organs. However, HSV1 usually is not cleared from these animals and typically establishes latency in neurons of sensory ganglia.Various HSV1 isolates possess a number of characteristics that make them promising as vectors for gene delivery.7 These properties include their capacity to package large amounts of heterologous DNA and an ability to establish persistent, lifelong infections, during which the viral genome remains as a circular nonintegrated episome. In addition, HSV1-based vectors can infect a wide range of human cell lines and primary cultures with high efficiencies. This attribute allows HSV1-based vectors to stably transduce neurons and provide sustained heterologous gene expression. As such, HSV1-based vectors offer the characteristics of an artificial chromosome combined with a highly efficient delivery system. HSV1 strains used for gene therapy typically are engineered to have decreased virulence; for example, strains with defective viral thymidine kinase cannot replicate in nervous tissue, will not cause encephalitis, and are avirulent to immunocompetent hosts.8Naked mole rats have been used to study pain because they do not produce substance P and calcitonin gene-related peptide from the C fibers in their skin17 and they lack C-fiber–related responses to capsaicin.18 In other mammals, these peptides play important roles in pain signaling in the spinal cord and in initiating local immune responses in the periphery.15,19 We infected naked mole rats with a thymidine-kinase–inactivated (replication-conditional) HSV1 engineered to express the preprotachykinin gene that encodes the pain-related neuropeptides substance P and neurokinin A.4 Viruses used in the comparative study did not carry transgenes.  相似文献   

16.
The herpes simplex virus type 1 (HSV‐1) VRTK? strain that was previously isolated in our laboratory as an acyclovir‐resistant thymidine kinase (TK)‐deficient mutant, is more sensitive to type 1 interferon than is the parent strain VR3. The properties of this mutant were investigated to clarify the mechanism for its hyper‐sensitivity to interferon (IFN). It was found that: (i) IFN‐pretreated cells, but not those treated with IFN after adsorption, are hyper‐sensitive to IFN; (ii) the mutant cannot inhibit protein kinase R phosphorylation efficiently during the early stage of replication (2 hrs post‐infection); (iii) expression of US11 in infected cells and its incorporation into the virion is reduced in the mutant compared to the wild type, despite the fact that a similar degree of DNA synthesis occurs during replication of both strains and; (iv) over‐expression of wild‐type viral TK has no effect on the phenotype of the VRTK? strain, indicating that the phenotype is induced by a mutation(s) that does not involve the TK gene. These results suggested that the presence of US11 in the virion, but not that expressed after infection, plays an important role in the escape function of HSV‐1 from the antiviral activity of type 1 IFN.
  相似文献   

17.
Recent studies have renewed the debate on infectious etiology in late-onset Alzheimer''s disease. Bocharova et al. reported that abundant expression of human beta amyloid (Aβ) in the mouse brain (5XFAD animals) failed to protect against acute herpes simplex virus type 1 infection relative to control mice. While this study does not confirm the antiviral actions of Aβ, it neither supports nor disproves the hypothesis that infection with microbial pathogens is the major cause of Alzheimer''s disease.

