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.
Neospora caninum and Toxoplasma gondii are closely related, obligate intracellular parasites infecting a wide range of vertebrate hosts and causing abortion and neonatal morbidity and mortality. Several lines of evidence suggest that cross immunity between these two pathogens could be exploited in the design of strategies for heterologous vaccination. We assessed the ability of an attenuated strain of T. gondii (“mic1-3KO strain”) conferring strong protection against chronic and congenital toxoplasmosis to protect mice against lethal N. caninum infection. Mice immunized with mic1-3KO tachyzoites by the oral and intraperitoneal routes developed a strong cellular Th1 response and displayed significant protection against lethal heterologous N. caninum infection, with survival rates of 70% and 80%, respectively, whereas only 30% of the nonimmunized mice survived. We report here the acquisition of heterologous protective immunity against N. caninum following immunization with a live attenuated mic1-3KO strain of T. gondii.Neospora caninum and Toxoplasma gondii are closely related apicomplexans displaying extensive morphological and genetical similarity (37). These cyst-forming coccidians of the family Sarcocystidae (11) cause similar disorders in different animals (15). Despite controversial documentation on their phylogenetic relationship (30), molecular (31) and biological studies have shown that these species have followed different evolutionary paths and have different life cycles and host preferences. Canids are the definitive hosts of N. caninum, which causes neosporosis, a major disease of cattle, whereas felids are the definitive hosts of T. gondii, which causes toxoplasmosis, a major disease of sheep, goats, and humans (10). Both parasites are responsible for important economic losses in livestock production through neonatal mortality and abortion. T. gondii also causes congenital neuropathology and opportunistic infections in immunocompromised humans (41), but there is no conclusive evidence to suggest that N. caninum can infect humans (29).Previous clinical and diagnostic studies have shown that specific antibodies directed against N. caninum or T. gondii cross-react in serological and immunohistochemical tests, suggesting a possible convergence of immune responses during infections with T. gondii and N. caninum (32, 38). It has recently been shown that antibodies directed against N. caninum antigens inhibit host cell invasion by both these parasites (22, 43). Similarly, the specific cellular responses stimulated upon experimental infections with N. caninum are also stimulated in vitro by T. gondii antigenic lysate (21, 26). Consistent with these findings, CD8+ T cells specific for N. caninum have been shown to protect mice against lethal T. gondii infection (19). The existence of cross-reactive epitopes between N. caninum and T. gondii antigens is supported by the high level of sequence identity between conserved proteins (13). A number of cross-reactive antigens have been identified in the micronemes, rhoptries, and dense granules of tachyzoites and in bradyzoites (2, 3, 28, 43). All these observations suggest that the conserved antigenicity between N. caninum and T. gondii might represent a rational basis for the development of efficient vaccines for the control of both parasitic diseases.A vaccine based on dead N. caninum tachyzoites is currently available for prophylaxy; this vaccine is thought to confer about 46% protection against N. caninum-induced abortion in cattle (36). However, in a number of countries, this vaccine has not been licensed, since more complete scientific documentation is required to authorize the use of a vaccine against N. caninum (8). The need for a more effective vaccine against transplacental infection in cattle is therefore of the utmost importance. Live vaccines are thought to induce complete protective immunity against N. caninum infection. In vaccination trials with the mouse model, the use of N. caninum tachyzoite crude extract as the immunogen resulted in an absence of protection against parasite-related neurological illness and death (5, 27). Such vaccinations have also proved ineffective for the prevention of abortion in cattle, even in the presence of adjuvants (42).Given that protective immunity against intracellular pathogens such as T. gondii and N. caninum involves T-cell-mediated immunity (12, 21) and that experimental evidence of protection against N. caninum transplacental transmission has been shown to involve high levels of gamma interferon (IFN-γ) production (17, 42), we propose an innovative approach based on heterologous vaccination.Taking into consideration the antigenic similarities between N. caninum and T. gondii, we used an attenuated strain of T. gondii as a heterologous vaccine against N. caninum. A mutant RH strain of T. gondii tachyzoites lacking the mic1 and mic3 genes was constructed in our laboratory, the “mic1-3KO strain.” The disruption of these two genes, both of which code for proteins involved in tachyzoite adhesion and invasion, greatly decreases virulence in mice (7). Vaccination with the mic1-3KO strain provides strong protection against chronic and congenital toxoplasmosis in mice through the induction of strong humoral and Th1 cellular immune responses (18). In this study, we used this attenuated strain as a heterologous vaccine. Our results provide evidence for protection against lethal N. caninum infection. This protection was associated with strong cross-reactive humoral and Th1 cellular immune responses, overcoming the biological and antigenic differences between the two species.  相似文献   

2.
We examined the susceptibilities to fluconazole of 642 bloodstream infection (BSI) isolates of Candida glabrata and grouped the isolates by patient age and geographic location within the United States. Susceptibility of C. glabrata to fluconazole was lowest in the northeast region (46%) and was highest in the west (76%). The frequencies of isolation and of fluconazole resistance among C. glabrata BSI isolates were higher in the present study (years 2001 to 2007) than in a previous study conducted from 1992 to 2001. Whereas the frequency of C. glabrata increased with patient age, the rate of fluconazole resistance declined. The oldest age group (≥80 years) had the highest proportion of BSI isolates that were C. glabrata (32%) and the lowest rate of fluconazole resistance (5%).Candidemia is without question the most important of the invasive mycoses (6, 33, 35, 61, 65, 68, 78, 86, 88). Treatment of candidemia over the past 20 years has been enhanced considerably by the introduction of fluconazole in 1990 (7, 10, 15, 28, 29, 31, 40, 56-58, 61, 86, 90). Because of its widespread usage, concern about the development of fluconazole resistance among Candida spp. abounds (2, 6, 14, 32, 47, 53, 55, 56, 59, 60, 62, 80, 86). Despite these concerns, fluconazole resistance is relatively uncommon among most species of Candida causing bloodstream infections (BSI) (5, 6, 22, 24, 33, 42, 54, 56, 65, 68, 71, 86). The exception to this statement is Candida glabrata, of which more than 10% of BSI isolates may be highly resistant (MIC ≥ 64 μg/ml) to fluconazole (6, 9, 15, 23, 30, 32, 36, 63-65, 71, 87, 91). Suboptimal fluconazole dosing practices (low dose [<400 mg/day] and poor indications) may lead to an increased frequency of isolation of C. glabrata as an etiological agent of candidemia in hospitalized patients (6, 17, 29, 32, 35, 41, 47, 55, 60, 68, 85) and to increased fluconazole (and other azole) resistance secondary to induction of CDR efflux pumps (2, 11, 13, 16, 43, 47, 50, 55, 69, 77, 83, 84) and may adversely affect the survival of treated patients (7, 10, 29, 40, 59, 90). Among the various Candida species, C. glabrata alone has increased as a cause of BSI in U.S. intensive care units since 1993 (89). Within the United States, the proportion of fungemias due to C. glabrata has been shown to vary from 11% to 37% across the different regions (west, midwest, northeast, and south) of the country (63, 65) and from <10% to >30% within single institutions over the course of several years (9, 48). It has been shown that the prevalence of C. glabrata as a cause of BSI is potentially related to many disparate factors in addition to fluconazole exposure, including geographic characteristics (3, 6, 63-65, 71, 88), patient age (5, 6, 25, 35, 41, 42, 48, 63, 82, 92), and other characteristics of the patient population studied (1, 32, 35, 51). Because C. glabrata is relatively resistant to fluconazole, the frequency with which it causes BSI has important implications for therapy (21, 29, 32, 40, 41, 45, 56, 57, 59, 80, 81, 86, 90).Previously, we examined the susceptibilities to fluconazole of 559 BSI isolates of C. glabrata and grouped the isolates by patient age and geographic location within the United States over the time period from 1992 to 2001 (63). In the present study we build upon this experience and report the fluconazole susceptibilities of 642 BSI isolates of C. glabrata collected from sentinel surveillance sites throughout the United States for the time period from 2001 through 2007 and stratify the results by geographic region and patient age. The activities of voriconazole and the echinocandins against this contemporary collection of C. glabrata isolates are also reported.  相似文献   

