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 62 毫秒
1.
Par-1 is an evolutionarily conserved protein kinase required for polarity in worms, flies, frogs, and mammals. The mammalian Par-1 family consists of four members. Knockout studies of mice implicate Par-1b/MARK2/EMK in regulating fertility, immune homeostasis, learning, and memory as well as adiposity, insulin hypersensitivity, and glucose metabolism. Here, we report phenotypes of mice null for a second family member (Par-1a/MARK3/C-TAK1) that exhibit increased energy expenditure, reduced adiposity with unaltered glucose handling, and normal insulin sensitivity. Knockout mice were protected against high-fat diet-induced obesity and displayed attenuated weight gain, complete resistance to hepatic steatosis, and improved glucose handling with decreased insulin secretion. Overnight starvation led to complete hepatic glycogen depletion, associated hypoketotic hypoglycemia, increased hepatocellular autophagy, and increased glycogen synthase levels in Par-1a−/− but not in control or Par-1b−/− mice. The intercrossing of Par-1a−/− with Par-1b−/− mice revealed that at least one of the four alleles is necessary for embryonic survival. The severity of phenotypes followed a rank order, whereby the loss of one Par-1b allele in Par-1a−/− mice conveyed milder phenotypes than the loss of one Par-1a allele in Par-1b−/− mice. Thus, although Par-1a and Par-1b can compensate for one another during embryogenesis, their individual disruption gives rise to distinct metabolic phenotypes in adult mice.Cellular polarity is a fundamental principle in biology (6, 36, 62). The prototypical protein kinase originally identified as a regulator of polarity was termed partitioning defective (Par-1) due to early embryonic defects in Caenorhabditis elegans (52). Subsequent studies revealed that Par-1 is required for cellular polarity in worms, flies, frogs, and mammals (4, 17, 58, 63, 65, 71, 89). An integral role for Par-1 kinases in multiple signaling pathways has also been established, and although not formally addressed, multifunctionality for individual Par-1 family members is implied in reviews of the list of recognized upstream regulators and downstream substrates (Table (Table1).1). Interestingly, for many Par-1 substrates the phosphorylated residues generate 14-3-3 binding sites (25, 28, 37, 50, 59, 61, 68, 69, 78, 95, 101, 103). 14-3-3 binding in turn modulates both nuclear/cytoplasmic as well as cytoplasmic/membrane shuttling of target proteins, thus allowing Par-1 activity to establish intracellular spatial organization (15, 101). The phosphorylation of Par-1 itself promotes 14-3-3 binding, thereby regulating its subcellular localization (37, 59, 101).

TABLE 1.

Multifunctionality of Par-1 polarity kinase pathwaysa
Regulator or substrateFunctionReference(s)
Regulators (upstream function)
    LKB1Wnt signaling, Peutz-Jeghers syndrome, insulin signal transduction, pattern formation2, 63, 93
    TAO1MEK3/p38 stress-responsive mitogen-activated protein kinase (MAPK) pathway46
    MARKKNerve growth factor signaling in neurite development and differentiation98
    aPKCCa2+/DAG-independent signal transduction, cell polarity, glucose metabolism14, 37, 40, 45, 59, 75, 95
    nPKC/PKDDAG-dependent, Ca2+-independent signal transduction (GPCR)101
    PAR-3/PAR-6/aPKC(−); regulates Par-1, assembly of microtubules, axon-dendrite specification19
    GSK3β(−); tau phosphorylation, Alzheimer''s dementia, energy metabolism, body patterning54, 97
    Pim-1 oncogene(−); G2/M checkpoint, effector of cytokine signaling and Jak/STAT(3/5)5
    CaMKI(−); Ca2+-dependent signal transduction, neuronal differentiation99
Substrates (downstream function)
    Cdc25CRegulation of mitotic entry by activation of the cdc2-cyclin B complex25, 72, 78, 103
    Class II HDACControl of gene expression and master regulator of subcellular trafficking28, 50
    CRTC2/TORC2Gluconeogenesis regulator via LKB1/AMPK/TORC2 signaling, PPARγ1a coactivator49
    Dlg/PSD-95Synaptogenesis and neuromuscular junction, tumor suppressor (102)104
    DisheveledWnt signaling, translocation of Dsh from cytoplasmic vesicles to cortex73, 94
    KSR1Regulation of the Ras-MAPK pathway68, 69
    MAP2/4/TAUDynamic instability (67, 83) of microtubules, Alzheimer''s dementia (30)11, 31-33, 47, 70, 96
    Mib/NotchMind bomb (Mib degradation and repression of Notch signaling results in neurogenesis)57, 74, 81
    Par3/OSKAR/LglCytoplasmic protein segregation, cell polarity, and asymmetric cell division7, 10
    Pkp2Desmosome assembly and organization; nuclear shuttling68, 69
    PTPH1Linkage between Ser/Thr and Tyr phosphorylation-dependent signaling103
    Rab11-FIPRegulation of endocytosis (23), trafficking of E-cadherin (64)34
Open in a separate windowaLKB1 also is known as Par-4; MARKK also is known as Ste20-like; (−), inhibitory/negative regulation has been shown; GPCR, G protein-coupled receptors. MARKK is highly homologous to TAO-1 (thousand-and-one amino acid kinase) (46).The mammalian Par-1 family contains four members (Table (Table2).2). Physiological functions of the Par-1b kinase have been studied using targeted gene knockout approaches in mice (9, 44). Two independently derived mouse lines null for Par-1b have implicated this protein kinase in diverse physiological processes, including fertility (9), immune system homeostasis (44), learning and memory (86), the positioning of nuclei in pancreatic beta cells (35, 38), and growth and metabolism (43).

TABLE 2.

Terminology and localization of mammalian Par-1 family members
SynonymsaSubcellular localization
Par-1a, MARK3, C-TAK1, p78/KP78, 1600015G02Rik, A430080F22Rik, Emk2, ETK-1, KIAA4230, mKIAA1860, mKIAA4230, M80359Basolateralb/apicalc
Par-1b, EMK, MARK2, AU024026, mKIAA4207Basolateral
Par1c, MARK1Basolateral
Par1d, MARK4, MARKL1Not asymmetricd
Open in a separate windowaPar should not to be confused with protease-activated receptor 1 (PAR1 [29]); C-TAK1, Cdc twenty-five C-associated kinase 1; MARK, microtubule affinity regulating kinase; MARKL, MAP/microtubule affinity-regulating kinase-like 1.bBasolateral to a lesser degree than Par-1b (37).cHuman KP78 is asymmetrically localized to the apical surface of epithelial cells (76).dVariant that does not show asymmetric localization in epithelial cells when overexpressed (95).Beyond Par-1b, most information regarding the cell biological functions of the Par-1 kinases comes from studies of Par-1a. Specifically, Par-1a has been implicated in pancreatic (76) and hepatocarcinogenesis (51), as well as colorectal tumors (77), hippocampal function (100), CagA (Helicobacter pylori)-associated epithelial cell polarity disruption (82), and Peutz-Jeghers syndrome (48), although the latter association has been excluded recently (27). As a first step toward determining unique and redundant functions of Par-1 family members, mice disrupted for a second member of the family (Par-1a/MARK3/C-TAK1) were generated. We report that Par-1a−/− mice are viable and develop normally, and adult mice are hypermetabolic, have decreased white and brown adipose tissue mass, and unaltered glucose/insulin handling. However, when challenged by a high-fat diet (HFD), Par-1a−/− mice exhibit resistance to hepatic steatosis, resistance to glucose intolerance, and the delayed onset of obesity relative to that of control littermates. Strikingly, overnight starvation results in a complete depletion of glycogen and lipid stores along with an increase in autophagic vacuoles in the liver of Par-1a−/− but not Par-1b−/− mice. Correspondingly, Par-1a−/− mice develop hypoketotic hypoglycemia. These findings reveal unique metabolic functions of two Par-1 family members.  相似文献   

