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 31 毫秒
1.
The mammalian gastrointestinal microbiota exerts a strong influence on host lipid and cholesterol metabolism. In this study, we have characterized the interplay among diet, gut microbial ecology, and cholesterol metabolism in a hamster model of hypercholesterolemia. Previous work in this model had shown that grain sorghum lipid extract (GSL) included in the diet significantly improved the high-density lipoprotein (HDL)/non-HDL cholesterol equilibrium (T. P. Carr, C. L. Weller, V. L. Schlegel, S. L. Cuppett, D. M. Guderian, Jr., and K. R. Johnson, J. Nutr. 135:2236-2240, 2005). Molecular analysis of the hamsters'' fecal bacterial populations by pyrosequencing of 16S rRNA tags, PCR-denaturing gradient gel electrophoresis, and Bifidobacterium-specific quantitative real-time PCR revealed that the improvements in cholesterol homeostasis induced through feeding the hamsters GSL were strongly associated with alterations of the gut microbiota. Bifidobacteria, which significantly increased in abundance in hamsters fed GSL, showed a strong positive association with HDL plasma cholesterol levels (r = 0.75; P = 0.001). The proportion of members of the family Coriobacteriaceae decreased when the hamsters were fed GSL and showed a high positive association with non-HDL plasma cholesterol levels (r = 0.84; P = 0.0002). These correlations were more significant than those between daily GSL intake and animal metabolic markers, implying that the dietary effects on host cholesterol metabolism are conferred, at least in part, through an effect on the gut microbiota. This study provides evidence that modulation of the gut microbiota-host metabolic interrelationship by dietary intervention has the potential to improve mammalian cholesterol homeostasis, which has relevance for cardiovascular health.The mammalian gut microbiota interacts intimately with its host, affecting both host metabolic and immunological phenotypes with important consequences for health (18, 22, 32). Recent studies have revealed complex linkages between the gut microbiome and host metabolism, with the microbes exerting effects on the energy balance by influencing glucose and lipid metabolism (2, 7, 28). This intimate metabolic relationship is most likely the consequence of a long coevolutionary process that resulted in a mutualistic relationship between the host and its microbial partners (25). However, life in industrialized societies has introduced profound changes into the human environment (e.g., diet, antibiotics, hospital deliveries, hygiene, etc.) that are markedly different from the conditions to which humans have evolved and that are likely to have occurred too abruptly for the human microbiome to adjust. Consequently, aberrations of the gut microbiota induced through lifestyle factors could be relevant to the etiology of several complex human diseases whose occurrence has markedly increased in developed countries. Interestingly, imbalances in the gut microbiota have been reported for obesity, type 1 and 2 diabetes, some allergies, and inflammatory bowel diseases in humans and animal models (7, 24, 43, 45, 48). The connection between gut bacteria and disease suggests an intriguing paradigm on how to view and potentially treat complex diseases. Specific bacterial populations in the intestine could be pharmaceutical targets to maintain or restore metabolic functions (6, 17).Coronary heart disease (CHD) continues to be a major cause of death in developed countries and is another example of a “western disease” that is less common in underdeveloped countries but increases in frequency with adoption of western customs (4). Most risk factors for CHD (obesity, high blood pressure, type 2 diabetes, heredity, high cholesterol, and diet) have been linked to the gut microbiota (7, 17, 20, 30, 45), and gut bacteria have been suggested to play a role in the etiology of cardiovascular disease (16, 33). Cholesterol metabolism is a key factor in susceptibility to CHD, and as early as 1959, it has been shown that germfree rats have higher serum cholesterol concentrations than their conventional counterparts do (12). Several mechanisms have been proposed by which gut bacteria could influence host cholesterol metabolism (13). Bacterial conversions of bile acids (such as the formation of secondary bile acids) are likely to play a role, as they affect enterohepatic circulation, de novo synthesis of bile acids, emulsification, and cholesterol absorption (10, 28, 30). A further mechanism by which gut bacteria might influence cholesterol metabolism is through Fiaf (fasting-induced adipocyte factor), which is selectively suppressed in the intestinal epithelium by the gut microbiota (1, 2). Fiaf is an important regulator of lipid metabolism (e.g., through its inhibition of lipoprotein lipase) and has been shown to increase total cholesterol and high-density lipoprotein (HDL) cholesterol levels when overexpressed in transgenic mice (26).There are several reasons why hamsters are an excellent model for studying the metabolic relationships among diet, cholesterol metabolism, and gut microbiota in relation to health. First, hamsters are omnivorous, and their blood lipid profiles respond to diets in a predictive manner similar to humans (5). Second, unlike mice and rats which lack cholesterol ester transfer protein, hamsters exhibit all of the enzymatic pathways in lipoprotein and bile metabolism that are also present in humans. They exhibit limited hepatic synthesis of cholesterol and bile acids, resulting in more relevant data when extrapolating to humans (23). Third, hamsters develop atherosclerosis in a predictive manner in response to dietary manipulation (31).Using the Golden Syrian hamster model, Carr and coworkers have shown that the hexane-extractable lipid fraction of grain sorghum whole kernels (GSL), when included in the hamsters'' diet, leads to a significant reduction of plasma non-HDL and liver cholesterol levels while increasing HDL cholesterol levels (8). We extended this research and performed a comprehensive molecular characterization of the fecal microbiota of the hamsters by pyrosequencing of 16S rRNA tags, denaturing gradient gel electrophoresis (DGGE), and Bifidobacterium specific quantitative real-time PCR (qRT-PCR) in order to test whether metabolic effects of GSL were associated with specific modifications of the gut microbiota.  相似文献   

2.