3.
In animal models, a defective Th1 response appears to be critical in the pathogenesis of brucellosis, but the Th1 response in human brucellosis patients remains partially undefined. Peripheral blood from 24 brucellosis patients was studied before and 45 days after antibiotherapy. Twenty-four sex- and age-matched healthy donors were analyzed in parallel. Significantly increased levels of interleukin 1β (IL-1β), IL-2, IL-4, IL-6, IL-12p40, gamma interferon (IFN-γ), and tumor necrosis factor alpha (TNF-α), but not of IL-10, in serum and/or significantly increased percentages of samples with detectable levels of these cytokines, measured by enzyme-linked immunosorbent assays (ELISA), were found for untreated brucellosis patients, but these levels were reduced and/or normalized after treatment. Flow cytometry studies showed that the intracytoplasmic expression of IFN-γ, IL-2, and TNF-α, but not that of IL-4, by phorbol myristate-activated CD4+ CD3+ and CD8+ CD3+ T lymphocytes was significantly increased in untreated brucellosis patients and was also partially normalized after antibiotherapy. The percentage of phagocytic cells, the mean phagocytic activity per cell, and the phagocytic indices for monocytes at baseline were defective and had only partially reverted at follow-up. T lymphocytes from untreated brucellosis patients are activated in vivo and show Th1 cytokine production polarization, with strikingly high serum IFN-γ levels. In spite of this Th1 environment, we found deficient effector phagocytic activity in peripheral blood monocytes.Brucellosis is a zoonotic disease of worldwide distribution. Despite its control in many countries, it remains endemic in the Mediterranean and Middle Eastern regions (20, 28, 41, 42). Brucella melitensis is the most frequent cause of human brucellosis in these geographical areas (19). In Spain, it has been reported that the majority (more than 97.5%) of isolates were identified as Brucella melitensis (13, 44, 45).Brucella organisms are facultatively intracellular Gram-negative coccobacilli that reside and replicate in a vacuolar compartment within myelomonocytic cells of the infected host (14, 15, 47). The response to Brucella involves the whole gamut of the immune system, from innate to adaptive immunity (21). In murine models, passive transfer of immune cells resulted in an effective anti-Brucella defensive response mediated by CD4+ and CD8+ T lymphocytes (5, 6, 32, 37, 51, 52). Furthermore, the pattern of T-lymphocyte cytokine secretion is considered to be critical for the effectiveness of the protective anti-Brucella immune response (3, 7). It has been postulated that Th1 cytokines confer resistance, while Th2 cytokines facilitate the development of brucellosis (2, 3, 24, 25, 40, 43, 52). In animal models, gamma interferon (IFN-γ) induces macrophage activation and control of Brucella infection (16, 18, 43). In Brucella-infected mice, administration of recombinant IFN-γ enhances host resistance, resulting in a deep decrease in the number of viable bacteria (51). Moreover, host IFN-γ depletion results in an increase in the number of viable bacteria (17, 37, 52). Several abnormalities in the immune system have been found in human brucellosis (27, 46, 49). It has been found that T and NK lymphocytes show defective functions in brucellosis patients (46, 49). Since mice are naturally resistant to Brucella infections, it is possible to suggest that the immune response elicited by Brucella in humans might have different characteristics. Thus, susceptibility to, or protection from, human brucellosis conferred by T-lymphocyte cytokines has not been established.In this work, we have further investigated the pattern of T-lymphocyte and monocyte responses to human Brucella infection. We have prospectively studied (i) the levels of Th1, Th2, and regulatory cytokines in serum, (ii) the distribution, activation stage, and pattern of Th1/Th2 cytokine production by T lymphocytes, and (iii) the phagocytic activity of monocytes in a group of brucellosis patients before and after antimicrobial treatment.  相似文献   

4.
Neospora caninum is an apicomplexan parasite closely related to Toxoplasma gondii. In nature this parasite is found especially in dogs and cattle, but it may also infect other livestock. The growth of N. caninum, which is an obligate intracellular parasite, is controlled mainly by the cell-mediated immune response. During infection the cytokine gamma interferon (IFN-γ) plays a prominent role in regulating the growth of N. caninum in natural and experimental disease. The present study showed that induction of the tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO) is responsible for the inhibition of parasite growth that is mediated by IFN-γ-activated bovine fibroblasts and endothelial cells. This antiparasite effect could be abrogated by addition of tryptophan, as well as by the IDO-specific inhibitor 1-l-methyltryptophan. In conclusion, our data show that human and bovine cells use the same effector mechanism to control the growth of N. caninum.Toxoplasma gondii, a protozoan belonging to the apicomplexan phylum, is one of the most successful parasites on earth. This parasite is capable of infecting nearly all warm-blooded animals, including humans. T. gondii can be transmitted via tissue cysts, raw meat, and environmental-resistant oocysts derived from cat feces and is able to spread transplacentally from mother to fetus. Furthermore, another special feature of this evolutionarily successful parasite is the fact that it usually causes asymptomatic infections and in most cases does not kill immunocompetent hosts. However, without sufficient therapy, reactivation of T. gondii in immunocompromised individuals frequently results in death of the host (31, 26, 18).In 1984 a T. gondii-like parasite was found in the cerebral tissue of dogs and described (5). This parasite was later detected in brain tissue from dogs which had clinical signs of neuromuscular disease and was named Neospora caninum (15). It took until 1998 to discover that dogs are not only intermediate hosts but also one of the definitive hosts of this parasite (29). In nature, dogs are frequently intermediate hosts of N. caninum, although canine neosporosis seems to be rare (2). N. caninum can also be isolated from cattle, and vertically transmitted N. caninum infection is considered an important cause of bovine abortion worldwide (17). In sheep N. caninum-associated abortion seems to be rare (16). This is in contrast to infections with T. gondii, which often cause abortion in sheep but seldom in cattle (14). Furthermore, so far, there is no evidence that N. caninum infection is zoonotic (16). It has been shown that under experimental conditions N. caninum is able to infect rhesus monkeys, indicating the zoonotic potential of this parasite. However, serologic studies with humans have shown that no or only small amounts of N. caninum-specific antibodies are detectable in some human sera, even sera from high-risk groups like farm workers (30, 16, 22). Despite the high levels of homology between T. gondii and N. caninum, many differences have also been detected. Both parasites can be transmitted via food, via oocysts in soil, and also transplacentally (23). Several species have been successfully infected experimentally with N. caninum, and in vitro N. caninum is capable of replicating in different types of cells derived from various animal species or humans.The variability in the susceptibility to natural T. gondii or N. caninum infection among various host species might be due to differences in the immune responses. Different antiparasite effector mechanisms might, at least in part, be involved in the evolutionary success of both parasites. In support of this, workers have obtained some data showing that experimental infection with attenuated or apathogenic N. caninum strains can induce immunity to this parasite in mice and cattle (3). Furthermore, a lot of data indicate that the cellular immune response is necessary to control infection with N. caninum. In addition, it was found that gamma interferon (IFN-γ), a product of activated T cells and natural killer (NK) cells, is one of the main cytokines conferring resistance to N. caninum (21). So far, the IFN-γ-induced effector mechanism that is active against N. caninum in cattle has not been defined.In this paper we provide evidence that induction of the tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO), which is the most prominent antiparasite effector mechanism active against T. gondii in human cells (28), is also effective for inhibiting N. caninum growth in tissue cells from humans and cattle.  相似文献   

5.
Acanthamoeba culbertsoni is an opportunistic pathogen that causes granulomatous amoebic encephalitis (GAE), a chronic and often fatal disease of the central nervous system (CNS). A hallmark of GAE is the formation of granulomas around the amoebae. These cellular aggregates consist of microglia, macrophages, lymphocytes, and neutrophils, which produce a myriad of proinflammatory soluble factors. In the present study, it is demonstrated that A. culbertsoni secretes serine peptidases that degrade chemokines and cytokines produced by a mouse microglial cell line (BV-2 cells). Furthermore, soluble factors present in cocultures of A. culbertsoni and BV-2 cells, as well as in cocultures of A. culbertsoni and primary neonatal rat cerebral cortex microglia, induced apoptosis of these macrophage-like cells. Collectively, the results indicate that A. culbertsoni can apply a multiplicity of cell contact-independent modes to target macrophage-like cells that exert antiamoeba activities in the CNS.Acanthamoeba culbertsoni belongs to a group of free-living amoebae, such as Balamuthia mandrillaris, Naegleria fowleri, and Sappinia pedata, that can cause disease in humans (46, 56). Acanthamoeba spp. are found worldwide and have been isolated from a variety of environmental sources, including air, soil, dust, tap water, freshwater, seawater, swimming pools, air conditioning units, and contaminated contact lenses (30). Trophozoites feed on bacteria and algae and represent the infective form (47, 56). However, under unfavorable environmental conditions, such as extreme changes in temperature or pH, trophozoites transform into a double-walled, round cyst (22, 45).Acanthamoeba spp. cause an infection of the eye known as amoebic keratitis (AK), an infection of the skin referred to as cutaneous acanthamoebiasis, and a chronic and slowly progressing disease of the central nervous system (CNS) known as granulomatous amoebic encephalitis (GAE) (22, 23, 30, 56). GAE is most prevalent in humans who are immunocompromised (30, 33, 40) and has been reported to occur among individuals infected with the human immunodeficiency virus (HIV) (28). It has been proposed that Acanthamoeba trophozoites access the CNS by passage through the olfactory neuroepithelium (32) or by hematogenous spread from a primary nonneuronal site of infection (23, 24, 33, 53).In immune-competent individuals, GAE is characterized by the formation of granulomas. These cellular aggregates consist of microglia, macrophages, polymorphonuclear cells, T lymphocytes, and B lymphocytes (24, 30). The concerted action of these immune cells results in sequestration of amoebae and is instrumental in slowing the progression of GAE. This outcome is consistent with the observation that granulomas are rarely observed in immunocompromised individuals (34) and in mice with experimentally induced immune suppression following treatment with the cannabinoid delta-9-tetrahydrocannabinol (Δ9-THC) (8).Microglia are a resident population of macrophages in the CNS. These cells, along with CNS-invading peripheral macrophages, appear to play a critical early effector role in the control of Acanthamoeba spread during GAE (4, 5, 29, 31). In vitro, microglia have been shown to produce an array of chemokines and cytokines in response to Acanthamoeba (31, 51). However, these factors appear not to have a deleterious effect on these amoebae (29).Acanthamoeba spp. produce serine peptidases, cysteine peptidases, and metallopeptidases (1, 2, 9, 10, 14, 16, 18, 19, 21, 25, 26, 37, 38, 41, 42, 52). In the present study, it is demonstrated that serine peptidases secreted by A. culbertsoni degrade chemokines and cytokines that are produced by immortalized mouse BV-2 microglia-like cells. In addition, soluble factors present in cocultures of A. culbertsoni and BV-2 cells induced apoptosis of the BV-2 cells. Collectively, these results suggest a mode through which A. culbertsoni can evade immune responsiveness in the CNS.  相似文献   