2.
3.
4.
5.
Human bocavirus (HBoV) was recently discovered and classified in the Bocavirus genus (family Parvoviridae, subfamily Parvovirinae) on the basis of genomic similarity to bovine parvovirus and canine minute virus. HBoV has been implicated in respiratory tract infections and gastroenteric disease in children worldwide, yet despite numerous epidemiological reports, there has been limited biochemical and molecular characterization of the virus. Reported here is the three-dimensional structure of recombinant HBoV capsids, assembled from viral protein 2 (VP2), at 7.9-Å resolution as determined by cryo-electron microscopy and image reconstruction. A pseudo-atomic model of HBoV VP2 was derived from sequence alignment analysis and knowledge of the crystal structure of human parvovirus B19 (genus Erythrovirus). Comparison of the HBoV capsid structure to that of parvoviruses from five separate genera demonstrates strong conservation of a β-barrel core domain and an α-helix, from which emanate several loops of various lengths and conformations, yielding a unique surface topology that differs from the three already described for this family. The highly conserved core is consistent with observations for other single-stranded DNA viruses, and variable surface loops have been shown to confer the host-specific tropism and the diverse antigenic properties of this family.Human bocavirus (HBoV), a newly discovered member of the family Parvoviridae, was originally isolated in randomly selected nasopharyngeal aspirates (5). Since this initial discovery, HBoV has also been detected worldwide, predominantly in children under the age of 2 years with respiratory infections, in serum, urine, and fecal samples (40). Symptomatic children commonly exhibit acute diseases of the upper and lower respiratory tracts (7, 36, 44, 56) and, possibly, gastroenteritis (31, 56) though the link to gastroenteritis outbreaks has been questioned (12). It is still unclear if HBoV is the sole etiologic agent of respiratory disease as higher rates of coinfections with other respiratory pathogens such as human rhinovirus and Streptococcus spp. are often observed (4). However, Allander et al. recently reported (4) that HBoV was found in 19% of children with acute wheezing, thereby making it the fourth most common virus, after rhinoviruses, enteroviruses, and respiratory syncytial virus, detected in children exhibiting this symptom. These findings suggest that, at high viral load, HBoV could be an etiologic agent of respiratory tract disease (4). HBoV infection is common in the first few years of life, and clinical research suggests it may follow the primary period for acquisition of human parvovirus B19 (B19) though there is no antigenic cross-reactivity between B19 and HBoV (28, 30). By age 5, most people have circulating antibodies against HBoV, as is also true for other respiratory viruses such as respiratory syncytial virus, rhinoviruses, and human metapneumovirus (17). HBoV has also been identified in adults, with ∼63% of samples tested being seropositive, showing a positive correlation with age and a slight positive bias toward women (14).The Parvoviridae is a family of small, nonenveloped viruses that package a single-stranded DNA (ssDNA) genome of ∼5,000 bases. These viruses are subdivided into two subfamilies: Parvovirinae and Densovirinae (Table (Table1).1). The Parvovirinae are further subdivided into five genera, all of whose members infect vertebrates. The Densovirinae (four genera) infect only invertebrates. Phylogenetic analysis places HBoV in the recently classified Bocavirus genus (Table (Table1).1). In addition to HBoV, numerous parvoviruses circulate among the human population. Among these are the following: several dependoviruses; adeno-associated virus (AAV) serotypes AAV1 to AAV3, AAV5, and AAV9; the Erythrovirus B19; and the newly discovered human parvovirus genotypes 4 (Parv4) and 5 (Parv5) (23, 27, 50). Of these, only B19 had been implicated in disease until the discovery of HBoV and Parv4, which has been isolated from patients who present symptoms of acute HIV infection (50).

TABLE 1.