Dynamic, multicompartment in vitro gastrointestinal simulators are often used to monitor gut microbial dynamics and activity. These reactors need to harbor a microbial community that is stable upon inoculation, colon region specific, and relevant to in vivo conditions. Together with the reproducibility of the colonization process, these criteria are often overlooked when the modulatory properties from different treatments are compared. We therefore investigated the microbial colonization process in two identical simulators of the human intestinal microbial ecosystem (SHIME), simultaneously inoculated with the same human fecal microbiota with a high-resolution phylogenetic microarray: the human intestinal tract chip (HITChip). Following inoculation of the in vitro colon compartments, microbial community composition reached steady state after 2 weeks, whereas 3 weeks were required to reach functional stability. This dynamic colonization process was reproducible in both SHIME units and resulted in highly diverse microbial communities which were colon region specific, with the proximal regions harboring saccharolytic microbes (e.g., Bacteroides spp. and Eubacterium spp.) and the distal regions harboring mucin-degrading microbes (e.g., Akkermansia spp.). Importantly, the shift from an in vivo to an in vitro environment resulted in an increased Bacteroidetes/Firmicutes ratio, whereas Clostridium cluster IX (propionate producers) was enriched compared to clusters IV and XIVa (butyrate producers). This was supported by proportionally higher in vitro propionate concentrations. In conclusion, high-resolution analysis of in vitro-cultured gut microbiota offers new insight on the microbial colonization process and indicates the importance of digestive parameters that may be crucial in the development of new in vitro models.The human gastrointestinal tract harbors a complex microbial ecosystem with a coding capacity exceeding that of the host genome by a factor of 100 (13). These gut microbes play a determining role in host health by converting otherwise indigestible compounds (14, 19), protecting against gut epithelial cell injury (46), regulating host fat storage (49), and inducing immunity (20, 48). Modulation of the composition and metabolic activity of these microbes to improve host health attracts a lot of attention and is referred to as gastrointestinal resource management (15, 37). Such new strategies are often evaluated during human trials or in vivo studies of animals associated with conventional or human microbiota (50).Despite the physiological relevance, in vivo experimental setups are inherently associated with some drawbacks. First, apart from fecal analyses over time, most in vivo data are derived from endpoint measurements, thereby limiting the dynamic monitoring of the gut microbiota. Second, troublesome sampling of different gut regions makes it difficult to locate the effects of a treatment. For mechanistic reasons, a third drawback of an in vivo approach is the inability to focus solely on gut microbial activity, because there is always a host involved. For these reasons, different types of in vitro systems have been developed, ranging from simple nonstirred batch cultures without pH control (44) to more complex continuous models involving pH-controlled single (55) or dynamic multicompartment (2, 29, 32, 34) culture systems. Other advantages are the lack of ethical constraints and a higher reproducibility due to strict control of environmental factors that can influence the microbiota, such as retention time, pH, temperature, and food intake. Therefore, in vitro methods are widely used to elucidate the mechanism behind the degradation of prebiotics (17, 52), bioactivation of polyphenols (10, 36, 38), adhesion of microbes to mucins (51), or bioavailability of environmental contaminants (53, 54).Dynamic in vitro gut models need to fulfil certain criteria before they can be used to monitor the modulating potency of specific treatments toward the microbiota. To ensure that effects are due solely to the treatment and not to the adaptation of microbes to the in vitro environment, steady-state conditions in terms of microbial community composition and metabolic activity need to be established prior to the actual start of the experiment (39). Moreover, the stabilization of this in vitro microbiota needs to be reproducible, as comparison of different treatments requires identical starting communities. Former studies assumed but never fully substantiated this requirement (17, 38). Further, in vitro microbiota need to be gut region specific, be representative for the in vivo situation, and maintain a high diversity. The potency of in vitro models thus relies on a good characterization of its microbiota. Molecular techniques, such as denaturing gradient gel electrophoresis (DGGE) (17, 39, 52), fluorescent in situ hybridization (FISH) (6), and quantitative real-time PCR (Q-PCR) (17, 29), provide useful information but do not provide direct phylogenetic information or target only a limited group of previously identified organisms, therefore limiting current knowledge. Recently, high-resolution techniques, such as microarrays (41, 42) and pyrosequencing (59), have provided access to phylogenetic and metagenomic analysis of the gut microbiota in unprecedented detail.In this study, we performed conventional metabolic analysis, applied existing molecular techniques (DGGE), and for the first time provided an in-depth phylogenetic analysis on the simulator of the human intestinal microbial ecosystem (SHIME) in vitro microbiota using the recently developed human intestinal tract chip (HITChip) microarray (41, 42). We evaluated the microbial colonization process in two parallel in vitro simulators (Twin-SHIME) simultaneously inoculated with the same human fecal microbiota. The aims of this study were (i) to determine when the microbial community composition and metabolic activity reach steady-state conditions, (ii) to assess the reproducibility of the stabilization process in two identical in vitro simulators, (iii) to obtain a high-resolution characterization of the colon region specificity of the residing communities, and (iv) to evaluate how the in vivo fecal inoculum changes to the in vitro colon microbial communities.  相似文献   

3.
4.