6.
We report the use of PCR techniques on a formalin-fixed and paraffin-embedded tissue specimen for direct detection of one dominant azole resistance mechanism in a case of disseminated invasive aspergillosis. Rapid detection of mutations associated with azole resistance directly in tissue significantly reduces diagnostic delay.Invasive infections due to Aspergillus fumigatus are associated with significant morbidity and mortality, although the prognosis of patients with invasive aspergillosis has improved with the clinical use of mold-active antifungal azoles, most notably voriconazole (9, 11). However, the survival of patients may be threatened by the emergence of azole resistance of aspergilli (1, 7, 13). Resistance is commonly due to point mutations in the cyp51A gene, which is the target for antifungal azoles (1, 4, 8, 13, 14). The isolates commonly exhibit a cross-resistant phenotype (4), and patients with azole-resistant disease may fail azole therapy (1, 7, 10, 12). One problem in the management of azole-resistant aspergillosis is the early detection of resistance as cultures are negative in up to 50% of patients with focal pulmonary lesions (2), and in vitro susceptibility testing takes at least 5 to 7 days to complete. In this report, molecular tools were utilized to rapidly confirm the diagnosis of disseminated azole-resistant aspergillosis.  相似文献   

7.
Helicobacter pylori is a human gastroduodenal pathogen that leads to active chronic inflammation characterized by T-cell responses biased toward a Th1 phenotype. It has been accepted that H. pylori infection induces a Th17 response. At mucosal sites, dendritic cells (DCs) have the capacity to induce effector T cells. Here, we evaluate the role of DCs in the H. pylori-induced interleukin-17 (IL-17) response. Immunohistochemistry and immunofluorescence were performed on human gastric mucosal biopsy samples and showed that myeloid DCs in H. pylori-infected patients colocalized with IL-23- and that IL-17-producing lymphocytes were present in H. pylori-infected antral biopsy samples. In parallel, human monocyte-derived DCs stimulated in vitro with live H. pylori cells produced significant levels of IL-23 in the absence of IL-12 release. The subsequent incubation of H. pylori-infected DCs with autologous CD4+ T cells led to gamma interferon (IFN-γ) and IL-17 expression. The inhibition of IL-1 and, to a lesser extent, IL-23 inhibited IL-17 production by T cells. Finally, isogenic H. pylori mutant strains not expressing major virulence factors were less effective in inducing IL-1 and IL-23 release by DCs and IL-17 release by T cells than parental strains. Altogether, we can conclude that DCs are potent inducers of IL-23/IL-17 expression following H. pylori stimulation. IL-1/IL-23 as well as H. pylori virulence factors seem to play an important role in mediating this response.Gram-negative Helicobacter pylori is a gastroduodenal pathogen identified as being the causative agent of a variety of disease including gastritis, peptic ulcer, gastric adenocarcinoma, and mucosa-associated lymphoma (23, 27, 41, 42). H. pylori infection of gastric mucosa leads to active chronic inflammation characterized by both a lymphocytic and neutrophil infiltrate with the induction of proinflammatory cytokines, mainly interleukin-1β (IL-1β), tumor necrosis factor alpha (TNF-α), IL-8, and IL-6 (13, 29).The H. pylori-specific gastric mucosal T-cell response is predominantly a CD4+ T-cell response polarized toward a T-helper 1 (Th1) phenotype with increased levels of gamma interferon (IFN-γ) (4, 38, 55). Although profound, this immune response does not clear the bacteria, and indeed, the cytokines secreted are more associated with pathogenesis (38, 45). Furthermore, neutrophil responses are associated with tissue damage and ulceration (7, 60). The release of the neutrophil chemoattractant IL-8 by gastric epithelial cells was previously shown to depend on the expression of an H. pylori virulence factor: the cytotoxin-associated gene (cag) pathogenicity island (PAI) (14, 62). The cag PAI encodes the immunodominant protein CagA and the type IV secretion system, which serves to transfer the bacterial CagA protein and other soluble factors, such as peptidoglycans, to the cytoplasm of the host cell (9, 52). Strains expressing the cag PAI have been associated with a more severe inflammatory response than that induced by cag PAI-negative strains (12). The cellular recognition of cag PAI-positive strains is mediated via signaling through the host-intracellular pathogen recognition molecule NOD1 (nucleotide-binding oligomerization domain 1), leading to NF-κB activation and the induction of proinflammatory responses (58).It was previously shown that H. pylori infection is also associated with a marked production of Th17 cytokines (2, 39, 44). By using real-time PCR and Western blotting, it was previously demonstrated that IL-17, a proinflammatory cytokine, is upregulated in H. pylori-infected stomach biopsy specimens in comparison to uninfected specimens (39). IL-17 is a cytokine that characterizes a distinct population of T cells, namely, Th17 (1, 28). IL-17 has been associated with chronic inflammatory conditions such as rheumatoid arthritis (10) and multiple sclerosis (37). In addition, IL-17 proinflammatory function leading to IL-8 stimulation raises the possibility that IL-17 may play a role during bacterial infections (39, 57). Major cytokines associated with the differentiation of human Th17 cells were identified to be IL-23, IL-1β, and IL-6 (11, 61). While IL-12 plays a key role in the differentiation of naïve T cells to Th1 cells, IL-23 promotes the expansion of Th17 cells. In contrast, IL-27, another IL-12 family member, has been shown to limit the development of Th17 cells (25). IL-12 and IL-23 are heterodimers with a shared subunit, p40. Both IL-23 and IL-12 are produced by activated antigen-presenting cells (APCs) such as DCs and macrophages (48, 53).DCs, which play a central role in the induction of adaptive immune responses, are widely distributed in tissues, including gastrointestinal mucosa (32, 33), and were previously shown to be capable of migrating through epithelial tight junctions to gain access to the gastrointestinal lumen (33, 49). Furthermore, we and others have shown that H. pylori interactions with DCs trigger maturation and activation events that lead to the production of cytokines, which are important for the induction and regulation of immune responses (5, 18, 34, 43, 46).Previous studies of DC activation by H. pylori have focused on the induction of the Th1-biased response. Much less is known about the mechanism of induction as well as the cells and cytokine stimuli responsible for the expression of IL-17 in Helicobacter infection. Here, we have reevaluated the role of DCs in the induction of immune responses to Helicobacter infection by addressing the interaction of H. pylori-infected DCs with CD4+ T lymphocytes in initiating a Th17 response.  相似文献   

8.
9.
10.
The pathogenic yeast Cryptococcus neoformans and C. gattii commonly cause severe infections of the central nervous system in patients with impaired immunity but also increasingly in immunocompetent individuals. Cryptococcus is phagocytosed by macrophages but can then survive and proliferate within the phagosomes of these infected host cells. Moreover, Cryptococcus is able to escape into the extracellular environment via a recently discovered nonlytic mechanism (termed expulsion or extrusion). Although it is well established that the host''s cytokine profile dramatically affects the outcome of cryptococcal disease, the molecular basis for this effect is unclear. Here, we report a systematic analysis of the influence of Th1, Th2, and Th17 cytokines on the outcome of the interaction between macrophages and cryptococci. We show that Th1 and Th17 cytokines activate, whereas Th2 cytokines inhibit, anticryptococcal functions. Intracellular yeast proliferation and cryptococcal expulsion rates were significantly lower after treatment with the Th1 cytokines gamma interferon and tumor necrosis factor alpha and the Th17 cytokine interleukin-17 (IL-17). Interestingly, however, the Th2 cytokines IL-4 and IL-13 significantly increased intracellular yeast proliferation while reducing the occurrence of pathogen expulsion. These results help explain the observed poor prognosis associated with the Th2 cytokine profile (e.g., in human immunodeficiency virus-infected patients).The two encapsulated yeast species Cryptococcus neoformans (serotypes A and D) and C. gattii (serotypes B and C), the causative agents of cryptococcosis, can cause life-threatening infections of the central nervous system (e.g., meningoencephalitis) (9).Initial infection with Cryptococcus is believed to occur via the inhalation of airborne propagules and the subsequent colonization of the respiratory tract (9). In mouse and rat model systems, C. neoformans is internalized by alveolar macrophages shortly after inhalation (17, 22). Furthermore, C. neoformans phagocytosis by mouse, rat, guinea pig, and human macrophages in vitro has been demonstrated repeatedly (8, 16, 37, 49) and is triggered by direct recognition of the yeast or by receptor-mediated recognition via complement or antibodies (38). However, Cryptococcus seems to have developed a unique method to manipulate host macrophages. After phagocytosis, C. neoformans can survive and proliferate within these infected host cells, eventually leading to macrophage lysis (2, 15, 17, 18, 33, 50). Moreover, a novel expulsive mechanism by which the yeast can exit macrophages without killing the host cell, thus avoiding a local inflammatory response, has recently been described (3, 34).Results from restriction fragment length polymorphism analyses suggest that initial infection with Cryptococcus often occurs in early childhood and can be followed by a long latent phase in immunocompetent individuals (21). However, C. neoformans is generally capable of disseminating to other organs within the human body and shows a predilection for the central nervous system, where it can lead to life-threatening meningitis and meningoencephalitis (27). Although the molecular basis of latency and expulsion is not known, the so-called Trojan Horse model suggests that replication in and eventual expulsion from macrophages may offer a potential explanation for how C. neoformans stays latent and spreads within the host without triggering immediate immune responses (11, 46). An improved understanding of the interaction between macrophages and Cryptococcus is therefore critical for the development of more effective therapies.In healthy hosts, the cryptococcal infection is usually self-limiting, suggesting effective clearance or maintenance in a latent state by phagocytic cells. The outcome of cryptococcosis depends on the immune status of the infected individual and the cytokine pattern generated in response to the pathogen. Although it is well established that the host''s cytokine profile dramatically affects the outcome of cryptococcal disease, the molecular basis for this effect is unclear. Both Th1 and Th2 cytokines are involved in protection against C. neoformans, but whereas Th1-associated cytokines are essential for natural immunity, Th2-associated immunity is not protective in mice (6, 24, 25). Increased expression of Th1 cytokines, such as tumor necrosis factor alpha (TNF-α) and gamma interferon (IFN-γ), results in improved fungal control (19, 29, 36, 53), while IFN-γ knockout mice show increased fungal burdens (4). In 2007, Müller et al. (39) showed a significant role for the Th17 response and the proinflammatory cytokine interleukin-17 (IL-17) in modulating the survival of Cryptococcus-infected mice. In contrast, Th2 cytokines such as IL-4 and IL-13 reduce the host''s ability to deal with C. neoformans in vivo (7, 14, 28, 39).Despite these observations from animal models, little work on the in vitro effects of Th1, Th17, and Th2 cytokines on the interaction between macrophages and Cryptococcus has been done. Here, we report a systematic study of cytokine influence on macrophage-Cryptococcus interactions for a representative selection of Cryptococcus strains. Our results demonstrate that Th1- and Th17-stimulated macrophages are significantly better at phagocytosing cryptococci and at controlling the intracellular proliferation of this pathogen than Th2-stimulated cells. In contrast, Th2-activated macrophages show a significantly lower rate of cryptococcal expulsion than Th1- or Th17-activated cells. Together, these data help explain the susceptibility phenotype associated with Th2 cytokine profiles in vivo.  相似文献   