Selected properties of representative members of the Parvoviridae
Subfamily (host) and genusMember(s)aNo. of VPsbGroupcMajor VP(s)d
Parvovirinae (vertebrate)
    ParvovirusMVM*, CPV*, FPV*3IVP2
    ErythrovirusB19*, SPV2IIIVP2
    DependovirusAAV2*, AAV4*, GPV3IIIVP3
    AmdovirusAMDV2IIIVP2
    BocavirusHBoV, BPV, CnMV2NAVP2
Densovirinae (invertebrate)
    DensovirusGmDNV*, JcDNV4IIVP4
    IteravirusBmDNV4-6IIVP1-VP4
    BrevidensovirusAaeDNV, AalDNV2-3NAVP1 or VP2/3
    PefudensovirusPfDNV5NAVP1
Open in a separate windowaAalDNV, Aedes albopictus densovirus; AaeDNV, Aedes aegypti densovirus; BmDNV, Bombyx mori densovirus; BPV, bovine parvovirus; CnMV, canine minute virus; CPV, canine parvovirus; FPV, feline panleukopenia virus; GPV, goose parvovirus; JcDNV, Junonia coenia densovirus; PfDNV, Periplaneta fuliginosa densovirus; SPV, simian parvovirus. *, structure determined by X-ray crystallography; †, structure determined by cryo-EM.bThe number of VPs in the virion capsid.cGroup refers to the surface topologies described in Results and Discussion (HBoV is currently the only bocavirus with a known structure; there is no structure available for Periplaneta fuliginosa densovirus or the brevidensoviruses). NA, structural group not assigned.dThe VP(s) that comprises most of the wild-type virion.The HBoV genome, like that of all members of the Bocavirus genus, contains three open reading frames (ORFs). The first ORF, at the 5′ end, encodes NS1, a nonstructural protein. The next ORF, unique to the bocaviruses, encodes NP1, a second nonstructural protein. The third ORF, at the 3′ end, encodes the two structural capsid viral proteins (VPs), VP1 and VP2. The HBoV VPs share 42% and 43% amino acid sequence identity with the corresponding VPs of bovine parvovirus and canine minute virus, respectively (5). More recently, two additional HBoV-like viruses, HBoV-2 and HBoV-3, were identified in stool samples from children (8, 31). The genome organization of these viruses is identical to that of HBoV, with the NS1, NP1, and VP proteins of HBoV-2 and HBoV-3 being, respectively, ∼80 and 90%, ∼70 and 80%, and ∼80 and 80% identical to the respective proteins in HBoV (8, 31).Parvovirus genomes are packaged into a T=1 icosahedral capsid that is assembled from 60 copies of a combination of up to six types of capsid VPs (VP1 to VP6), all of which share a C terminus. VP1 is always a minor component, typically comprising about five copies per capsid, whereas the smallest VP is always the major component. The unique N-terminal region of VP1 (VP1u) contains a conserved phospholipase A2 (PLA2) motif within the first 131 amino acids that is essential for infection (49, 61). Interestingly, Aleutian mink disease virus (AMDV), the only member of the Amdovirus genus, is the only exception in that this motif is absent, which suggests that this virus employs a different mechanism to escape the endosome during infection (54).The X-ray crystal structures of several parvoviruses show that all VPs contain a conserved, eight-stranded β-barrel motif (βB to βI) that forms the core of the capsid (15). There is also a conserved α-helix (αA) observed in all parvovirus structures determined to date. The bulk of the VP consists of elaborate loops between the strands that form the surface of the capsid. For example, the GH loop between the βG and βH strands is ∼230 residues. The composition and topology of these loops encode several important functions, including tissue tropism, pathogenicity, and the antigenic response directed against each parvovirus during infection (2).A number of parvoviruses have been studied by cryo-electron microscopy (cryo-EM) and three-dimensional (3D) image reconstruction in concert with and complementary to X-ray crystallographic studies (reviewed in reference 15). Reported here is the 3D structure of a recombinant HBoV capsid solved to 7.9-Å resolution using cryo-EM. The capsid of HBoV was compared to that of representative members of the Parvoviridae (Table (Table1)1) with known atomic structures (AAV2, minute virus of mice [MVM], B19, and Galleria mellonella densovirus [GmDNV]) or pseudo-atomic models built into cryo-EM reconstructed density (AMDV) to identify similarities and differences. The capsid topology of the newly emerging HBoV incorporates a combination of surface structural features seen in other members of the Parvovirinae and is closest to that of B19, the only other structurally characterized parvovirus that is pathogenic to humans. A pseudo-atomic model of the HBoV VP2, built into the reconstructed density, identified conserved core secondary structure elements, which are known to be important for parvovirus capsid assembly, and variable surface loops, which likely govern host specific interactions.  相似文献   

6.
7.
The human gut microbe Bacteroides fragilis can alter the expression of its surface molecules, such as capsular polysaccharides and SusC/SusD family outer membrane proteins, through reversible DNA inversions. We demonstrate here that DNA inversions at 12 invertible regions, including three gene clusters for SusC/SusD family proteins, were controlled by a single tyrosine site-specific recombinase (Tsr0667) encoded by BF0667 in B. fragilis strain YCH46. Genetic disruption of BF0667 diminished or attenuated shufflon-type DNA inversions at all three susC/susD genes clusters, as well as simple DNA inversions at nine other loci, most of which colocalized with susC/susD family genes. The inverted repeat sequences found within the Tsr0667-regulated invertible regions shared the consensus motif sequence AGTYYYN4GDACT. Tsr0667 specifically mediated the DNA inversions of 10 of the 12 regions, even under an Escherichia coli background when the invertible regions were exposed to BF0667 in E. coli cells. Thus, Tsr0667 is an additional globally acting DNA invertase in B. fragilis, which probably involves the selective expression of SusC/SusD family outer membrane proteins.The human gut harbors an abundant and diverse microbiota. Bacteroides is one of the most abundant genera of human gut microflora (10, 17, 20), and the biological activities of Bacteroides species are deeply integrated into human physiology through nutrient degradation, the production of short-chain fatty acids, or immunomodulatory molecules (11-14, 24). Recent genomic analyses of Bacteroides have revealed that the bacteria possess redundant abilities not only to bind and degrade otherwise indigestible dietary polysaccharides but also to produce vast arrays of capsular polysaccharide (5, 19, 38, 39). These functional redundancies have been established by the extensive duplication of various genes that encode molecules such as glycosylhydrolases, glycosyltransferases, and outer membrane proteins of the SusC/SusD family (starch utilization system) known to be involved in polysaccharide recognition and transport (7, 27, 28, 30). It has been assumed that these functional redundancies of Bacteroides contribute to the stability of the gut ecosystem (3, 21, 23, 32, 39).Another characteristic feature common in Bacteroides species is that the expression of some of the genes is altered in an on-off manner by reversible DNA inversions at gene promoters or within the protein-coding regions (5, 9, 19, 38, 39). These phase-variable phenotypes are associated with surface architectures such as capsular polysaccharides and SusC/SusD family proteins (5, 6, 16, 19). Our previous genomic analyses of Bacteroides fragilis strain YCH46 revealed that it contained as many as 31 invertible regions in its chromosome (19). These invertible regions can be grouped into six classes according to the internal motif sequences within inverted repeat sequences (IRs) (Table (Table1).1). The DNA inversions within these regions are thought to be controlled by site-specific DNA invertases specific to each class. B. fragilis strain YCH46 contains 33 tyrosine site-specific recombinases (Tsr) genes and four serine site-specific recombinases (Ssr) genes. Generally, DNA invertases mediate DNA inversions at adjacent regions, such as FimB and FimE, that flip their immediate downstream promoters to generate a phase-variable phenotype of type I pili in Escherichia coli (15). B. fragilis is unique in that this anaerobe possesses not only locally acting DNA invertases but also globally acting DNA invertases that mediate DNA inversions at distant loci (8, 29). It has been reported that B. fragilis possesses at least two types of master DNA invertase that regulate DNA inversions at multiple loci simultaneously (8, 29). One is Mpi, an Ssr that mediates the on-off switching of 13 promoter regions (corresponding to class I regions in B. fragilis strain YCH46), including seven promoter regions for capsular polysaccharide biosynthesis in B. fragilis strain NCTC9343 (8). The other master DNA invertase is Tsr19, a Tsr that regulates DNA inversions at two distantly located promoter regions (corresponding to class IV regions in B. fragilis strain YCH46) associated with the large encapsulation phenotype (6, 26, 29). The invertible regions contain specific consensus motifs within the IRs corresponding to each DNA invertase and constitute a regulatory unit. We designated this type of regulatory unit as an “inverlon,” which consists of at least two invertible regions controlled by a single master DNA invertase.

TABLE 1.