Gut microbiota carry out key functions in health and participate in the pathogenesis of a growing number of diseases. The aim of this study was to develop a custom microarray that is able to identify hundreds of intestinal bacterial species. We used the Entrez nucleotide database to compile a data set of bacterial 16S rRNA gene sequences isolated from human intestinal and fecal samples. Identified sequences were clustered into separate phylospecies groups. Representative sequences from each phylospecies were used to develop a microbiota microarray based on the Affymetrix GeneChip platform. The designed microbiota array contains probes to 775 different bacterial phylospecies. In our validation experiments, the array correctly identified genomic DNA from all 15 bacterial species used. Microbiota array has a detection sensitivity of at least 1 pg of genomic DNA and can detect bacteria present at a 0.00025% level of overall sample. Using the developed microarray, fecal samples from two healthy children and two healthy adults were analyzed for bacterial presence. Between 227 and 232 species were detected in fecal samples from children, whereas 191 to 208 species were found in adult stools. The majority of identified phylospecies belonged to the classes Clostridia and Bacteroidetes. The microarray revealed putative differences between the gut microbiota of healthy children and adults: fecal samples from adults had more Clostridia and less Bacteroidetes and Proteobacteria than those from children. A number of other putative differences were found at the genus level.In the healthy adult, there are 1011 to 1014 bacteria colonizing the intestine. This outnumbers the total tissue cells in the body by at least an order of magnitude. The composition and activity of this complex microbial system (called microbiota or microflora) have a major influence on health and disease (9). Commensal microbiota contribute to the trophic functions of the gut (producing fermentation products and vitamins that can be used by intestinal epithelial cells), stimulate the immune function of the gastrointestinal tract, transform or excrete toxic substances, protect the host against invasion by pathogenic species, and modulate gut motility (28, 36). At the same time, recent research incriminates a dysfunctional cross-talk between the host and the microbiota in the pathogenesis of a growing number of disorders, such as irritable bowel syndrome, inflammatory bowel disease, allergic diseases, and gastrointestinal cancer (28).While the intestine in a newborn contains no microbes, immediately after birth the intestine of the infant is colonized by enterobacteria and enterococci. Gradual changes in microbiota composition occur during childhood, with a general reduction in the number of aerobes and facultative anaerobes and an increase in the populations of obligate anaerobic species (27). It is considered that by 2 years of age the microbiota resembles that of an adult, which is dominated in health and disease by species from only four phyla, Firmicutes (predominantly Clostridia; 50 to 70% total bacterial numbers), Bacteroidetes (10 to 30%), Proteobacteria (up to 10%), and Actinobacteria (up to 5%), with 90% believed to be obligate anaerobes (4, 10, 11, 22).Traditionally, microorganisms were detected in intestinal samples and feces by microscopic, biochemical, or physiological methods, or by culturing on selective nutrient media. However, since most intestinal microbiota species are obligate anaerobes, their isolation and culturing are difficult (21, 38, 42). In recent years, new methods based on the use of microarray technology have been utilized for the characterization of complex microbial communities (18, 32, 41, 43, 47). Microarrays represent an excellent choice for the high-throughput analysis of bacterial populations, because many different probes can be placed on one slide or synthesized on one chip, and samples thus can be tested for the presence of many different species simultaneously. Environmental and clinical samples can be interrogated directly, circumventing any need for culturing, and thus nonculturable species can be reliably detected.Several types of microarrays have been used to date to characterize the composition of microbial communities (47). Community genome arrays are constructed using whole genomic DNA (gDNA) isolated from pure culture strains (46). Functional gene arrays contain genes encoding key enzymes that are involved in various biochemical processes, and they are useful for monitoring physiological changes in microbial communities (14, 45). Phylogenetic oligonucleotide arrays contain probes derived from rRNA sequence information and are ideally suited for the analysis of microbial community composition structure and variance. Different types of phylogenetic arrays have been designed for these purposes (26, 30, 31).A number of projects performed in the last several years focused on sampling the diversity of human microbiota by the cloning and subsequent sequencing of the 16S rRNA genes isolated from gastrointestinal and fecal samples (5, 10, 13, 23, 38). In this project, we have designed, developed, and validated a custom microbiota microarray containing 16S rRNA genes probes to 775 different microbial phylospecies of human intestinal bacteria. We also have tested the applicability of this array to profiling the microbiota populations in fecal samples isolated from two adult and two child volunteers.  相似文献   

5.