11.
Ixodes scapularis ticks transmit a number of human pathogens, including the Lyme disease spirochete Borrelia burgdorferi. I. scapularis suppresses host immunity in the skin to promote feeding and systemically skew T-helper (Th)-cell differentiation toward Th2 cells in secondary lymphoid organs. Although components of tick saliva are known to influence Th-cell polarization, the mechanism whereby tick feeding in the skin modulates regional and systemic Th-cell responses is unknown. In this study, the role of the epidermal Langerhans cell (LC) subset of skin dendritic cells in tick-mediated Th1/Th2-cell immunomodulation was assessed. Mice deficient in LCs (Langerin-DTA mice) exhibited enhanced lymph node (LN) concanavalin A (ConA)-induced Th1 responses after tick infestation in comparison to results for uninfested Langerin-DTA or wild-type (WT) mice, whereas effects on Th2-cell production of interleukin 4 were more variable. Nonetheless, the altered T-cell response did not impact tick feeding or refeeding. Gamma interferon production by ConA-stimulated LN cells of both WT and LC-deficient mice was enhanced by as much as fourfold after B. burgdorferi-infected-tick feeding, indicating that immunomodulatory effects of tick saliva were not able to attenuate the Th1 immune responses induced by this pathogen. Taken together, these findings show a requirement for LCs in the tick-mediated attenuation of Th1 responses in regional lymph nodes but not in the spleens of mice and show that the presence of a pathogen can overcome the Th1-inhibitory effects of tick feeding on the host.Blood-feeding arthropods are responsible for a significant proportion of all vector-borne infections, which number in the hundreds of millions annually worldwide (35, 36). In the United States, Lyme disease, due to infection with the spirochete Borrelia burgdorferi, is the most common arthropod-transmitted disease, with more than 20,000 cases reported in 2007 alone (3a, 32). Ixodes scapularis is the primary vector for B. burgdorferi in North America, and it also transmits several other pathogens associated with emerging infectious diseases, including Anaplasma phagocytophilum and Babesia microti. Each of these pathogens has established a life cycle that requires their sequential passage between a vertebrate host and ticks, underscoring the importance of tick feeding in maintaining these pathogens in nature.Interaction between the tick and the blood meal host has a significant impact on the host immune system and has been shown to enhance pathogen transmission (24). I. scapularis ticks must remain attached to their hosts for several days to acquire their blood meals and consequently have developed strategies to avoid immune rejection by the host (8, 11, 18, 25, 26, 31, 34, 39). I. scapularis saliva has pharmacologic properties that allow feeding to occur without host detection of the tick, which vector-borne pathogens like B. burgdorferi have exploited to establish infection and survive in the mammalian host (25-27, 34). Proteomic analysis of tick saliva has revealed antihemostatic, anti-inflammatory, and immunomodulatory components, which are thought to subdue the host cutaneous immune response and promote ingestion of the blood meal (18, 26, 34). In addition to effects on local immunity, the feeding of I. scapularis ticks on mice has been reported to result in systemic immune deviation of T-helper (Th) responses toward the anti-inflammatory Th2 type, as assessed by mitogen (concanavalin A) stimulation of lymphocytes harvested from the spleens 7 to 10 days after tick placement (30, 40-42).Although many of the effects of tick feeding have been linked to components of tick saliva (9, 10, 26, 34, 38), the mechanism(s) whereby tick feeding leads to systemic modulation of Th-cell cytokine production, distant from the tick bite in the skin, remains poorly understood. Dendritic cells in the skin, including epidermal Langerhans cells (LCs) and dermal dendritic cells (DCs), are key antigen-presenting cells that prime T-cell responses in regional draining lymph nodes after skin disruption. LCs in particular perform a sentinel function by detecting foreign antigens in the skin and migrating to the lymph nodes, where they are thought to interact with T cells and influence the Th-cell response through specific cytokine production (28). Although they were originally thought to play a seminal role in the priming of T-cell responses, more-recent studies have revealed that they can also modulate T-cell priming by other antigen-presenting cells (14-17, 20, 28, 37). Disruption of E-cadherin attachments of LCs to keratinocytes in the skin can induce LC migration in an immature state in which they can induce tolerance in the host, in part through the secretion of cytokines that promote Th2 responses (12). LCs appear to suppress the development of Th1 responses, as has been shown by the increase in the type IV hypersensitivity reaction to the contact allergen 2,4-dinitrofluorobenzene in mice genetically deficient in epidermal LCs (Langerin-DTA mice) (14). Additionally, studies using a mouse model of skin allograft rejection demonstrated that an absence of LCs leads to graft rejection, suggesting that LCs play a regulatory role in skin graft acceptance (23). In this study, we evaluated the role of LCs in the modulation of regional and systemic T-cell responses by tick feeding. Our results implicate LCs in tick-feeding-induced suppression of Th1 responses in the regional draining lymph nodes but not in modulation of systemic splenic Th-cell responses.  相似文献   

12.
Fasciola hepatica is a helminth pathogen that drives Th2/Treg immune responses in its mammalian host. The parasite releases a large number of molecules that are critical to inducing this type of immune response. Here we have selected recombinant forms of two major F. hepatica secreted molecules, the protease cathepsin L (rFhCL1) and an antioxidant, sigma class glutathione transferase (rFhGST-si), to examine their interactions with dendritic cells (DCs). Despite enzymatic and functional differences between these molecules, both induced interleukin-6 (IL-6), IL-12p40, and macrophage inflammatory protein 2 (MIP-2) secretion from DCs and enhanced CD40 expression. While this induction was mediated by Toll-like receptor 4 (TLR4), the subsequent intracellular signaling pathways differed; rFhCL1 signaled through p38, and rFhGST-si mediated its effect via c-Jun N-terminal kinase (JNK), p38, p-NF-κBp65, and IRF5. Neither rFhCL1 nor rFhGST-si enhanced DC phagocytosis or induced Th2 immune responses in vivo. However, DCs matured in the presence of either enzyme attenuated IL-17 production from OVA peptide-specific T cells in vivo. In addition, DCs exposed to either antigen secreted reduced levels of IL-23. Therefore, both F. hepatica FhCL1 and FhGST-si modulate host immunity by suppressing responses associated with chronic inflammation—an immune modulatory mechanism that may benefit the parasite''s survival within the host.Dendritic cells (DCs) have a central role among innate immune cells in presenting antigen and priming naïve T cells to differentiate into Th1/Th17 and Th2/Treg subsets. In response to pathogen-associated molecular patterns (PAMPs) that bind to pattern recognition receptors (PPRs), DCs express surface molecules and produce cytokines that modulate the effector functions of responding T cells (29). While much is known of how DCs respond to bacterial and viral pathogens that drive Th1/Th17 subsets (13), comparatively little is understood about how these cells respond to and influence the Th2/Treg adaptive immune response to helminth parasites (33).Gene expression and proteomic analyses of DCs have revealed that remarkably few genes are induced following stimulation with helminth antigens (8, 17). DCs activated and matured in the presence of helminth antigens lack the classical markers, such as high levels of proinflammatory cytokines (interleukin-12 p70 [IL-12p70], tumor necrosis factor alpha [TNF-α], and nitric oxide) and expression of costimulatory markers (CD80 and CD86), observed in DCs matured with Toll-like receptor (TLR) ligands such as lipopolysaccharide (LPS) (41). Additionally, TLR-mediated activation of DCs can be inhibited significantly if they are first exposed to helminths or helminth-derived products (24, 29).Despite their limited maturation, helminth-primed DCs can nevertheless activate naïve T cells (35). Dendritic cells exposed to a soluble preparation of Schistosoma mansoni egg antigen (SEA) and either cocultured with naive T cells in vitro or injected into mice can polarize T-cell responses toward a Th2 phenotype (34). Similar findings have been reported for S. mansoni larval antigens (26, 27) and excretory-secretory (ES) material from Nippostrongylus brasiliensis (35), Acanthocheilonema viteae (52), and Echinococcus granulosus (43). However, Segura et al. found that adoptive transfer of DCs treated with ES from Heligmosomoides polygyrus resulted in suppression of both Th1 and Th2 responses in recipient mice (46). The same DCs promoted the differentiation of T cells with a regulatory phenotype and an ability to suppress effector CD4+ cell proliferation and cytokine secretion (46). Therefore, a diverse range of DC phenotypes can be induced by using complex mixtures of helminth antigens. For this reason, it is important to investigate the interactions of defined helminth-derived molecules with DCs and to elucidate the mechanism by which they alter DC function.The liver fluke Fasciola hepatica is an important global helminth of humans and livestock (36). During infection, this pathogen induces potent polarized Th2/Treg immune responses coincident with a suppression of Th1 cytokines (15, 16, 18, 39, 40). Furthermore, infection also results in the bystander suppression of Th1 responses to a concurrent bacterial infection or to immunization with a Th1-inducing bacterial vaccine (6, 19). We previously demonstrated that ES molecules of F. hepatica can mimic the immunomodulatory effect observed with active infection. One of the predominant secreted products, a cathepsin L1 cysteine protease (FhCL1), suppressed the onset of protective Th1 immune responses to bacterial infections in mice and prevented the development of a Th1 response to vaccination (11, 22). Another major antigen, comprising 4% of F. hepatica ES material, is the antioxidant glutathione transferase (FhGST) (30), which in dimeric form significantly inhibited the proliferation of rat spleen cells in response to concanavalin A (ConA) stimulation in vitro (7).Using recombinant forms of FhCL1 and FhGST, we show that both molecules partially activate DCs, via the PRR TLR4. However, despite activating DCs via different intracellular signaling pathways, both rFhCL1- and rFhGST-treated DCs suppressed the development of Th17 cells and did not induce the differentiation of Th2 cells. Our data suggest that helminth parasites secrete multiple molecules possessing a unique mechanism of modulation which suppresses inflammatory Th1/Th17 responses with redundancy, thus permitting the uninhibited development of Th2/Treg cells in response to other secretory molecules.  相似文献   