Classification of the invertible regions in B. fragilis strain YCH46 based on internal motif sequences within IRs
ClassaNo.Consensus motif sequencesbMaster DNA invertase genecRegulated genesSource or reference(s)
I14ARACGTWCGTBF2765 (mpi)Capsular polysaccharide biosynthesis genes8
II10AGTTC{N5}GAACTBF0667susC/susD paralogsThis study
III3GTTAC{N7}GTAACBF3038, BF4033, BF4283Putative outer membrane protein genes36
IV2TACTTANTAGGTAANAGAABF2766Extracellular polysacharide biosynthesis genes6, 26, 29
V1TCTGCAAAGNCTTTGCAGABF0667susC/susD paralogsThis study
VI1ACTAAGTTCTATCGGBF0667susC/susD paralogsThis study
Open in a separate windowaOur previous classification of the invertible regions identified in B. fragilis strain YCH46 genome (19).bConsensus motif sequences found within IRs are shown. R = A or G, W = A or T, and N = A, G, C, or T.cThe gene identifications in B. fragilis strain YCH46 genome are shown.Our previous studies indicated that an additional inverlon other than the Mpi- and Tsr19-regulated inverlons is present in B. fragilis, based on the finding that at least 10 invertible regions (corresponding to class II regions in B. fragilis strain YCH46) contain a particular consensus motif sequence (AAGTTCN5GAACTT) within their IRs (19) but do not appear to colocalize with a DNA invertase gene. The majority of the class II regions were associated with the selective switching of a particular set of susC/susD family genes. Since the SusC/SusD family of outer membrane proteins play an important role in polysaccharide utilization by Bacteroides (3, 23, 32), the inverlon associated with the phase variation of SusC/SusD family proteins would likely be involved in the survival of this anaerobe in the distal gut.In the present study, we sought to identify the DNA invertase regulating the additional inverlon in B. fragilis. Our results indicated that the Tsr encoded by BF0667 is a master DNA invertase for this inverlon (designated the Tsr0667-inverlon) in B. fragilis.  相似文献   

8.
9.
10.
11.
Predator-prey relationships among prokaryotes have received little attention but are likely to be important determinants of the composition, structure, and dynamics of microbial communities. Many species of the soil-dwelling myxobacteria are predators of other microbes, but their predation range is poorly characterized. To better understand the predatory capabilities of myxobacteria in nature, we analyzed the predation performance of numerous Myxococcus isolates across 12 diverse species of bacteria. All predator isolates could utilize most potential prey species to effectively fuel colony expansion, although one species hindered predator swarming relative to a control treatment with no growth substrate. Predator strains varied significantly in their relative performance across prey types, but most variation in predatory performance was determined by prey type, with Gram-negative prey species supporting more Myxococcus growth than Gram-positive species. There was evidence for specialized predator performance in some predator-prey combinations. Such specialization may reduce resource competition among sympatric strains in natural habitats. The broad prey range of the Myxococcus genus coupled with its ubiquity in the soil suggests that myxobacteria are likely to have very important ecological and evolutionary effects on many species of soil prokaryotes.Predation plays a major role in shaping both the ecology and evolution of biological communities. The population and evolutionary dynamics of predators and their prey are often tightly coupled and can greatly influence the dynamics of other organisms as well (1). Predation has been invoked as a major cause of diversity in ecosystems (11, 12). For example, predators may mediate coexistence between superior and inferior competitors (2, 13), and differential trajectories of predator-prey coevolution can lead to divergence between separate populations (70).Predation has been investigated extensively in higher organisms but relatively little among prokaryotes. Predation between prokaryotes is one of the most ancient forms of predation (27), and it has been proposed that this process may have been the origin of eukaryotic cells (16). Prokaryotes are key players in primary biomass production (44) and global nutrient cycling (22), and predation of some prokaryotes by others is likely to significantly affect these processes. Most studies of predatory prokaryotes have focused on Bdellovibrionaceae species (e.g., see references 51, 55, and 67). These small deltaproteobacteria prey on other Gram-negative cells, using flagella to swim rapidly until they collide with a prey cell. After collision, the predator cells then enter the periplasmic space of the prey cell, consume the host cell from within, elongate, and divide into new cells that are released upon host cell lysis (41). Although often described as predatory, the Bdellovibrionaceae may also be considered to be parasitic, as they typically depend (apart from host-independent strains that have been observed [60]) on the infection and death of their host for their reproduction (47).In this study, we examined predation among the myxobacteria, which are also deltaproteobacteria but constitute a monophyletic clade divergent from the Bdellovibrionaceae (17). Myxobacteria are found in most terrestrial soils and in many aquatic environments as well (17, 53, 74). Many myxobacteria, including the model species Myxococcus xanthus, exhibit several complex social traits, including fruiting body formation and spore formation (14, 18, 34, 62, 71), cooperative swarming with two motility systems (64, 87), and group (or “wolf pack”) predation on both bacteria and fungi (4, 5, 8, 9, 15, 50). Using representatives of the genus Myxococcus, we tested for both intra- and interspecific variation in myxobacterial predatory performance across a broad range of prey types. Moreover, we examined whether prey vary substantially in the degree to which they support predatory growth by the myxobacteria and whether patterns of variation in predator performance are constant or variable across prey environments. The latter outcome may reflect adaptive specialization and help to maintain diversity in natural populations (57, 59).Although closely related to the Bdellovibrionaceae (both are deltaproteobacteria), myxobacteria employ a highly divergent mode of predation. Myxobacteria use gliding motility (64) to search the soil matrix for prey and produce a wide range of antibiotics and lytic compounds that kill and decompose prey cells and break down complex polymers, thereby releasing substrates for growth (66). Myxobacterial predation is cooperative both in its “searching” component (6, 31, 82; for details on cooperative swarming, see reference 64) and in its “handling” component (10, 29, 31, 32), in which secreted enzymes turn prey cells into consumable growth substrates (56, 83). There is evidence that M. xanthus employs chemotaxis-like genes in its attack on prey cells (5) and that predation is stimulated by close contact with prey cells (48).Recent studies have revealed great genetic and phenotypic diversity within natural populations of M. xanthus, on both global (79) and local (down to centimeter) scales (78). Phenotypic diversity includes variation in social compatibility (24, 81), the density and nutrient thresholds triggering development (33, 38), developmental timing (38), motility rates and patterns (80), and secondary metabolite production (40). Although natural populations are spatially structured and both genetic diversity and population differentiation decrease with spatial scale (79), substantial genetic diversity is present even among centimeter-scale isolates (78). No study has yet systematically investigated quantitative natural variation in myxobacterial predation phenotypes across a large number of predator genotypes.Given the previous discovery of large variation in all examined phenotypes, even among genetically extremely similar strains, we anticipated extensive predatory variation as well. Using a phylogenetically broad range of prey, we compared and contrasted the predatory performance of 16 natural M. xanthus isolates, sampled from global to local scales, as well as the commonly studied laboratory reference strain DK1622 and representatives of three additional Myxococcus species: M. flavescens (86), M. macrosporus (42), and M. virescens (63) (Table (Table1).1). In particular, we measured myxobacterial swarm expansion rates on prey lawns spread on buffered agar (31, 50) and on control plates with no nutrients or with prehydrolyzed growth substrate.

TABLE 1.