The intestinal flora of mammals contains lactic acid bacteria (LAB) that may provide positive health effects for the host. Such bacteria are referred to as probiotic bacteria. From a pig, we have isolated a Lactobacillus reuteri strain that produces an antimicrobial peptide (AMP). The peptide was purified and characterized, and it was unequivocally shown that the AMP was a well-defined degradation product obtained from the mucus adhesion-promoting protein (MapA); it was therefore termed AP48-MapA. This finding demonstrates how large proteins might inherit unexpected pleiotropic functions by conferring antimicrobial capacities on the producer. The MapA/AP48-MapA system is the first example where a large protein of an intestinal LAB is shown to give rise to such an AMP. It is also of particular interest that the protein that provides this AMP is associated with the binding of the bacterium producing it to the surface/lining of the gut. This finding gives us new perspective on how some probiotic bacteria may successfully compete in this environment and thereby contribute to a healthy microbiota.Mammals have a microbiota in their digestive tract that contains lactic acid bacteria (LAB). It has been increasingly evident that some of these lactic acid bacteria produce antimicrobial peptides that may contribute to the positive effect on their host. Such bacteria are often referred to as probiotics, and one of their important beneficial effects is their ability to produce antimicrobial compounds that prevent or interfere with the growth of pathogenic bacteria in the host.It is known that the fecal microflora of pigs/piglets is large and diverse and develops rapidly after birth. Lactobacillus reuteri is among the very first lactic acid bacteria that colonize the intestine of new-born piglets, and their numbers gradually increase until they become the most dominant LAB in pigs (5, 17, 28). Other lactobacilli that are also part of the gut microbiota of pigs include L. amylovorus, L. acidophilus, L. salivarius, and L. casei (4, 8). Probiotic isolates have been identified within all these species, and many of them are today used as food/feed supplements to support good health (4, 11, 27). An important part of the antimicrobial arsenal produced by lactic acid bacteria (LAB) is a group of peptides called bacteriocins, which are ribosomally synthesized antibiotic-like peptides (antimicrobial peptides [AMPs]) (3, 7, 19). The bacteriocins constitute a wide range of structurally different peptides that are divided into different classes and subclasses. Some are modified (the lantibiotics, or class I), while others are basically unmodified (class II) (3, 6, 19).Most bacteriocins are derived from prepeptides, each containing a short leader sequence (14 to 30 amino acids [aa]) which is cleaved off during the secretion of the mature peptide (19). In recent years, a new group of AMPs have been recognized (18); these are different from regular bacteriocins in that they are derived from larger proteins through specific degradations, leading to a defined peptide possessing antimicrobial activity. Such antimicrobial peptides have been known for a long time in mammalian systems. For instance, lactoferrin, a protein in milk, is readily degraded to a specific antimicrobial peptide through heat, acid treatment, or pepsin digestion (14, 24, 26). Defined histone fragments with antimicrobial properties have been isolated from different eukaryotic species (1, 2, 15, 21, 23), and a few antimicrobial peptides derived from larger proteins have been isolated in bacteria, including Helicobacter pylori (22), propionic acid bacteria (9, 10), and Clostridium beijerinckii (13). Such antimicrobial peptides are most likely formed by proteolytic degradation during cell proliferation or death.  相似文献   

6.
7.
Soil substrate membrane systems allow for microcultivation of fastidious soil bacteria as mixed microbial communities. We isolated established microcolonies from these membranes by using fluorescence viability staining and micromanipulation. This approach facilitated the recovery of diverse, novel isolates, including the recalcitrant bacterium Leifsonia xyli, a plant pathogen that has never been isolated outside the host.The majority of bacterial species have never been recovered in the laboratory (1, 14, 19, 24). In the last decade, novel cultivation approaches have successfully been used to recover “unculturables” from a diverse range of divisions (23, 25, 29). Most strategies have targeted marine environments (4, 23, 25, 32), but soil offers the potential for the investigation of vast numbers of undescribed species (20, 29). Rapid advances have been made toward culturing soil bacteria by reformulating and diluting traditional media, extending incubation times, and using alternative gelling agents (8, 21, 29).The soil substrate membrane system (SSMS) is a diffusion chamber approach that uses extracts from the soil of interest as the growth substrate, thereby mimicking the environment under investigation (12). The SSMS enriches for slow-growing oligophiles, a proportion of which are subsequently capable of growing on complex media (23, 25, 27, 30, 32). However, the SSMS results in mixed microbial communities, with the consequent difficulty in isolation of individual microcolonies for further characterization (10).Micromanipulation has been widely used for the isolation of specific cell morphotypes for downstream applications in molecular diagnostics or proteomics (5, 15). This simple technology offers the opportunity to select established microcolonies of a specific morphotype from the SSMS when combined with fluorescence visualization (3, 11). Here, we have combined the SSMS, fluorescence viability staining, and advanced micromanipulation for targeted isolation of viable, microcolony-forming soil bacteria.  相似文献   

8.
9.
10.
The intestinal microbiota of broiler chickens and the microbiota in the litter have been well studied, but the interactions between these two microbiotas remain to be determined. Therefore, we examined their reciprocal effects by analyzing the intestinal microbiotas of broilers reared on fresh pine shavings versus reused litter, as well as the litter microbiota over a 6-week cycle. Composite ileal mucosal and cecal luminal samples from birds (n = 10) reared with both litter conditions (fresh versus reused) were collected at 7, 14, 21, and 42 days of age. Litter samples were also collected at days 7, 14, 21, and 42. The microbiotas were profiled and compared within sample types based on litter condition using PCR and denaturing gradient gel electrophoresis (PCR-DGGE). The microbiotas were further analyzed using 16S rRNA gene clone libraries constructed from microbiota DNA extracted from both chick intestinal and litter samples collected at day 7. Results showed significant reciprocal effects between the microbiotas present in the litter and those in the intestines of broilers. Fresh litter had more environmental bacteria, while reused litter contained more bacteria of intestinal origin. Lactobacillus spp. dominated the ileal mucosal microbiota of fresh-litter chicks, while a group of bacteria yet to be classified within Clostridiales dominated in the ileal mucosal microbiota in the reused-litter chicks. The Litter condition (fresh versus reused) seemed to have a more profound impact on the ileal microbiota than on the cecal