13.
14.
Moraxella catarrhalis is a human pathogen causing otitis media in infants and respiratory infections in adults, particularly patients with chronic obstructive pulmonary disease. The surface protein Hag (also designated MID) has previously been shown to be a key adherence factor for several epithelial cell lines relevant to pathogenesis by M. catarrhalis, including NCIH292 lung cells, middle ear cells, and A549 type II pneumocytes. In this study, we demonstrate that Hag mediates adherence to air-liquid interface cultures of normal human bronchial epithelium (NHBE) exhibiting mucociliary activity. Immunofluorescent staining and laser scanning confocal microscopy experiments demonstrated that the M. catarrhalis wild-type isolates O35E, O12E, TTA37, V1171, and McGHS1 bind principally to ciliated NHBE cells and that their corresponding hag mutant strains no longer associate with cilia. The hag gene product of M. catarrhalis isolate O35E was expressed in the heterologous genetic background of a nonadherent Haemophilus influenzae strain, and quantitative assays revealed that the adherence of these recombinant bacteria to NHBE cultures was increased 27-fold. These experiments conclusively demonstrate that the hag gene product is responsible for the previously unidentified tropism of M. catarrhalis for ciliated NHBE cells.Moraxella catarrhalis is a gram-negative pathogen of the middle ear and lower respiratory tract (29, 40, 51, 52, 69, 78). The organism is responsible for ∼15% of bacterial otitis media cases in children and up to 10% of infectious exacerbations in patients with chronic obstructive pulmonary disease (COPD). The cost of treating these ailments places a large financial burden on the health care system, adding up to well over $10 billion per annum in the United States alone (29, 40, 52, 95, 97). In recent years, M. catarrhalis has also been increasingly associated with infections such as bronchitis, conjunctivitis, sinusitis, bacteremia, pneumonia, meningitis, pericarditis, and endocarditis (3, 12, 13, 17-19, 24, 25, 27, 51, 67, 70, 72, 92, 99, 102-104). Therefore, the organism is emerging as an important health problem.M. catarrhalis infections are a matter of concern due to high carriage rates in children, the lack of a preventative vaccine, and the rapid emergence of antibiotic resistance in clinical isolates. Virtually all M. catarrhalis strains are resistant to β-lactams (34, 47, 48, 50, 53, 65, 81, 84). The genes specifying this resistance appear to be gram positive in origin (14, 15), suggesting that the organism could acquire genes conferring resistance to other antibiotics via horizontal transfer. Carriage rates as high as 81.6% have been reported for children (39, 104). In one study, Faden and colleagues analyzed the nasopharynx of 120 children over a 2-year period and showed that 77.5% of these patients became colonized by M. catarrhalis (35). These investigators also observed a direct relationship between the development of otitis media and the frequency of colonization. This high carriage rate, coupled with the emergence of antibiotic resistance, suggests that M. catarrhalis infections may become more prevalent and difficult to treat. This emphasizes the need to study pathogenesis by this bacterium in order to identify vaccine candidates and new targets for therapeutic approaches.One key aspect of pathogenesis by most infectious agents is adherence to mucosal surfaces, because it leads to colonization of the host (11, 16, 83, 93). Crucial to this process are surface proteins termed adhesins, which mediate the binding of microorganisms to human cells and are potential targets for vaccine development. M. catarrhalis has been shown to express several adhesins, namely UspA1 (20, 21, 59, 60, 77, 98), UspA2H (59, 75), Hag (also designated MID) (22, 23, 37, 42, 66), OMPCD (4, 41), McaP (61, 100), and a type 4 pilus (63, 64), as well as the filamentous hemagglutinin-like proteins MhaB1, MhaB2, MchA1, and MchA2 (7, 79). Each of these adhesins was characterized by demonstrating a decrease in the adherence of mutant strains to a variety of human-derived epithelial cell lines, including A549 type II pneumocytes and Chang conjunctival, NCIH292 lung mucoepidermoid, HEp2 laryngeal, and 16HBE14o-polarized bronchial cells. Although all of these cell types are relevant to the diseases caused by M. catarrhalis, they lack important aspects of the pathogen-targeted mucosa, such as the features of cilia and mucociliary activity. The ciliated cells of the respiratory tract and other mucosal membranes keep secretions moving out of the body so as to assist in preventing colonization by invading microbial pathogens (10, 26, 71, 91). Given this critical role in host defense, it is interesting to note that a few bacterial pathogens target ciliated cells for adherence, including Actinobacillus pleuropneumoniae (32), Pseudomonas aeruginosa (38, 108), Mycoplasma pneumoniae (58), Mycoplasma hyopneumoniae (44, 45), and Bordetella species (5, 62, 85, 101).In the present study, M. catarrhalis is shown to specifically bind to ciliated cells of a normal human bronchial epithelium (NHBE) culture exhibiting mucociliary activity. This tropism was found to be conserved among isolates, and analysis of mutants revealed a direct role for the adhesin Hag in binding to ciliated airway cells.  相似文献   

15.
Pseudallescheria boydii has long been known to cause white grain mycetoma in immunocompetent humans, but it has recently emerged as an opportunistic pathogen of humans, causing potentially fatal invasive infections in immunocompromised individuals and evacuees of natural disasters, such as tsunamis and hurricanes. The diagnosis of P. boydii is problematic since it exhibits morphological characteristics similar to those of other hyaline fungi that cause infectious diseases, such as Aspergillus fumigatus and Scedosporium prolificans. This paper describes the development of immunoglobulin M (IgM) and IgG1 κ-light chain monoclonal antibodies (MAbs) specific to P. boydii and certain closely related fungi. The MAbs bind to an immunodominant carbohydrate epitope on an extracellular 120-kDa antigen present in the spore and hyphal cell walls of P. boydii and Scedosporium apiospermum. The MAbs do not react with S. prolificans, Scedosporium dehoogii, or a large number of clinically relevant fungi, including A. fumigatus, Candida albicans, Cryptococcus neoformans, Fusarium solani, and Rhizopus oryzae. The MAbs were used in immunofluorescence and double-antibody sandwich enzyme-linked immunosorbent assays (DAS-ELISAs) to accurately differentiate P. boydii from other infectious fungi and to track the pathogen in environmental samples. Specificity of the DAS-ELISA was confirmed by sequencing of the internally transcribed spacer 1 (ITS1)-5.8S-ITS2 rRNA-encoding regions of environmental isolates.Pseudallescheria boydii is an infectious fungal pathogen of humans (7, 16, 40, 58, 59). It is the etiologic agent of white grain mycetoma in immunocompetent humans (7) and has emerged over recent years as the cause of fatal disseminated infections in individuals with neutropenia, AIDS, diabetes, renal failure, bone marrow or solid organ transplants, systemic lupus erythematous, and Crohn''s disease; in those undergoing corticosteroid treatment; and in leukemia and lymphoma patients (1, 2, 3, 18, 27, 31, 32, 34, 36, 37, 38, 47, 49, 52). The fungus is the most prevalent species after Aspergillus fumigatus in the lungs of cystic fibrosis patients (8), where it causes allergic bronchopulmonary disease (5) and chronic lung lesions simulating aspergillosis (24). Near-drowning incidents and recent natural disasters, such as the Indonesian tsunami in 2004, have shown P. boydii and the related species Scedosporium apiospermum and Scedosporium aurantiacum to be the causes of fatal central nervous system infections and pneumonia in immunocompetent victims who have aspirated polluted water (4, 11, 12, 21, 22, 25, 30, 33, 57). Its significance as a potential pathogen of disaster evacuees has led to its recent inclusion in the Centers for Disease Control and Prevention list of infectious etiologies in persons with altered mental statuses, central nervous system syndromes, or respiratory illness.P. boydii is thought to be an underdiagnosed fungus (60), and misidentification is one of the reasons that the mortality rate due to invasive pseudallescheriasis is high. Detection of invasive P. boydii infections, based on cytopathology and histopathology, is problematic since it can occur in tissue and bronchoalveolar and bronchial washing specimens with other hyaline septated fungi, such as Aspergillus and Fusarium spp. (7, 23, 53, 60), which exhibit similar morphological characteristics upon microscopic examination (2, 23, 24, 28, 37, 44, 53, 60). Early diagnosis of infection by P. boydii and differentiation from other agents of hyalohyphomycosis is imperative, since it is refractory to antifungal compounds, such as amphotericin B, that are commonly administered for the control of fungal infections (10, 39, 58).The immunological diagnosis of Pseudallescheria infections has focused on the detection of antigens by counterimmunoelectrophoresis, and by immunohistological techniques using polyclonal fluorescent antibodies, but cross-reactions with antigens from other fungi, such as Aspergillus species, occurs (7, 19, 23). Pinto and coworkers (41, 42) isolated a peptidorhamnomannan from hyphae of P. boydii and proposed the antigen as a diagnostic marker for the pathogen. Cross-reactivity with Sporothrix schenckii and with Aspergillus have, however, been noted (23, 41). Furthermore, it is uncertain whether a similar antigen is present in the related pathogenic species S. prolificans, an important consideration in patient groups susceptible to mixed Scedosporium infections (6, 18).Hybridoma technology allows the production of highly specific MAbs that are able to differentiate between closely related species of fungi (54, 55, 56). The purpose of this paper is to report the development of MAbs specific to P. boydii and certain closely related species and their use to accurately discriminate among P. boydii, A. fumigatus, and other human pathogenic fungi by using immunofluorescence and double-antibody sandwich enzyme-linked immunosorbent assays (DAS-ELISAs).Currently, the natural environmental habitat of P. boydii is unknown, but nutrient-rich, brackish waters, such as estuaries, have been suggested (9, 17). In combination with a semiselective isolation procedure, I show how the DAS-ELISA can be used to rapidly and accurately track the pathogen in naturally infested estuarine muds, and in doing so illustrate the potential of the DAS-ELISA as a diagnostic platform for detection of P. boydii and related species within the Pseudallescheria complex.  相似文献   