List of myxobacteria used, with geographical origin
Organism abbreviation used in textSpeciesStrainGeographic originReference(s)
A9Myxococcus xanthusA9Tübingen, Germany78
A23Myxococcus xanthusA23Tübingen, Germany78
A30Myxococcus xanthusA30Tübingen, Germany78
A41Myxococcus xanthusA41Tübingen, Germany78
A46Myxococcus xanthusA46Tübingen, Germany78
A47Myxococcus xanthusA47Tübingen, Germany78
A75Myxococcus xanthusA75Tübingen, Germany78
A85Myxococcus xanthusA85Tübingen, Germany78
TVMyxococcus xanthusTvärminneTvärminne, Finland79
PAKMyxococcus xanthusPaklenicaPaklenica, Croatia79
MADMyxococcus xanthusMadeira 1Madeira, Portugal79
WARMyxococcus xanthusWarwick 1Warwick, UK79
TORMyxococcus xanthusToronto 1Toronto, Ontario, Canada79
SUL2Myxococcus xanthusSulawesi 2Sulawesi, Indonesia79
KALMyxococcus xanthusKalalauKalalau, HI79
DAVMyxococcus xanthusDavis 1ADavis, CA79
GJV1Myxococcus xanthusGJV 1Unknown35, 72
MXFL1Myxococcus flavescensMx fl1Unknown65
MXV2Myxococcus virescensMx v2Unknown65
CCM8Myxococcus macrosporusCc m8Unknown65
Open in a separate window  相似文献   

12.
13.
Animal-to-Animal Variation in Fecal Microbial Diversity among Beef Cattle   总被引:1,自引:0,他引:1  
The intestinal microbiota of beef cattle are important for animal health, food safety, and methane emissions. This full-length sequencing survey of 11,171 16S rRNA genes reveals animal-to-animal variation in communities that cannot be attributed to breed, gender, diet, age, or weather. Beef communities differ from those of dairy. Core bovine taxa are identified.The gastrointestinal tracts (GIT) of beef cattle are colonized by microorganisms that profoundly impact animal physiology, nutrition, health, and productivity (5). The GIT microbiota potentially impact food safety via pathogen shedding (13) by interacting with organisms such as Salmonella and competing for resources in the GIT. Cattle intestinal microbiota also play an important role in methane emissions, with U.S. beef cattle alone contributing an estimated 3.87 million metric tons of methane into the environment each year, both from rumen and large-intestine fermentations (7). Although the bovine fecal microbiota have been well characterized using culture-based methods, these techniques are necessarily limited to characterizing bacteria that can be grown in the laboratory. Culture-independent methods can reveal community members that are recalcitrant to culture. Only a handful of deep-sequencing studies have been done using culture-independent 16S rRNA-based methods (1, 11, 12, 14), all with dairy cattle, which have a fundamentally different diet and metabolism from beef cattle. Despite the potential contributions of the beef cattle GIT microbiota to animal health, food safety, and global warming, these communities remain poorly characterized. With the advent of pyrosequencing technology, researchers now have the tools to characterize these important communities. Pyrosequencing will allow rapid characterization of large-sample data sets (1). However, the taxonomic information generated by rapid sequencing is approximate by necessity (9), and full-length 16S-rRNA sequencing remains the “gold standard” method. Accordingly, we have characterized fecal bacteria from six feedlot cattle by full-length capillary sequence analysis of 11,171 16S rRNA gene clones (Fig. (Fig.11).Open in a separate windowFIG. 1.Bacterial diversity of six feedlot beef cattle. Gray bars represent the percentages of all 16S sequences that were assigned to each taxonomy. Colored dots represent the percentages of 16S sequences from each library that were assigned to each taxonomic group. Asterisks indicate unclassified members of the named taxon. Panel A shows the data for the first 99% of all the sequences. Panel B shows the data for the remaining 1% of sequences. Note differences in scales for panels A and B.Rectal grab fecal samples (n = 6) were collected according to institutional animal care guidelines. All animals were female cross-bred MARCIII beef heifers, 6 to 8 months of age, 214 to 241 kg, housed in the same feedlot pen for 2 months prior to fecal collection, and fed the same typical feedlot beef production growing rations consisting of 61.6% corn silage (41.3% dry matter), 15.2% alfalfa hay, 20.9% corn, and 2.3% liquid supplement.Total fecal DNA was isolated from homogenized samples using MoBio UltraClean fecal kit (Carlsbad, CA). PCR was performed using 27F and 1392R primers (11). Amplification consisted of 25 cycles, with an annealing temperature of 55°C. Amplicons from three reactions per sample were pooled (8), cloned using the Invitrogen TOPO TA cloning kit (Carlsbad, CA), and sequenced bidirectionally with M13 primers using an ABI 3700 sequencer (17). Low-quality and chimeric sequences (6) were excluded from further analysis. Distance matrices were compiled from ClustalW alignments (18) in PHYLIP (4). Pairwise estimates of shared richness were calculated using EstimateS, version 8.2 (R. K. Colwell; http://purl.oclc.org/estimates). DOTUR (16) was used to identify operational taxonomic units (OTUs) and to generate rarefaction curves (Fig. (Fig.2),2), richness and evenness estimates, and Shannon''s and Simpson''s diversity indices (Table (Table1).1). A 97% similarity cutoff and an 85% similarity cutoff for estimating OTUs were used to approximate species and class-level designations (15). Taxonomies were assigned to one member of each OTU using the RDP “classifier” tool (19), and the RDP taxonomic information was used for Fig. Fig.11 and and3.3. Common bovine taxa were identified based on inclusion in all three U.S. culture-independent studies (this study and references 1 and 11).Open in a separate windowFIG. 2.Rarefaction curves for six feedlot beef cattle. OTUs were assigned at the 85% DNA sequence similarity level. For comparison purposes, all six curves were truncated after 1,321 sequences.Open in a separate windowFIG. 3.Phylum-level distribution of bacterial sequences from six beef feedlot cattle. Asterisks indicate unclassified members of the named taxon.

TABLE 1.