microbiota. The data suggest that the influence of fresh litter on ileal microbiota decreased as broilers grew, compared with temporal changes observed under reused-litter rearing conditions.The intestines of broiler chickens harbor a complex microbiota that plays an important role in the growth and health of the bird. Nurmi and Rantala (27) provided the first reported evidence that a healthy gut microbiota could protect broiler chicks against a challenge by enteric pathogens. That study led to the concept of “competitive exclusion.” Since then, the colonization of the intestines by beneficial bacteria has been shown to promote epithelial cell turnover (34), increase mucous production (26), upregulate expression of genes involved in several important intestinal functions (14), and help with reinforcement of the mucosal barrier, modulation of the immune system, and metabolism of nutrients by the host (46). Although studies conducted using either cultivation-based or molecular biology analyses have documented that the intestinal microbiota of mature broiler chickens is relatively stable (10, 24, 45), this microbiota is still dynamic and can be manipulated to a large extent (8, 31, 37).Commercial broiler flocks in the United States are primarily floor raised in enclosed, environmentally controlled facilities. Within these commercial broiler houses, poultry litter can be reused for a year or longer if managed well and maintained in a relatively dry state (6). At an estimated accumulation rate of 1.45 metric tons per 1,000 broilers, the U.S. poultry industry, which produces >8.5 billion broilers per year (39), produces >12.3 million metric tons of poultry litter annually (4). Management and disposal of such a large quantity of poultry litter are two of the greatest challenges faced by U.S. broiler producers (6). Reuse of poultry litter can help alleviate this challenge, but there is concern that the reused-litter environment, both biotic and abiotic, may negatively affect the intestinal microbiota of the bird, potentially resulting in poor health and reduced production efficiency.Poultry litter is primarily a mixture of bedding materials and bird excreta. In addition to variation in the physiochemical properties, reused poultry litter harbors microbes of typically intestinal origin that are not commonly present in fresh litter (23, 35). During a broiler growth cycle, a constant influx of nutrients and intestinal microbes results in a complex litter microbiota. With continued reuse, the litter environment becomes more complex, which may have a profound impact on flock growth performance and health. Recognizing the importance of the litter environment, especially in commercial broiler houses with high stocking densities, several research groups have investigated how the physiochemical properties of broiler litter affect the growth and health of the birds (15) and the microbiota in the litter, using either cultivation-based or molecular biology techniques (22, 23, 33). Early studies focused on the effect of litter on enteric pathogens, including Salmonella (35, 12, 28) and cellulitis-causing Escherichia coli (38). From analyzing 16S rRNA gene clone libraries, Lu et al. (23) showed that in broiler litter at various stages of reuse, low-G+C Gram-positive bacteria predominated. Recent studies have focused on the interplay between the unique physiochemical conditions within the litter and the endogenous microbiota (22, 29), as well as the occurrence and persistence of antibiotic resistance in poultry litter (13, 17), providing some insight into the litter microbiota and the intestinal microbiota in broiler chickens. However, the relationship between litter and intestinal microbiotas has not been investigated. Since chickens consume some litter materials, we hypothesized that the intestinal and litter microbiotas may affect each other with respect to their composition and diversity. Using PCR with denaturing gradient gel electrophoresis (PCR-DGGE) and 16S rRNA gene clone library analysis, we tested this hypothesis by examining the effect of the litter microbiota on the ontogeny of the intestinal microbiota in broilers over a 6-week growth cycle.  相似文献   

11.
12.
13.
Analysis of Lyme borreliosis (LB) spirochetes, using a novel multilocus sequence analysis scheme, revealed that OspA serotype 4 strains (a rodent-associated ecotype) of Borrelia garinii were sufficiently genetically distinct from bird-associated B. garinii strains to deserve species status. We suggest that OspA serotype 4 strains be raised to species status and named Borrelia bavariensis sp. nov. The rooted phylogenetic trees provide novel insights into the evolutionary history of LB spirochetes.Multilocus sequence typing (MLST) and multilocus sequence analysis (MLSA) have been shown to be powerful and pragmatic molecular methods for typing large numbers of microbial strains for population genetics studies, delineation of species, and assignment of strains to defined bacterial species (4, 13, 27, 40, 44). To date, MLST/MLSA schemes have been applied only to a few vector-borne microbial populations (1, 6, 30, 37, 40, 41, 47).Lyme borreliosis (LB) spirochetes comprise a diverse group of zoonotic bacteria which are transmitted among vertebrate hosts by ixodid (hard) ticks. The most common agents of human LB are Borrelia burgdorferi (sensu stricto), Borrelia afzelii, Borrelia garinii, Borrelia lusitaniae, and Borrelia spielmanii (7, 8, 12, 35). To date, 15 species have been named within the group of LB spirochetes (6, 31, 32, 37, 38, 41). While several of these LB species have been delineated using whole DNA-DNA hybridization (3, 20, 33), most ecological or epidemiological studies have been using single loci (5, 9-11, 29, 34, 36, 38, 42, 51, 53). Although some of these loci have been convenient for species assignment of strains or to address particular epidemiological questions, they may be unsuitable to resolve evolutionary relationships among LB species, because it is not possible to define any outgroup. For example, both the 5S-23S intergenic spacer (5S-23S IGS) and the gene encoding the outer surface protein A (ospA) are present only in LB spirochete genomes (36, 43). The advantage of using appropriate housekeeping genes of LB group spirochetes is that phylogenetic trees can be rooted with sequences of relapsing fever spirochetes. This renders the data amenable to detailed evolutionary studies of LB spirochetes.LB group spirochetes differ remarkably in their patterns and levels of host association, which are likely to affect their population structures (22, 24, 46, 48). Of the three main Eurasian Borrelia species, B. afzelii is adapted to rodents, whereas B. valaisiana and most strains of B. garinii are maintained by birds (12, 15, 16, 23, 26, 45). However, B. garinii OspA serotype 4 strains in Europe have been shown to be transmitted by rodents (17, 18) and, therefore, constitute a distinct ecotype within B. garinii. These strains have also been associated with high pathogenicity in humans, and their finer-scale geographical distribution seems highly focal (10, 34, 52, 53).In this study, we analyzed the intra- and interspecific phylogenetic relationships of B. burgdorferi, B. afzelii, B. garinii, B. valaisiana, B. lusitaniae, B. bissettii, and B. spielmanii by means of a novel MLSA scheme based on chromosomal housekeeping genes (30, 48).  相似文献   

14.