Alzheimer''s disease (AD) is a devastating and progressive neurodegenerative disorder and the most common form of dementia in the elderly. AD is associated with the pathological deposition of neurofibrillary tau tangles and extracellular amyloid β (Aβ) plaques as well as with chronic neuroinflammation. Numerous studies showed that specific microbial infections, including with herpes simplex virus type 1 (HSV1), Chlamydia pneumoniae, and several types of spirochaete are linked to AD etiology, as these pathogens have been detected in some AD brains, particularly in senile plaques (1). HSV1 is the most common of these and strongly links human pathogens to AD etiology (supported by over 100 publications reporting direct or indirect association), whereas only a handful of reports are contradictory. The following lines of evidence support this connection: (i) the presence of viruses and other microbes in the brain of most elderly individuals; (ii) in AD brains, pathogen signatures like HSV1 DNA specifically colocalize with AD amyloid plaques; (iii) active HSV1 infection can trigger chronic neuroinflammatory responses that lead to herpes simplex encephalitis, known to cause severe damage in brain regions associated with memory and cognitive functions; (iv) circulating levels of anti-HSV antibodies, an indicator of HSV1 reactivation, is positively correlated with AD pathology; (v) HSV1 infection activates neurotoxic pathways and causes an AD-like phenotype in mice, while in clinical studies, treatment with antiherpetic drugs like valacyclovir show improved cognitive functions in patients with AD compared with controls; and (vi) key features of AD pathology are transmissible upon intracerebral injection of AD brain homogenates (1, 2), indicating that microbial infection could represent an important contributor to late-onset AD and supporting “the Aβ antimicrobial protection hypothesis.” This hypothesis proposes that Aβ deposition is an early response against microbial infection, which consequently drives chronic neuroinflammation and neurodegeneration, which is not necessarily contrary to with the established “amyloid hypothesis” that proposes that amyloidosis of Aβ is the root cause of AD (1). Recently, contrasting evidence was provided to continue the debate on the antiviral role of Aβ and the causative role of HSV1 infection in AD and to highlight that chronic neuroinflammation could represent a central mechanism in infectious etiology of late-onset AD (Fig. 1).Open in a separate windowFigure 1Schematic presentation of AD pathogenesis according to the “Aβ antimicrobial protection hypothesis” and the “amyloid β hypothesis.” Chronic microbial infections activate the immune system and lead to sustained inflammatory responses, which allow penetration of microbial pathogens and/or their products to cross the blood–brain barrier. In the brain, they colocalize with Aβ and induce Aβ fibrillation to form senile plaques. Disruption of the blood–brain barrier causes penetration of peripheral inflammatory molecules as well as inflammatory cells into the brain and promotes gliosis. Furthermore, various risk factors, including genetics, stress, sleep, diet, head injury, and aging, influence disease progression. Together, these form a vicious inflammatory response, perpetuated by chronic infections or reactivation, which further stimulate chronic neuroinflammation. This process eventually leads to neuron death and AD pathology. AD, Alzheimer''s disease.Previously, an elegant and high profile study by Eimer et al. (3) reported that human Aβ interacts with viral surface glycoproteins, which mediates Aβ oligomerization and viral entrapment, leading to protection against HSV1 infection in AD mice and 3D human neuronal cell cultures. Others also showed that Aβ amyloidosis protects against bacterial and fungal infections in animal and worm models of AD (4), suggesting that in addition to various genetic, biochemical, and environmental factors, microbial infection could represent one of the possible triggers of amyloidosis in AD neuropathology (Fig. 1).In a recent article in the Journal of Biochemistry, Bocharova et al. (5) questioned the antiviral properties of Aβ and the role of viral infection in AD pathology in contrast to the findings of Eimer et al. (3), demonstrating protective antiherpetic actions mediated by human Aβ-expressing mice (5XFAD mice). Bocharova et al. used three different age groups of 5XFAD mice expressing transgenes for mutant human amyloid precursor protein and human presenilin 1, one of the four core proteins of the gamma secretase complex that generates Aβ from amyloid precursor protein, with five AD-linked mutations. WT control and 5XFAD mice were infected with three different doses of two strains of HSV1 ranging from 5- to 10-fold below or above the dose lethal to 50% of the mice (LD50). Despite the use of different viral strains, doses, and ages of the animals, survival analysis revealed no statistically significant differences between 5XFAD and WT mice, which confirmed the lack of protective effect of the 5XFAD genotype against HSV1-induced encephalitis (5). Bocharova et al. also noted the region-specific or cell-specific tropisms of HSV1 strains that were not affected in 5XFAD mice compared with controls, which suggested that host–pathogen interactions remained unaltered by Aβ overexpression. Also contrary to the study by Eimer et al. (3), Bocharova et al. found no evidence of HSV1-induced Aβ aggregation and Aβ-mediated viral entrapment. This observation was attributed to the microglia transitioning to a chronic reactive stage and subsequently phagocytosing the virus in Aβ aggregate-dense brain regions. Reactive microglia, often seen in human AD brains, might cause chronic neuroinflammation in response to recurrent reactivation or repetitive HSV1 infections (6). This repeated and sustained microglia activation could eventually potentiate deregulated chronic neuroinflammation and development of AD pathology (7).These studies raise obvious fundamental questions, such as what are the targets for antimicrobial actions of Aβ in human? It is noteworthy to mention that mouse models are not natural hosts for HSV1 and hence do not adequately mimic spontaneous viral shedding or recurrent symptomatic diseases in humans (8). Furthermore, antimicrobial proteins show varying activity against different microbial pathogens, and therefore, the lack of protection against HSV1 infection in 5XFAD mice contrasts the strong protective effects against bacterial and fungal infections (5). This could correlate with diverse microbial infections in human brain. The discrepancies between the two studies could also be due to the use of different doses and variations of HSV1 strains. Nevertheless, the current work neither supports nor refutes the hypothesis of the viral etiology of late-onset AD. Indeed, since no protection was found against acute HSV1 infection in 5XFAD mice, it indicates that viral pathogens could even increase the risk of late-onset AD through multiple Aβ-dependent and independent mechanisms.In the future, it would be of interest to examine the antimicrobial actions of Aβ against diverse microbial pathogens, including several types of spirochete, Treponema pallidum, C. pneumoniae, and the protozoan Toxoplasma gondii, implicated in human brain diseases. Certainly, HSV1 is not the sole contributor to late-onset AD, as it is a multifactorial disease with many contributing factors (including other potential pathogens). HSV1 may contribute to a minority of AD cases, and even this etiology may depend on the presence or the absence of other risk factors such as genetic factors (apolipoprotein E4 variant carriers), age, stress, sleep, diet, head injury, cardiovascular disease, and many others (7). Collectively, these contradictory studies question the contributory role of HSV1 to AD development and the antiviral activity of Aβ. Fundamentally, the findings of Bocharova et al. (5) support the notion that chronic infections with viral, bacterial, and fungal pathogens might cause deregulated neuroinflammatory responses, which subsequently increase amyloidosis in the brain and contribute to AD pathogenesis (2, 9, 10). Thus, inflammatory responses against infections might provide the missing link between infectious etiology and late-onset AD, if such a link exists.It is worth mentioning that the infectious etiology in late-onset AD is a puzzle not yet solved, as—at least so far—no specific microbial infection has been conclusively linked to causation of AD in humans. However, interesting data from many laboratories renewed the concern of infectious etiology in late-onset AD, which will provide new opportunities for anti-inflammatory therapy development.  相似文献   