Richness and diversity indices for 6 beef feedlot cattle
Library and animal (n)No. of OTUs observedSpecies richness (CI)a by:
Diversity (CI) by:
ChaoACEShannon''s indexSimpson''s index
97% DNA sequence similarity
    Animal 1 (2,485)198372 (294-515)329 (280-408)3.89 (3.83-3.95)0.0422
    Animal 2 (2,084)416600 (538-694)604 (552-675)5.40 (5.35-5.45)0.0066
    Animal 3 (1,710)6961,393 (1,224-1,615)1,418 (1,327-1,523)6.13 (6.08-6.18)0.0027
    Animal 4 (1,512)294526 (439-665)483 (425-566)4.71 (4.63-4.78)0.0237
    Animal 5 (2,059)314612 (495-805)488 (434-566)4.93 (4.88-4.99)0.0126
    Animal 6 (1,321)174320 (252-447)289 (244-361)4.18 (4.11-4.25)0.0286
85% DNA sequence similarity
    Animal 1 (2,485)4861 (51-99)62 (52-90)2.64 (2.59-2.68)0.1056
    Animal 2 (2,084)77107 (87-165)102 (87-139)3.38 (3.34-3.43)0.0505
    Animal 3 (1,710)130153 (139-186)151 (140-174)4.07 (4.02-4.12)0.0254
    Animal 4 (1,512)6675 (68-98)77 (70-96)2.71 (2.64-2.78)0.0931
    Animal 5 (2,059)6980 (72-109)84 (75-110)3.31 (3.26-3.36)0.0545
    Animal 6 (1,321)5465 (57-102)61 (56-76)2.90 (2.83-2.97)0.0939
Open in a separate windowaCI, confidence interval.The GIT community of beef feedlot cattle characterized in this study was found to share many taxa with the bovine GIT community described for dairy cattle (1, 11, 14), although the relative abundances of the major bacterial groups differed considerably. The fecal microbiota of beef cattle were dominated by members of the Firmicutes, with 62.8% of the OTUs belonging to this taxonomic group (Fig. (Fig.3).3). Bacteroidetes (29.5% of the OTUs) and Proteobacteria (4.4% of the OTUs) were also represented in feces (Fig. (Fig.3).3). A total of seven phyla were found in our six animals.Total estimated species richness values (Chao) for each of the six animals were 372, 600, 1,393, 526, 612, and 320 (Table (Table1).1). These cattle richness numbers are higher than those observed for three human subjects (164, 332, and 297) (2). The mean of Chao pairwise estimates of shared richness between any two of the six cattle fecal libraries was 230.Our findings, in addition to those from pyrosequencing studies (1), identify a core set of bovine GIT bacterial taxa, including the Bacteroidetes Prevotella and Bacteroides; the Firmicutes Faecalibacterium, Ruminococcus, Roseburia, and Clostridium; and the proteobacterium Succinovibrio (Fig. (Fig.1).1). These genera are consistently identified in bovine feces and likely compose part of the bovine resident microbiota. Although the potential exists for culture-independent methods to reveal minority microbial community members, 16S rRNA gene sequencing in dairy (1, 11) and beef cattle supports the list of core taxa identified using culture-based methods.Comparisons between our data set and recent studies done with dairy cattle (1, 11, 12) suggest that although beef and dairy cattle share many of the same major bacterial groups, the relative abundances of these groups in beef and dairy cattle may differ, and there may be differences between the two groups in the compositions of minority community members. The most common genus in beef cattle from our study was Prevotella, representing 24% of the total number of sequences evaluated. In comparison, Dowd et al. (1) found that Prevotella spp. represented only 5.5% of the total 16S genes sequenced from 20 dairy cattle, and Prevotella was not listed in the top 10 most frequently occurring OTUs in either of the studies from McGarvey et al. (11, 12). Likewise, Clostridium represented only 1.5% of the total beef sequences but 19% of the dairy pyrosequences (1). There were a number of bacterial sequences present in the beef cattle sequences but not reported in the dairy sequences, including Arthrobacter, Asteroleplasma, Bifidobacterium, Collinsella, Delftia, Eggerthella, Lactobacillus, Mitsuokella, Olsenella, and Propionibacterium (1, 11), although a number of these genera have been cultured from dairy animals in the past. It must be noted that all of these sequencing studies examined only a small number of animals, and each method has limitations which affect interpretation of the results. The full-length sequencing performed as part of this beef cattle study and two dairy studies (11, 12) relies on a PCR step which can potentially affect the relative numbers of each taxon observed due to PCR bias, while the pyrosequeincg method used in the 20-animal dairy study suffers from artifacts that potentially affect taxonomic assignment and richness estimates due to short read lengths and potential biases in evenness (how many of each group) due to primer and template mismatches (3). Nonetheless, these studies indicate that there may be fundamental differences between the gastrointestinal communities of beef and dairy cattle, they provide a comprehensive examination of the communities present in the specific animals tested, and they serve to provide important baseline information for further studies examining various factors which can impact cattle gastrointestinal communities.The taxonomic information generated by deep sequencing of beef cattle feces revealed considerable animal-to-animal variation in the operational taxonomic unit (OTU) composition of the individual libraries (Fig. (Fig.1).1). The OTU designation facilitates an analysis of the community data without forcing the assignment of sequences into an incomplete and imperfect bacterial taxonomic system. It relies on DNA sequence similarity to assign sequences to a particular OTU defined by the level of DNA sequence similarity. In total, 1,906 OTUs (97% OTU designation) were identified in the six libraries. Of these, only 24 OTUs (1.2%) (comprising 1,253 [11.2%] of sequences) were present in all six libraries, while 1,348 OTUs (69%) were found only in individual libraries. Of these, 1,064 OTUs (77%) were unique, represented by a solitary clone (range of 3% to 29% of the total clones from each individual animal). These data hint at considerable animal-to-animal variation in bacterial community structure at the species level that cannot be readily attributed to breed, gender, age, macroecologic factors such as weather conditions, or diet, given that the animals in this study were controlled for these variables, and support the conclusions of Manter et al. (10) that pooling samples can obscure rare phylotypes.Our results from beef cattle suggest that there may be differences in the bacterial community members present in the GIT of each individual animal that cannot be attributed to diet, breed, gender, age, or macroecologic factors such as weather and suggest the need for the high-resolution community sequencing of much larger numbers of animals before “core” minority community members can be identified. Considering the limited nature of the community surveys to date and all of the genetic, management, geographic, and temporal factors that can contribute to the composition of GIT microbiota, much work remains before we are able to understand and predict the community composition of any individual animal.  相似文献   

14.
Melioidosis has been considered an emerging disease in Brazil since the first cases were reported to occur in the northeast region. This study investigated two municipalities in Ceará state where melioidosis cases have been confirmed to occur. Burkholderia pseudomallei was isolated in 26 (4.3%) of 600 samples in the dry and rainy seasons.Melioidosis is an endemic disease in Southeast Asia and northern Australia (2, 4) and also occurs sporadically in other parts of the world (3, 7). Human melioidosis was reported to occur in Brazil only in 2003, when a family outbreak afflicted four sisters in the rural part of the municipality of Tejuçuoca, Ceará state (14). After this episode, there was one reported case of melioidosis in 2004 in the rural area of Banabuiú, Ceará (14). And in 2005, a case of melioidosis associated with near drowning after a car accident was confirmed to occur in Aracoiaba, Ceará (11).The goal of this study was to investigate the Tejuçuoca and Banabuiú municipalities, where human cases of melioidosis have been confirmed to occur, and to gain a better understanding of the ecology of Burkholderia pseudomallei in this region.We chose as central points of the study the residences and surrounding areas of the melioidosis patients in the rural areas of Banabuiú (5°18′35″S, 38°55′14″W) and Tejuçuoca (03°59′20″S, 39°34′50′W) (Fig. (Fig.1).1). There are two well-defined seasons in each of these locations: one rainy (running from January to May) and one dry (from June to December). A total of 600 samples were collected at five sites in Tejuçuoca (T1, T2, T3, T4, and T5) and five in Banabuiú (B1, B2, B3, B4, and B5), distributed as follows (Fig. (Fig.2):2): backyards (B1 and T1), places shaded by trees (B2 and T2), water courses (B3 and T3), wet places (B4 and T4), and stock breeding areas (B5 and T5).Open in a separate windowFIG. 1.Municipalities of Banabuiú (5°18′35″S, 38°55′14″W) and Tejuçuoca (03°59′20″S, 39°34′50″W).Open in a separate windowFIG. 2.Soil sampling sites in Banabuiú and Tejuçuoca.Once a month for 12 months (a complete dry/rainy cycle), five samples were gathered at five different depths: at the surface and at 10, 20, 30 and 40 cm (Table (Table1).1). The samples were gathered according to the method used by Inglis et al. (9). Additionally, the sample processing and B. pseudomallei identification were carried out as previously reported (1, 8, 9).