16.
Mannheimia haemolytica is an important member of the bovine respiratory disease complex, which is characterized by abundant neutrophil infiltration into the alveoli and fibrin deposition. Recently several authors have reported that human neutrophils release neutrophil extracellular traps (NETs), which are protein-studded DNA matrices capable of trapping and killing pathogens. Here, we demonstrate that the leukotoxin (LKT) of M. haemolytica causes NET formation by bovine neutrophils in a CD18-dependent manner. Using an unacylated, noncytotoxic pro-LKT produced by an ΔlktC mutant of M. haemolytica, we show that binding of unacylated pro-LKT stimulates NET formation despite a lack of cytotoxicity. Inhibition of LKT binding to the CD18 chain of lymphocyte function-associated antigen 1 (LFA-1) on bovine neutrophils reduced NET formation in response to LKT or M. haemolytica cells. Further investigation revealed that NETs formed in response to M. haemolytica are capable of trapping and killing a portion of the bacterial cells. NET formation was confirmed by confocal microscopy and by scanning and transmission electron microscopy. Prior exposure of bovine neutrophils to LKT enhanced subsequent trapping and killing of M. haemolytica cells in bovine NETs. Understanding NET formation in response to M. haemolytica and its LKT provides a new perspective on how neutrophils contribute to the pathogenesis of bovine respiratory disease.Mannheimia haemolytica is a member of the bovine respiratory disease complex (BRD), causing a severe fibrinous pleuropneumonia sometimes referred to as shipping fever. The pneumonia is characterized by intense neutrophil infiltration in alveoli, intra-alveolar hemorrhage, fibrin deposition, and consolidation of the lungs (42, 56). The importance of neutrophils in the production of inflammatory mediators, recruitment of other leukocytes, and lung damage (17, 56, 67, 74) was demonstrated in calves that were depleted of neutrophils before challenge with M. haemolytica (10, 56). Neutrophil-depleted calves displayed less lung pathology than did control calves infected with M. haemolytica (10, 56). From these data, it is clear that neutrophils are a key player in the pathology of bovine pleuropneumonia; however, the mechanisms by which they contribute to host defense and tissue destruction are not clearly defined.The most important virulence factor for M. haemolytica is its leukotoxin (LKT), a 104-kDa exotoxin produced during logarithmic-phase growth (18, 32). LKT is a member of the repeats-in-toxin (RTX) toxin family of exoproteins produced by a wide variety of Gram-negative bacteria, including Escherichia coli, Actinobacillus pleuoropneumoniae, and Aggregatibacter actinomycetemcomitans (70). RTX toxins are characterized by a C-terminal glycine-rich nonapeptide repeat region (-G-G-X-G-X-D-X-U-X, where U is a hydrophobic residue) that binds calcium (Ca2+). The latter is required for membrane binding and cytotoxicity (30, 70). RTX toxins can insert into the plasma membrane of target cells, causing lysis and necrotic cell death (30, 70). The N-terminal domain contains amphipathic and hydrophobic domains believed to be required for pore stabilization and formation, respectively (70). More recently, it was shown that LKT also causes apoptosis via a caspase 9-dependent pathway and that LKT is internalized and transported via the cytoskeleton to mitochondria (4-6).The leukotoxin operon contains the genes lktC, lktA, lktB, and lktD (36, 37, 58). lktA encodes the inactive pro-LKT protein that is not cytotoxic until acylated (62) by the transacylase encoded by lktC. lktB and lktD encode proteins responsible for leader sequence-independent secretion of LKT from the bacterial cell (36, 37, 58). The acylated LKT then binds the CD18 chain of the β2-integrin lymphocyte function-associated antigen 1 (LFA-1) (3, 21-26, 33, 40, 41, 44, 55, 63) on ruminant leukocytes. LKT binding to amino acids 5 to 17 of the signal sequence of CD18 is required for cell death and restricts cytotoxicity to ruminant leukocytes, because the signal sequence for CD18 is not present on mature leukocytes from other mammalian species (55). Other investigators have shown that both the pro- form and mature LKT are capable of binding CD18, although the pro-LKT does not cause cytotoxicity (62). No biological role has been assigned to the pro- form of LKT.Recently, several authors have shown that human neutrophils are able to undergo a form of cell death, called NETosis, that is distinct from apoptosis and necrosis (12, 13, 31, 51, 69). NETosis is defined as the release of nuclear DNA from an activated neutrophil into the extracellular environment, with little concomitant release of lactate dehydrogenase (LDH) (12). The extracellular DNA and associated proteins (e.g., histones) released by activated neutrophils have been termed neutrophil extracellular traps (NETs) (12). There are four steps leading to NET formation. These are neutrophil activation, nuclear envelope degradation, mixing of nuclear DNA with cytosolic proteins, and extrusion of the DNA-protein mixture from the cell (31). Treatment of human neutrophils with interleukin-8 (IL-8), phorbol 12-myristate 13-acetate (PMA), or lipopolysaccharide (LPS) causes NET formation (12, 31, 69). NET formation also occurs in response to prokaryotic and eukaryotic pathogens (12, 35, 64). To date, no bacterial exotoxin has been shown to cause NET formation.NETs are composed of extracellular DNA that is studded with antimicrobial proteins. The latter include nuclear histones and primary, secondary, and tertiary granular components such as neutrophil elastase, myeloperoxidase, lactoferrin, and gelatinase (51, 69). When neutrophils become activated and commit to NET formation, they also are capable of trapping and killing pathogens. To date, NETs have been shown to kill a variety of Gram-negative and Gram-positive bacteria, fungi, and protozoans (2, 7-9, 12, 13, 15, 19, 20, 27, 28, 31, 34, 35, 43, 50-53, 59, 64, 67, 70). Here, we examine if M. haemolytica and its LKT cause NET formation by bovine neutrophils and whether NETs are capable of trapping and killing M. haemolytica cells in vitro.  相似文献   