Human immunodeficiency virus type 1 (HIV-1) infects target cells by binding to CD4 and a chemokine receptor, most commonly CCR5. CXCR4 is a frequent alternative coreceptor (CoR) in subtype B and D HIV-1 infection, but the importance of many other alternative CoRs remains elusive. We have analyzed HIV-1 envelope (Env) proteins from 66 individuals infected with the major subtypes of HIV-1 to determine if virus entry into highly permissive NP-2 cell lines expressing most known alternative CoRs differed by HIV-1 subtype. We also performed linear regression analysis to determine if virus entry via the major CoR CCR5 correlated with use of any alternative CoR and if this correlation differed by subtype. Virus pseudotyped with subtype B Env showed robust entry via CCR3 that was highly correlated with CCR5 entry efficiency. By contrast, viruses pseudotyped with subtype A and C Env proteins were able to use the recently described alternative CoR FPRL1 more efficiently than CCR3, and use of FPRL1 was correlated with CCR5 entry. Subtype D Env was unable to use either CCR3 or FPRL1 efficiently, a unique pattern of alternative CoR use. These results suggest that each subtype of circulating HIV-1 may be subject to somewhat different selective pressures for Env-mediated entry into target cells and suggest that CCR3 may be used as a surrogate CoR by subtype B while FPRL1 may be used as a surrogate CoR by subtypes A and C. These data may provide insight into development of resistance to CCR5-targeted entry inhibitors and alternative entry pathways for each HIV-1 subtype.Human immunodeficiency virus type 1 (HIV-1) infects target cells by binding first to CD4 and then to a coreceptor (CoR), of which C-C chemokine receptor 5 (CCR5) is the most common (6, 53). CXCR4 is an additional CoR for up to 50% of subtype B and D HIV-1 isolates at very late stages of disease (4, 7, 28, 35). Many other seven-membrane-spanning G-protein-coupled receptors (GPCRs) have been identified as alternative CoRs when expressed on various target cell lines in vitro, including CCR1 (76, 79), CCR2b (24), CCR3 (3, 5, 17, 32, 60), CCR8 (18, 34, 38), GPR1 (27, 65), GPR15/BOB (22), CXCR5 (39), CXCR6/Bonzo/STRL33/TYMSTR (9, 22, 25, 45, 46), APJ (26), CMKLR1/ChemR23 (49, 62), FPLR1 (67, 68), RDC1 (66), and D6 (55). HIV-2 and simian immunodeficiency virus SIVmac isolates more frequently show expanded use of these alternative CoRs than HIV-1 isolates (12, 30, 51, 74), and evidence that alternative CoRs other than CXCR4 mediate infection of primary target cells by HIV-1 isolates is sparse (18, 30, 53, 81). Genetic deficiency in CCR5 expression is highly protective against HIV-1 transmission (21, 36), establishing CCR5 as the primary CoR. The importance of alternative CoRs other than CXCR4 has remained elusive despite many studies (1, 30, 70, 81). Expansion of CoR use from CCR5 to include CXCR4 is frequently associated with the ability to use additional alternative CoRs for viral entry (8, 16, 20, 63, 79) in most but not all studies (29, 33, 40, 77, 78). This finding suggests that the sequence changes in HIV-1 env required for use of CXCR4 as an additional or alternative CoR (14, 15, 31, 37, 41, 57) are likely to increase the potential to use other alternative CoRs.We have used the highly permissive NP-2/CD4 human glioma cell line developed by Soda et al. (69) to classify virus entry via the alternative CoRs CCR1, CCR3, CCR8, GPR1, CXCR6, APJ, CMKLR1/ChemR23, FPRL1, and CXCR4. Full-length molecular clones of 66 env genes from most prevalent HIV-1 subtypes were used to generate infectious virus pseudotypes expressing a luciferase reporter construct (19, 57). Two types of analysis were performed: the level of virus entry mediated by each alternative CoR and linear regression of entry mediated by CCR5 versus all other alternative CoRs. We thus were able to identify patterns of alternative CoR use that were subtype specific and to determine if use of any alternative CoR was correlated or independent of CCR5-mediated entry. The results obtained have implications for the evolution of env function, and the analyses revealed important differences between subtype B Env function and all other HIV-1 subtypes.  相似文献   

15.