TABLE 1.

Distribution of samples with isolates by site and soil depth
Sitesa and depth (cm)No. of B. pseudomallei isolates in samples from:
Banabuiú (n = 300)Tejuçuoca (n = 300)Total (n = 600)
B1/T13
    Surface2
    10
    201
    30
    40
B2/T21
    Surface1
    10
    20
    30
    40
B3/T315
    Surface2
    102
    204
    303
    404
B4/T45
    Surface
    101
    201
    3011
    401
B5/T52
    Surface
    10
    20
    302
    40
Total62026
Open in a separate windowaSites designated with B are in Banabuiú, and sites designated with T are in Tejuçuoca. See the text for details.The data on weather and soil composition were obtained from specialized government institutions, such as FUNCEME, IPECE, and EMBRAPA. The average annual temperature in both municipalities is between 26 and 28°C. In 2007, the annual rainfall in Tejuçuoca was 496.8 mm, and that in Banabuiú was 766.8 mm. There are a range of soil types in both Tejuçuoca and Banabuiú: noncalcic brown, sodic planossolic, red-yellow podzolic, and litholic. In Banabuiú, there are also alluvial and cambisol soils. The characteristic vegetation in both municipalities is caatinga (scrublands).There were isolates of B. pseudomallei in 26 (4.3%) of the 600 samples collected. The bacterium was isolated at a rate (3%) similar to that previously reported (9). The bacterium isolation occurred in both the dry (53.8%) and the rainy (46.2%) seasons. Tejuçuoca represented 76.9% (20/26) of the strains isolated. Four sites in Tejuçuoca (T1, T3, T4, and T5) and three in Banabuiú (B1, B2, and B4) presented isolates of the bacterium (Table (Table1).1). The isolation of the B. pseudomallei strains varied from the surface down to 40 cm. However, 17 of the 26 positive samples (65.3%) were found at depths between 20 and 40 cm (Table (Table1).1). Only two isolates were found at the surface during the dry season.A study in Vietnam (13) and one in Australia (9) reported the presence of B. pseudomallei near the houses of melioidosis patients. In our study, the same thing happened. Site T3 (15/26; 57.6%) was located 290 m from the patient''s house, as reported by the Rolim group (14).B. pseudomallei was isolated from a sheep paddock in Australia, where animals sought shelter below mango and fig trees (17). In our study, the bacterium was isolated at site T5, a goat corral alongside the house where the outbreak occurred in Tejuçuoca. Four sites in places shaded by trees yielded positive samples (30.7%) in both Tejuçuoca (palm trees) and Banabuiú (mango trees). Additionally, B. pseudomallei was isolated on three occasions from a cornfield (site 4B) located alongside the house of the melioidosis patient in Banabuiú.In the main areas of endemicity, the disease is more prevalent in the rainy season (4, 5, 16). The outbreak in Tejuçuoca was related to rainfall (14). Besides the association of cases of the disease with rainfall itself, the isolation of B. pseudomallei in soil and water was also demonstrated during the dry season (12, 15). An Australian study isolated strains from soil and water during the dry and rainy seasons (17). A Thai study also reported B. pseudomallei in the dry season (18). In our study, the isolation of B. pseudomallei took place either at the end of the wet season or in the dry months. Fourteen of the positive samples (53.8%) were collected during the dry season, albeit near a river or reservoir (sites T3 and B4).Physical, biological, and chemical soil features appear to influence the survival of B. pseudomallei (6, 10). In the present study, the soil was classified as litholic with sandy or clayey textures. It is susceptible to erosion, and when there is a lack of water, it is subject to salinization. During the dry season, the clay layer becomes dried, cracked, and very hard. During the rainy season, it becomes soggy and sticky. The isolation of B. pseudomallei in the dry season is possibly related to the capacity for adaptation of this soil, since the extreme conditions of lithosols do not prevent the bacterial growth and survival.It has been shown that B. pseudomallei is more often isolated at depths between 25 and 45 cm (17). In our study, 65.3% of the positive samples were taken at depths between 20 and 40 cm. Moreover, of these 17 samples, 10 (58.8%) were collected during the dry months. Also, unlike in other regions, two positive samples were taken from the surface in the period without rainfall.The rainfall in Tejuçuoca and Banabuiú is generally low, and temperatures do not vary significantly during the year. Therefore, the isolation of B. pseudomallei in these places occurs outside the rainfall, temperature, and moisture conditions observed in other regions of endemicity. Our data thus suggest that peculiar environmental features, such as soil composition, might favor the multiplication of B. pseudomallei in northeast Brazil.  相似文献   