17.
Yersinia pestis, the causative agent of plague, evades host immune responses and rapidly causes disease. The Y. pestis adhesin Ail mediates host cell binding and is critical for Yop delivery. To identify the Ail receptor(s), Ail was purified following overexpression in Escherichia coli. Ail bound specifically to fibronectin, an extracellular matrix protein with the potential to act as a bridge between Ail and host cells. Ail expressed by E. coli also mediated binding to purified fibronectin, and Ail-mediated E. coli adhesion to host cells was dependent on fibronectin. Ail expressed by Y. pestis bound purified fibronectin, as did the Y. pestis adhesin plasminogen activator (Pla). However, a KIM5 Δail mutant had decreased binding to host cells, while a KIM5 Δpla mutant had no significant defect in adhesion. Furthermore, treatment with antifibronectin antibodies decreased Ail-mediated adhesion by KIM5 and the KIM5 Δpla mutant, indicating that the Ail-fibronectin interaction was important for cell binding. Finally, antifibronectin antibodies inhibited the KIM5-mediated cytotoxicity of host cells in an Ail-dependent fashion. These data indicate that Ail is a key adhesin that mediates binding to host cells through interaction with fibronectin on the surface of host cells, and this interaction is important for Yop delivery by Y. pestis.The three species of Yersinia pathogenic for humans, Yersinia enterocolitica, Y. pseudotuberculosis, and Y. pestis, cause distinct diseases. Y. pseudotuberculosis and Y. enterocolitica typically cause acute gastroenteritis and mesenteric lymphadenitis. On the other hand, Y. pestis, the causative agent of the plague, is one of the most deadly human infectious diseases (8). Y. pestis is a close relative of Y. pseudotuberculosis, diverging only 1,500 to 20,000 years ago (1). To accommodate flea-borne transmission, Y. pestis has acquired two unique plasmids not harbored by enteropathogenic Yersinia species. All three pathogenic Yersinia species inject cytotoxic Yersinia outer proteins (Yops) into host cells via the Ysc type III secretion system (TTSS) to establish an infection (11). Host cell contact is essential for engagement of the TTSS and secretion of Yops (9, 54). Within the host cell, Yops effect actin rearrangements, inhibit phagocytosis, and block proinflammatory signals (4, 40, 42). Both Y. enterocolitica and Y. pseudotuberculosis express the well-studied adhesin molecules invasin (Inv) and YadA, capable of mediating Yop delivery (9, 54). However, Y. pestis does not express either adhesin due to an IS1541 element insertion within inv (58) and a frameshift mutation in yadA (44, 55). Y. pestis has a number of other adhesins capable of mediating host cell interaction. Both the pH 6 antigen (Psa [29, 63]) and plasminogen activator (Pla [28]) of Y. pestis have been shown to be adhesins. Psa is a tightly regulated pilus expressed at a pH of <6.7 and 37°C (52, 67) and is known to bind to β-linked galactosylated glycosphingolipids (46), low-density lipoprotein (31), and human IgG (69). Pla, expressed at 26°C but further induced at 37°C (49), is known to bind to several extracellular matrix components (23, 28, 30). The putative autotransporter, YapC, is also capable of mediating cell adhesion when it is expressed in Escherichia coli (15), as is the pilus encoded by the chaperone/usher system locus y0561-0563 (16), but neither yapC nor y0561-0563 results in significantly decreased adhesion when they are deleted from Y. pestis (15, 16).Recently, an additional adhesin of Y. pestis, Ail (adherence and invasion locus), was determined to facilitate cell binding (14, 25). Ail (encoded by y1324) is a 21.5-kDa outer membrane protein of the OmpX family that is predicted to have eight transmembrane domains and four extracellular loops extending above the surface of the bacterium (17, 65). Ail homologues include OmpX of Escherichia coli (32) and Enterobacter cloacae (61), PagC in Salmonella (53), and Opa proteins from Neisseria (10). Ail from Y. enterocolitica has been studied previously and shown to have three activities: cell adhesion, cell invasion (36), and the ability to confer serum resistance (5, 51) by binding to complement regulatory proteins (24). The residues for all three activities have been mapped to particular amino acids in the surface-exposed loops (35). Y. pseudotuberculosis Ail also confers adhesion and invasion functions (T. M. Tsang and E. S. Krukonis, unpublished data) and serum resistance (68), although the two amino acid changes between Y. pseudotuberculosis Ail and Y. pestis Ail result in decreased adhesion and invasion mediated by the former (Tsang and Krukonis, unpublished). More recently, Y. pestis Ail was also shown to mediate cell adhesion (14, 25), autoaggregation (25), and serum resistance (3, 24, 25) and to facilitate Yop delivery to host cells (14). Furthermore, Y. pestis Ail is required for virulence, as a Y. pestis Δail mutant has a >3,000-fold increase in the 50% lethal dose (14). A Y. pestis Δail mutant shows reduced binding to both epithelial and phagocytic human-derived cell lines, and in a mouse model of infection, a Y. pestis KIM5 Δail mutant colonizes host tissue to much lower levels than the parental KIM5 strain (14). Over the course of 7 days, the Δail mutant is cleared from the host (14). Together, these data demonstrate that Ail is an important adhesin that contributes to colonization and virulence.Cell adhesion is important for the establishment of a successful infection. Adhesion is also significant in Y. pestis pathogenesis because host cell contact is required for the production and translocation of the Yop effector proteins (48, 54). Bacteria can bind directly to host cell receptors (21) or use molecules like extracellular matrix (ECM) components to mediate attachment to host cells (12, 22, 30, 45, 57, 64). Common components of the cellular matrix that facilitate bacterial binding include fibronectin (22, 28, 64), collagen (23, 45), and laminin (28, 30, 45). Interactions between bacteria and ECM can lead to bridge-like attachments to host cells.Fibronectin is a large glycoprotein that is a key structural component in many tissues. This ∼220-kDa protein is commonly found as a dimer that is linked by two disulfide bonds located near the C terminus. Fibronectin is a complex molecule made up of three types of modular repeating units (43, 47). Fibronectin can bind to many substrates, including collagen (13), integrin receptors on host cells (50, 56), and heparin (60). Additionally, fibronectin contains a binding site for several bacterial pathogens at the N-terminal end of the molecule (39, 59).A number of fibronectin binding proteins on bacterial pathogens have been identified and studied, including SigB from Staphylococcus aureus (34), protein F from Streptococcus pyogenes (41), and YadA from Y. pseudotuberculosis (12, 19) and Y. enterocolitica (64). Binding of Y. pseudotuberculosis YadA to fibronectin allows Y. pseudotuberculosis to utilize β1 integrins on the surface of host cells for invasion (12). Given the key role of Y. pestis Ail in cell adhesion, Yop delivery, and virulence, we sought to determine the component on host cells to which Ail binds.Although Ail has been studied extensively in other Yersinia species, the substrate on host cells with which Ail interacts is not known. In this study, we used a purified Y. pestis Ail to identify the extracellular matrix component, fibronectin, as a protein bound by Ail. Furthermore, Ail-mediated binding to host cells through fibronectin is important for the delivery of Yop effector proteins.  相似文献   

18.
Rift Valley fever (RVF) is an epizootic viral disease of sheep that can be transmitted from sheep to humans, particularly by contact with aborted fetuses. A capripoxvirus (CPV) recombinant virus (rKS1/RVFV) was developed, which expressed the Rift Valley fever virus (RVFV) Gn and Gc glycoproteins. These expressed glycoproteins had the correct size and reacted with monoclonal antibodies (MAb) to native glycoproteins. Mice vaccinated with rKS1/RVFV were protected against RVFV challenge. Sheep vaccinated with rKS1/RVFV twice developed neutralizing antibodies and were significantly protected against RVFV and sheep poxvirus challenge. These findings further document the value of CPV recombinants as ruminant vaccine vectors and support the inclusion of RVFV genes encoding glycoproteins in multivalent recombinant vaccines to be used where RVF occurs.Rift Valley fever (RFV) virus (RVFV) is a mosquito-borne member of the genus Phlebovirus, family Bunyaviridae. It is widely distributed in Africa, causing endemic and epidemic disease in both humans and livestock, including sheep, cattle, and goats. RVF was first described in Kenya and was shown to be caused by a filterable virus transmissible via blood (9). Acute RVF in lambs is characterized by fever and death within 24 to 48 h of being detected (43). Signs in adult sheep include fever, mucopurulent nasal discharge, hemorrhagic diarrhea, and abortion in pregnant ewes (43). RVFV can be transmitted from infected sheep to humans, particularly when humans are exposed to aborted sheep fetuses and blood.Attenuated live RVFV vaccines are available for use in livestock. A mutagen-attenuated RVFV vaccine induces protective immune responses in lambs and appears to be safe (25); however, other studies documented teratogenic effects on lambs from vaccinated pregnant ewes similar to those caused by the attenuated RVFV strain Smithburn (18). An inactivated RVFV vaccine induces neutralizing antibody responses in humans (33), and its use in sheep would not induce teratogenic effects or abortions. However, the inactivated vaccine requires 3 doses (33) and is expensive to produce. Efforts to make RVFV vaccines without these disadvantages include an attenuated RVFV developed by reverse genetics and lacking the NSs and NSm genes (4) and other new-generation RVFV vaccines (reviewed in reference 19) that protect mice against virus challenge (7, 16, 24, 27).The middle (M) RNA segment of the RVFV genome encodes the viral glycoproteins Gn and Gc (8, 20), and recombinant vaccinia virus expressing these glycoproteins induces neutralizing antibody and protective immunity to RVFV in mice (7). Vaccinia virus is safe for animals, but there is some risk to humans, as it was reported previously to spread from human vaccinees to contacts (28, 55) and to cause serious clinical disease in human immunodeficiency virus-infected patients (36). Although modified vaccinia virus Ankara is a safer alternative for humans (6, 57), there are animal poxviruses with naturally restricted host ranges for vaccine vectors in animals (1, 13, 30, 31, 40, 46, 47, 52, 53).For ruminants, the genus Capripoxvirus (CPV) of the family Poxviridae has been an effective recombinant vector to induce protective immunity against several other viruses (3, 17, 29, 32, 40, 41, 51). This genus has three closely related species causing sheep pox, goat pox, and lumpy skin disease (LSD) of cattle. A recombinant LSD vaccine expressing the Gn and Gc glycoproteins of RVFV induced protection against RVFV challenge in mice (52, 53) and sheep (52). The three species of CPV have 96 to 97% nucleotide identity (49) and are restricted to ruminants, with no evidence of human infections (10, 11). Furthermore, attenuated CPV vaccines are in use in Africa and the Middle East to control ruminant poxvirus disease (11, 21). The use of a CPV vector to deliver virus vaccines to ruminants also induces immunity to the CPV vector, thus increasing the valence of the vaccine (3, 17, 39, 40). We report here the construction of a recombinant CPV that expresses the RVFV Gn and Gc glycoproteins and induces protective immunity against RVFV and sheep poxvirus (SPV) challenge in sheep.  相似文献   