Immunogold localization revealed that OmcS, a cytochrome that is required for Fe(III) oxide reduction by Geobacter sulfurreducens, was localized along the pili. The apparent spacing between OmcS molecules suggests that OmcS facilitates electron transfer from pili to Fe(III) oxides rather than promoting electron conduction along the length of the pili.There are multiple competing/complementary models for extracellular electron transfer in Fe(III)- and electrode-reducing microorganisms (8, 18, 20, 44). Which mechanisms prevail in different microorganisms or environmental conditions may greatly influence which microorganisms compete most successfully in sedimentary environments or on the surfaces of electrodes and can impact practical decisions on the best strategies to promote Fe(III) reduction for bioremediation applications (18, 19) or to enhance the power output of microbial fuel cells (18, 21).The three most commonly considered mechanisms for electron transfer to extracellular electron acceptors are (i) direct contact between redox-active proteins on the outer surfaces of the cells and the electron acceptor, (ii) electron transfer via soluble electron shuttling molecules, and (iii) the conduction of electrons along pili or other filamentous structures. Evidence for the first mechanism includes the necessity for direct cell-Fe(III) oxide contact in Geobacter species (34) and the finding that intensively studied Fe(III)- and electrode-reducing microorganisms, such as Geobacter sulfurreducens and Shewanella oneidensis MR-1, display redox-active proteins on their outer cell surfaces that could have access to extracellular electron acceptors (1, 2, 12, 15, 27, 28, 31-33). Deletion of the genes for these proteins often inhibits Fe(III) reduction (1, 4, 7, 15, 17, 28, 40) and electron transfer to electrodes (5, 7, 11, 33). In some instances, these proteins have been purified and shown to have the capacity to reduce Fe(III) and other potential electron acceptors in vitro (10, 13, 29, 38, 42, 43, 48, 49).Evidence for the second mechanism includes the ability of some microorganisms to reduce Fe(III) that they cannot directly contact, which can be associated with the accumulation of soluble substances that can promote electron shuttling (17, 22, 26, 35, 36, 47). In microbial fuel cell studies, an abundance of planktonic cells and/or the loss of current-producing capacity when the medium is replaced is consistent with the presence of an electron shuttle (3, 14, 26). Furthermore, a soluble electron shuttle is the most likely explanation for the electrochemical signatures of some microorganisms growing on an electrode surface (26, 46).Evidence for the third mechanism is more circumstantial (19). Filaments that have conductive properties have been identified in Shewanella (7) and Geobacter (41) species. To date, conductance has been measured only across the diameter of the filaments, not along the length. The evidence that the conductive filaments were involved in extracellular electron transfer in Shewanella was the finding that deletion of the genes for the c-type cytochromes OmcA and MtrC, which are necessary for extracellular electron transfer, resulted in nonconductive filaments, suggesting that the cytochromes were associated with the filaments (7). However, subsequent studies specifically designed to localize these cytochromes revealed that, although the cytochromes were extracellular, they were attached to the cells or in the exopolymeric matrix and not aligned along the pili (24, 25, 30, 40, 43). Subsequent reviews of electron transfer to Fe(III) in Shewanella oneidensis (44, 45) appear to have dropped the nanowire concept and focused on the first and second mechanisms.Geobacter sulfurreducens has a number of c-type cytochromes (15, 28) and multicopper proteins (12, 27) that have been demonstrated or proposed to be on the outer cell surface and are essential for extracellular electron transfer. Immunolocalization and proteolysis studies demonstrated that the cytochrome OmcB, which is essential for optimal Fe(III) reduction (15) and highly expressed during growth on electrodes (33), is embedded in the outer membrane (39), whereas the multicopper protein OmpB, which is also required for Fe(III) oxide reduction (27), is exposed on the outer cell surface (39).OmcS is one of the most abundant cytochromes that can readily be sheared from the outer surfaces of G. sulfurreducens cells (28). It is essential for the reduction of Fe(III) oxide (28) and for electron transfer to electrodes under some conditions (11). Therefore, the localization of this important protein was further investigated.  相似文献   

16.
17.
Human cytomegalovirus (HCMV) UL37 proteins traffic sequentially from the endoplasmic reticulum (ER) to the mitochondria. In transiently transfected cells, UL37 proteins traffic into the mitochondrion-associated membranes (MAM), the site of contact between the ER and mitochondria. In HCMV-infected cells, the predominant UL37 exon 1 protein, pUL37x1, trafficked into the ER, the MAM, and the mitochondria. Surprisingly, a component of the MAM calcium signaling junction complex, cytosolic Grp75, was increasingly enriched in heavy MAM from HCMV-infected cells. These studies show the first documented case of a herpesvirus protein, HCMV pUL37x1, trafficking into the MAM during permissive infection and HCMV-induced alteration of the MAM protein composition.The human cytomegalovirus (HCMV) UL37 immediate early (IE) locus expresses multiple products, including the predominant UL37 exon 1 protein, pUL37x1, also known as viral mitochondrion-localized inhibitor of apoptosis (vMIA), during lytic infection (16, 22, 24, 39, 44). The UL37 glycoprotein (gpUL37) shares UL37x1 sequences and is internally cleaved, generating pUL37NH2 and gpUL37COOH (2, 22, 25, 26). pUL37x1 is essential for the growth of HCMV in humans (17) and for the growth of primary HCMV strains (20) and strain AD169 (14, 35, 39, 49) but not strain TownevarATCC in permissive human fibroblasts (HFFs) (27).pUL37x1 induces calcium (Ca2+) efflux from the endoplasmic reticulum (ER) (39), regulates viral early gene expression (5, 10), disrupts F-actin (34, 39), recruits and inactivates Bax at the mitochondrial outer membrane (MOM) (4, 31-33), and inhibits mitochondrial serine protease at late times of infection (28).Intriguingly, HCMV UL37 proteins localize dually in the ER and in the mitochondria (2, 9, 16, 17, 24-26). In contrast to other characterized, similarly localized proteins (3, 6, 11, 23, 30, 38), dual-trafficking UL37 proteins are noncompetitive and sequential, as an uncleaved gpUL37 mutant protein is ER translocated, N-glycosylated, and then imported into the mitochondria (24, 