18.
Two herpes simplex virus type 1 (HSV-1) entry pathways have been described: direct fusion between the virion envelope and the plasma membrane, as seen on Vero cells, and low-pH-dependent endocytosis, as seen on CHO nectin-1 and HeLa cells. In this paper, we studied HSV entry into C10 murine melanoma cells and identified a third entry pathway for this virus. During entry into C10 cells, virion envelope glycoproteins rapidly became protected from the membrane-impermeable chemical cross-linker BS3 and from proteinase K. Protection was gD receptor dependent, and the time taken to detect protected protein was proportional to the rate of virus entry. Ultrastructural examination revealed that virions attached to the surface of C10 cells were localized to membrane invaginations, whereas those on the surface of receptor-negative B78 cells were peripherally attached. Virus entry into C10 cells was energy dependent, and intracellular enveloped virions were seen within membrane-bound vesicles consistent with endocytic entry. Entry was not inhibited by bafilomycin A1 or ammonium chloride, showing that passage of the virion through a low-pH environment was not required for infection. Resistance to similar reagents should therefore not be taken as proof of HSV entry by a nonendosomal pathway. These data define a novel gD receptor-dependent acid-independent endocytic entry pathway for HSV.  相似文献   

19.
Herpes simplex virus triggers activation of calcium-signaling pathways   总被引:10,自引:0,他引:10       下载免费PDF全文
The cellular pathways required for herpes simplex virus (HSV) invasion have not been defined. To test the hypothesis that HSV entry triggers activation of Ca2+-signaling pathways, the effects on intracellular calcium concentration ([Ca2+]i) after exposure of cells to HSV were examined. Exposure to virus results in a rapid and transient increase in [Ca2+]i. Pretreatment of cells with pharmacological agents that block release of inositol 1,4,5-triphosphate (IP3)-sensitive endoplasmic reticulum stores abrogates the response. Moreover, treatment of cells with these pharmacological agents inhibits HSV infection and prevents focal adhesion kinase (FAK) phosphorylation, which occurs within 5 min after viral infection. Viruses deleted in glycoprotein L or glycoprotein D, which bind but do not penetrate, fail to induce a [Ca2+]i response or trigger FAK phosphorylation. Together, these results support a model for HSV infection that requires activation of IP3-responsive Ca2+-signaling pathways and that is associated with FAK phosphorylation. Defining the pathway of viral invasion may lead to new targets for anti-viral therapy.  相似文献   

20.
During viral entry, herpes simplex virus (HSV) glycoprotein D (gD) interacts with a specific cellular receptor such as nectin-1 (PRR1/HveC/CD111) or the herpesvirus entry mediator A (HVEM/HveA). Nectin-1 is involved in cell-to-cell adhesion. It is located at adherens junctions, where it bridges cells through homophilic or heterophilic interactions with other nectins. Binding of HSV gD prevents nectin-1-mediated cell aggregation. Since HSV gD affects the natural function of nectin-1, we further investigated the effects of gD expression on nectin-1 during HSV infection or in transfected cells. We also studied the importance of the interaction between nectin-1 and the cytoplasmic protein afadin for HSV entry and spread as well as the effects of infection on this interaction. In these investigations, we used a panel of cells expressing nectin-1 or nectin-1-green fluorescent protein fusions as the only mediators of HSV entry. During HSV infection, nectin-1 localization at adherens junction was dramatically altered in a manner dependent on gD expression. Nectin-1 and gD colocalized at cell contact areas between infected and noninfected cells and at the edges of plaques. This specific accumulation of gD at junctions was driven by expression of nectin-1 in trans on the surface of adjacent cells. Reciprocally, nectin-1 was maintained at junctions by the trans expression of gD in the absence of a cellular natural ligand. Our observations indicate that newly synthesized gD substitutes for nectin-1 of infected cells at junctions with noninfected cells. We propose that gD attracts and maintains the receptor at junctions where it can be used for virus spread.  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