19.
Cryptococcus neoformans and distantly related fungal species release extracellular vesicles that traverse the cell wall and contain a varied assortment of components, some of which have been associated with virulence. Previous studies have suggested that these extracellular vesicles are produced in vitro and during animal infection, but the role of vesicular secretion during the interaction of fungi with host cells remains unknown. In this report, we demonstrate by fluorescence microscopy that mammalian macrophages can incorporate extracellular vesicles produced by C. neoformans. Incubation of cryptococcal vesicles with murine macrophages resulted in increased levels of extracellular tumor necrosis factor alpha (TNF-α), interleukin-10 (IL-10), and transforming growth factor β (TGF-β). Vesicle preparations also resulted in a dose-dependent stimulation of nitric oxide production by phagocytes, suggesting that vesicle components stimulate macrophages to produce antimicrobial compounds. Treated macrophages were more effective at killing C. neoformans yeast. Our results indicate that the extracellular vesicles of C. neoformans can stimulate macrophage function, apparently activating these phagocytic cells to enhance their antimicrobial activity. These results establish that cryptococcal vesicles are biologically active.Cryptococcus neoformans is an encapsulated yeast that causes disease in diverse species, including humans. Infection is most commonly acquired by inhalation of environmental propagules. C. neoformans rarely causes disease in immunocompetent individuals, but patients with immunological disorders can develop disseminated and neural cryptococcosis (63).Extracellular microbial products have been amply demonstrated to modulate the interaction between host cells and pathogens. Many virulence factors and immunogens are released in their soluble forms by fungal cells to the extracellular space (4, 9, 16, 19, 37, 49, 53, 60, 62, 65, 67). C. neoformans, for instance, constitutively secretes large amounts of its capsular polysaccharide glucuronoxylomannan (GXM) (61). Disease progress is associated with detection of GXM, which is a potent modulator of the immune response (reviewed in reference 81). Other secreted virulence-related factors include galactoxylomannan (GalXM) (14), phospholipases (16), and urease (12, 62). In addition to acting as virulence factors, culture supernatant components are immunogenic, conferring protection against C. neoformans infection (51, 53).Phagocytes are particularly important effector cells in the control of systemic mycoses (54). The interaction of C. neoformans with phagocytes, including macrophages, monocytes, dendritic cells, and neutrophils, has been widely studied (23, 32, 43, 46, 50, 59, 68, 77). Cryptococcal GXM is antiphagocytic (34) and a powerful immunomodulator (45, 79). C. neoformans capsule size directly correlates with the efficacy of phagocytosis in vitro (6, 15, 82). Phagocytosis of C. neoformans can result in either fungal killing (24, 30) or survival (2, 3, 39-41, 71, 80). Killing of C. neoformans apparently involves the production of oxidative species (24), while the mechanisms of fungal escape include phagosome extrusion, cell-to-cell spread, and phagosomal permeabilization (2, 3, 40, 41, 71). Capsular polysaccharides and melanin are known to modulate the interaction of C. neoformans with phagocytes in favor of the fungus (27, 39, 47, 48, 71, 72, 74, 76), but the role of other structures in the outcome of yeast phagocytosis is virtually unknown.A number of recent studies have shown that GXM, GalXM, pigments, proteins, and lipids are trafficked in vesicles that traverse the cell wall (7, 14, 20, 56, 57, 62, 64, 65). Extracellular vesicles are also produced by the pathogens Candida albicans, C. parapsilosis, Sporothrix schenckii, and Histoplasma capsulatum, as well as by the model yeast Saccharomyces cerevisiae (1), suggesting that extracellular vesicle secretion is a general property of fungal cells. Secreted vesicles are heterogeneous. For instance, vesicles secreted by C. neoformans were classified into four different groups based on morphology and electron density (64). Additionally, vesicle diameter ranges from 30 to 400 nm, with the majority having dimensions of 100 to 150 nm (20, 64, 65). The combined use of serology, biochemistry, proteomics, and lipidomics led to the identification of 2 polysaccharides, phospholipids, 4 neutral lipids, and 76 proteins as extracellular vesicle components secreted by C. neoformans, which means that at least 81 different molecules are released to the extracellular milieu by vesicular secretion (14, 57, 64). It is likely that this number is an underestimate resulting from the difficulty of proteomic studies in vesicles from highly encapsulated cryptococcal cells, since a higher number of vesicular proteins were characterized in other fungi. For example, in H. capsulatum, proteomics and lipidomics of extracellular vesicles revealed an even more complex composition, including 283 proteins and 17 different phospholipids (1).In this study, we evaluated the influence of extracellular vesicles on the fate of C. neoformans after phagocytosis by mouse macrophages. Our results show that fungal vesicles are biologically active and stimulate macrophages. Moreover, our results demonstrate that vesicles from an acapsular mutant strain were more effective in eliciting macrophage activation and augmenting fungal killing than vesicles from encapsulated strains. Taken together, our findings suggest that fungal secretory vesicles have the potential to influence the interaction of C. neoformans with host cells.  相似文献   

20.
Toxoplasma gondii modulates pro- and anti-inflammatory responses to regulate parasite multiplication and host survival. Pressure from the immune response causes the conversion of tachyzoites into slowly dividing bradyzoites. The regulatory mechanisms involved in this switch are poorly understood. The aim of this study was to investigate the immunomodulatory role of T. gondii cyclophilin 18 (TgCyp18) in macrophages and the consequences of the cellular responses on the conversion machinery. Recombinant TgCyp18 induced the production of nitric oxide (NO), interleukin-12 (IL-12), and tumor necrosis factor alpha through its binding with cysteine-cysteine chemokine receptor 5 (CCR5) and the production of gamma interferon and IL-6 in a CCR5-independent manner. Interestingly, the treatment of macrophages with TgCyp18 resulted in the inhibition of parasite growth and an enhancement of the conversion into bradyzoites via NO in a CCR5-dependent manner. In conclusion, T. gondii possesses sophisticated mechanisms to manipulate host cell responses in a TgCyp18-mediated process.Toxoplasma gondii is a ubiquitous protozoan parasite that is able to infect a broad range of warm-blooded animals, including humans (17, 38). Fortunately, T. gondii is a well-adapted parasite which generally causes very little disease unless the host''s immune system is compromised in situations such as AIDS (41). Toxoplasma gondii affects pro- and anti-inflammatory host cell signaling in such a way as to maximize parasite multiplication and spread while maintaining host survival (14). One aspect of this manipulation is the upregulation of the interleukin-12 (IL-12)-dependent production of gamma interferon (IFN-γ), which is critical to host survival of acute toxoplasmosis (7, 8, 26, 28). This effect appears to occur by a pathway unique to T. gondii and involves the triggering of cysteine-cysteine chemokine receptor 5 (CCR5) in dendritic cells (DC) and macrophages by secreted T. gondii cyclophilin 18 (TgCyp18) (2). High et al. previously isolated genes encoding two Toxoplasma gondii cyclophilins, TgCyp18 and TgCyp20 (27). In T. gondii, the isolation of cyclosporine-binding proteins on affinity columns yielded only the cyclophilins TgCyp18 and TgCyp20 (27). Both cyclophilins were highly similar to human cyclophilin (hCyp18) in the central core region, but TgCyp20 differed in a 7-amino-acid “insertion” in the same region as that in Plasmodium falciparum cyclophilins (27).TgCyp18, but not hCyp18 or P. falciparum cyclophilin 19A (PfCyp19A), appears to induce IL-12 production by interacting directly with CCR5, an effect that was blocked by the addition of cyclosporine (2, 4, 63). These observations implied that structural determinants of TgCyp18, related to cyclosporine binding, were responsible for the induction of IL-12 synthesis (4, 63). This idea was confirmed by modeling of the TgCyp18 structure on that of PfCyp19A and site-directed mutagenesis of putatively surface-exposed residues that were absent in PfCyp19A (63). Two of the TgCyp18 mutants, namely, 17GEH19 to 17AAA19 and 149RP150 to 149YV150, located in the N and C termini of the protein, respectively, had reduced interactions with CCR5 and reduced IL-12 induction (63). Moreover, TgCyp18 peptidyl-prolyl cis-trans isomerase (PPIase) activity was not required for its interaction with CCR5, but IL-12 induction by TgCyp18 required both CCR5 binding and PPIase enzymatic activities (63). TgCyp18 appears to act as a structural mimic of CCR5-binding ligands, albeit one with no sequence similarity to the known host ligands, macrophage inflammatory protein 1α/chemokine (C-C motif) ligand 3 (CCL3), macrophage inflammatory protein 1β/CCL4, regulated on activation normal T-cell expressed and secreted (RANTES)/CCL5, or monocyte chemotactic protein 2/CCL8, for this receptor (4, 63). There is also evidence that the closely related protozoan Neospora caninum cyclophilin plays a role in stimulating IFN-γ production by bovine peripheral blood mononuclear cells and N. caninum-specific CD4+ T cells (59). This effect is also blocked by cyclosporine (59). IFN-γ production induced by N. caninum tachyzoites is thought to be critical in controlling the acute phase of neosporosis (59).Pressure from the immune response causes tachyzoites to differentiate into slowly multiplying bradyzoites, which form cysts within muscle and brain cells (19). The cysts are protected from the host immune response and establish a life-long chronic infection (18). If the tissue cysts are ingested, for example, through the consumption of undercooked meat, bradyzoites are released into the gut, invade epithelial cells, and differentiate into tachyzoites, initiating a new asexual cycle (10). In vitro models of tachyzoite-to-bradyzoite differentiation have been established by using a variety of stress conditions that mimic the stresses of the host immune response. These conditions include treatment with IFN-γ (5), mitochondrial inhibitors (6), alkaline pH (pH 8.1) (53), and high temperature (54). The stress response is controlled in part by eukaryotic initiation factor 2 kinase in the parasite, which is well characterized as a stress response in eukaryotic cells (55), and differentiation also involves parasite-derived cyclic nucleotide kinases (20). Large-scale sequencing of stage-specific cDNA (36, 39), microarray studies (10), and serial analysis of gene expression tags (44) revealed that stage conversion involves changes in the levels of expression of a large number of genes, although the regulatory mechanism(s) involved in the conversion is poorly understood.Although previous reports mentioned the ability of TgCyp18 to induce the production of the IL-12 (1, 2, 14, 63), the productions of other cytokines were not elucidated. Here we show that TgCyp18 induces the production of other cytokines and nitric oxide (NO) and enhances the bradyzoite conversion of T. gondii.  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