26).Ninety-nine percent of ∼1,000 mitochondrial proteins are synthesized in the cytosol and directly imported into the mitochondria (13). However, the mitochondrial import of ER-synthesized proteins is poorly understood. One potential pathway is the use of the mitochondrion-associated membrane (MAM) as a transfer waypoint. The MAM is a specialized ER subdomain enriched in lipid-synthetic enzymes, lipid-associated proteins, such as sigma-1 receptor, and chaperones (18, 45). The MAM, the site of contact between the ER and the mitochondria, permits the translocation of membrane-bound lipids, including ceramide, between the two organelles (40). The MAM also provides enriched Ca2+ microdomains for mitochondrial signaling (15, 36, 37, 43, 48). One macromolecular MAM complex involved in efficient ER-to-mitochondrion Ca2+ transfer is comprised of ER-bound inositol 1,4,5-triphosphate receptor 3 (IP3R3), cytosolic Grp75, and a MOM-localized voltage-dependent anion channel (VDAC) (42). Another MAM-stabilizing protein complex utilizes mitofusin 2 (Mfn2) to tether ER and mitochondrial organelles together (12).HCMV UL37 proteins traffic into the MAM of transiently transfected HFFs and HeLa cells, directed by their NH2-terminal leaders (8, 47). To determine whether the MAM is targeted by UL37 proteins during infection, we fractionated HCMV-infected cells and examined pUL37x1 trafficking in microsomes, mitochondria, and the MAM throughout all temporal phases of infection. Because MAM domains physically bridge two organelles, multiple markers were employed to verify the purity and identity of the fractions (7, 8, 19, 46, 47).(These studies were performed in part by Chad Williamson in partial fulfillment of his doctoral studies in the Biochemistry and Molecular Genetics Program at George Washington Institute of Biomedical Sciences.)HFFs and life-extended (LE)-HFFs were grown and not infected or infected with HCMV (strain AD169) at a multiplicity of 3 PFU/cell as previously described (8, 26, 47). Heavy (6,300 × g) and light (100,000 × g) MAM fractions, mitochondria, and microsomes were isolated at various times of infection and quantified as described previously (7, 8, 47). Ten- or 20-μg amounts of total lysate or of subcellular fractions were resolved by SDS-PAGE in 4 to 12% Bis-Tris NuPage gels (Invitrogen) and examined by Western analyses (7, 8, 26). Twenty-microgram amounts of the fractions were not treated or treated with proteinase K (3 μg) for 20 min on ice, resolved by SDS-PAGE, and probed by Western analysis. The blots were probed with rabbit anti-UL37x1 antiserum (DC35), goat anti-dolichyl phosphate mannose synthase 1 (DPM1), goat anti-COX2 (both from Santa Cruz Biotechnology), mouse anti-Grp75 (StressGen Biotechnologies), and the corresponding horseradish peroxidase-conjugated secondary antibodies (8, 47). Reactive proteins were detected by enhanced chemiluminescence (ECL) reagents (Pierce), and images were digitized as described previously (26, 47).  相似文献   

18.
This study examined bacteria-immune interactions in a mouse model possessing microbiota-dependent immune regulatory features similar to those occurring in human atopy, colitis, and immune regulation. Associations between the abundance of several bacterial phylotypes and immunoregulatory target cell types were identified, suggesting that they may play a role in these phenotypes.Bacteria are involved in critical aspects of immune system development and regulation (5, 23, 26, 29). Mice raised under germfree conditions exhibit a variety of abnormalities, including hypoplastic Peyer''s patches, reduced numbers of IgA-producing cells, relatively unstructured spleen and lymph nodes, and hypogammaglobulinemic serum (23). Remarkably, after several weeks of exposure to standard intestinal microbiota, normal immune structure and function are restored. Mechanistic details underlying microbe-immune interactions have been recently elucidated for two common intestinal bacteria. Bacteroides thetaiotaomicron was shown to induce the angiogenin Ang4, a component of innate immunity possessing microbicidal activity against a wide range of intestinal microbes, including both bacterial and fungal pathogens (16). In addition, studies of the Bacteroides fragilis zwitterionic capsular polysaccharide A have established it as a cognate antigen of certain CD4+ T cells, which programs immune effector polarization (24) and protection of mice from infection by Helicobacter hepaticus through several immune-mediated mechanisms (25). Resident microbiota also modify the interaction of dendritic cells with regulatory T-cell populations, with resultant susceptibility to chronic inflammatory disease, like colitis (15, 28).Recent work by Braun and colleagues has characterized a mouse model with unique immunologic features linking resident microbiota with levels of regulatory CD8+ T cells (13, 17, 39). This model is comprised of two physically isolated colonies of isogenic mice harboring distinct microbial communities: conventional floras (CF) and restricted floras (RF). CF refers to C57BL/6 mice housed in a standard specific-pathogen-free facility, while RF refers to C57BL/6 mice containing a different complement of intestinal microorganisms (13, 30), originally created by transferring several nonpathogenic anaerobic bacteria into antibiotic-treated mice (13). RF mice differ from CF mice in several immunologic phenotypes, including selective reduction of marginal zone (MZ) B cells (39), plasmacytoid dendritic cells (pDC) (13), and invariant natural killer (iNK) T cells (38a), as well as naïve CD4+ and CD8+ T cells (17). In addition, RF mice were shown to be resistant to colitis under genetic or adoptive transfer conditions that permit disease activity in CF mice (2). RF mice also cleared experimental infections by Campylobacter jejuni more slowly than did their CF counterparts (6). The resulting concept is that certain resident microbiota, which may be more abundant in RF mice than in CF mice, induce invariant Qa-1 T cells, with resultant changes in host immunoregulation and microbial surveillance (2).An important issue raised by the foregoing observations is the identity of resident microbiota responsible for this host immunoregulatory response. The objective of this study was to develop a methodology, based on bacteria-immune interactions in the RF/CF mouse model, to identify candidate microbiota. In this study, we employed a series of experiments examining associations between the population densities of bacterial rRNA genes and several immunologic features that differ between CF and RF mice.  相似文献   

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

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