15.
This study demonstrates for the first time that a thioether-containing peptide, an azurin fragment, can be translocated via the Sec pathway. This methyl-lanthionine was introduced by the nisin modification enzymes. The Sec pathway can therefore be a successful alternative for those cyclized peptides that are inefficiently transported via NisT.Azurin, a cupredoxin produced by Pseudomonas aeruginosa, can selectively enter human cancer cells and induce apoptosis (24) via binding to the tumor suppressor protein p53 (1). The azurin peptide fragment p28, containing amino acids 50 to 77 (LSTAADMQGVVTDGMASGLDKDYLKPDD), still enters human cancer cells and inhibits tumor proliferation (20). Importantly, novel cancer treatments can be based on azurin peptide fragments and derivatives thereof (T. Das Gupta and A. Chakrabarty, 20 March 2008, patent application WO2008033820). Although the pharmacokinetic property of therapeutic peptides is promising, lack of biostability is the major hurdle for their successful application. Consequently, it is very relevant to explore the possibilities for enhancing biostability of these peptides.In our group, we developed a technology to improve the stability of therapeutic peptides by exploiting the nisin synthetase enzymes NisB and NisC for the introduction of thioether bridges. We applied a two-plasmid expression system (7, 8, 12), in which the NisBTC-encoding plasmid is compatible with the substrate-peptide-encoding plasmid. Lactococcus lactis containing this expression system can secrete nonlantibiotic peptides which are dehydrated or stabilized by a thioether ring (8, 16). NisB dehydrates serines and threonines in substrate peptides, NisC couples dehydrated residues stereo- and regioselectively to cysteines, and NisT, the ABC transporter, translocates the modified peptides out of the cell (10, 11, 13, 15). The leader peptide is essential for targeting and modification of the propeptides (23).When transport via NisT is impaired or is less efficient, the Sec pathway of L. lactis is a successful alternative in translocation of dehydrated peptides. When the nisin leader is preceded by a Sec signal peptide or a Tat signal peptide 27 or 44 amino acids long, respectively, modification by NisB and NisC still occurs (12; G. N. Moll, A. Kuipers, R. Rink, A. J. M. Driessen, and O. P. Kuipers, 15 June 2006, patent application WO 2006062398). However, NisC-cyclized prenisin was not translocated via the Sec system (12). This is likely due to the dimensions of fully modified nisin (3), which is too large to fit in the SecY pore (12, 21). Here, we report for the first time that the Sec pathway of L. lactis can translocate a p28 azurin fragment analog with a thioether ring.We previously demonstrated that under culturing conditions, the highly reactive dehydroalanines can spontaneously couple to cysteines, either intra- or extracellularly, whereas the less reactive dehydrobutyrines do not (16). To exclude spontaneous thioether ring formation by dehydroalanines, we mutated serines in positions 51 and 66 to alanines, whereas a single dehydratable threonine was kept in position 52 of the azurin(50-77) peptide fragment. Position 56 was mutated to a cysteine to allow posttranslational introduction of a thioether bridge (Table (Table1;1; pNZ8048-derived plasmids).

TABLE 1.

Bacterial strains and plasmids used in this study
Strain or plasmidCharacteristicsaReference
Strain
    NZ9000MG1363 derivative; pepN::nisRK+5
Plasmids
    pIL253 derived19
        pIL3BTCnisBTC cloned behind Pnis promoter; Cmr17
        pILBCnisBC cloned behind Pnis promoter; Emr12
        pILBnisB cloned behind Pnis promoter; Emr12
    pNZ8048 derived14
        pNZazuPnis + sequence encoding nisin leader fused to azurin(50-77) peptide fragment; EmrThis study
        pNG41azuPnis + sequence encoding SPUsp45 fused to nisin leader fused to azurin(50-77) peptide fragment; CmrThis study
        pNG51azuPnis + sequence encoding SPYwbN fused to nisin leader fused to azurin(50-77) peptide fragment; CmrThis study
Open in a separate windowaPnis is the nisin-inducible nisA promoter; Emr, erythromycin resistance; Cmr, chloramphenicol resistance. Boldface indicates mutations: S51A, M56C, and S66A in the azurin(50-77) peptide fragment (LATAADCQGVVADGMAAGLDKDYLKPDD).When the azurin peptide fragment fused behind the nisin leader was coexpressed with the enzymes NisB, NisC, and NisT in L. lactis, no secreted (un)modified peptides in the supernatant were detected. Hence, we made use of the Sec pathway of L. lactis for export of the azurin peptide fragment. The nisin leader of the substrate peptide was preceded by the Sec signal peptide of Usp45, SPUsp45. This fusion peptide was coexpressed with NisB and NisC in L. lactis in the absence of NisT. The strains and plasmids used are listed in Table Table1.1. The culture of L. lactis NZ9000(pNG41azurin)(pILBC) was grown in minimal medium. Peptides from 4-ml induced cultures were isolated and purified with bond elute C18 cartridges from Varian. Dissolved peptides were analyzed by mass spectrometry directly, as well as after incubation with TCEP [Tris(2-carboxyethyl)phosphine] and, subsequently, CDAP (1-cyano-4-dimethylamino-pyridinium tetrafluoroborate) (16). Mass spectra were recorded with a Voyager DE PRO matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometer in the linear mode.We were able to detect Sec-secreted azurin peptides in the supernatant by mass spectrometry. Some of the dehydrated peptides in the supernatant contained disulfide-bonded cysteine adducts, meaning that not all of the formed dehydrobutyrines were coupled to the peptide''s cysteines (data not shown). This observation of partial ring formation was confirmed by mass spectrometry. Peptides were first reduced with the phosphine TCEP and then alkylated with CDAP; formation of the thiocyanate results in a mass shift of +25 Da. (Fig. (Fig.1A).1A). These data indicate that transport of an azurin peptide fragment with a thioether ring via the Sec pathway is possible, when preceded by SPUsp45 and the nisin leader.Open in a separate windowFIG. 1.Transport of cyclized and dehydrated azurin peptide fragments via the Sec pathway. Culture supernatant was analyzed by matrix-assisted laser desorption ionization-time of flight mass spectrometry. The expected mass of the processed, protonated, and fully dehydrated fusion peptide (STKDFNLDLVSVSKKDSGASPR-LATAADCQGVVADGMAAGLDKDYLKPD) is 5,141 Da. (A) Supernatant of L. lactis NZ9000(pILBC)(pNG41azu), with the fusion peptide preceded by SPUsp45. (B) Supernatant of L. lactis NZ9000(pILBC)(pNG51azu), with the fusion peptide preceded by SPYwbN. (C) Supernatant of control L. lactis NZ9000(pILB)(pNG51azu).As demonstrated before, replacement of SPUsp45 with a Tat signal peptide of YwbN from Bacillus subtilis, SPYwbN, resulted in reduced transport efficiency and simultaneously enhanced the extent of dehydration of the substrate peptide (12). To examine the effect of reduced transport on the extent of NisC-mediated cyclization, SPYwbN was fused N terminally to the nisin leader and azurin peptide fragment (Table (Table1)1) and coexpressed with NisB and NisC. Peptides in the supernatant of induced cultures were analyzed by mass spectrometry as described above and analyzed on a Tricine gel (18) by silver staining (Invitrogen). As a control, the substrate peptide was also coexpressed with only NisB. In the case of coexpression of NisB and NisC, the secreted peptides were almost fully dehydrated and no cysteinylations were seen (data not shown), suggesting that the peptides were fully ring closed. TCEP treatment and CDAP incubation (Fig. (Fig.1B)1B) of the purified isolated peptides confirmed this observation. As expected, in the supernatant of the control which had only coexpression of NisB (pILB), fully dehydrated peptides were seen. No cyclization took place, and therefore all of the free cysteines reacted with CDAP (Fig. (Fig.1C).1C). This control experiment clearly demonstrated that no spontaneous cyclization had occurred in the absence of NisC. Hence, the thioether bridge formation observed in the experiments presented in Fig. 1A and B should result from intracellular NisC-mediated cyclization. The data therefore convincingly prove that indeed this intrinsically stable thioether-bridged peptide is transported via the Sec system.Analyses on silver-stained gel showed that no transport at all via the transporter NisT had occurred when the azurin peptide fragment was preceded by only the nisin leader (Fig. (Fig.2,2, lane 1). In full contrast, transport of the modified azurin peptide fragment via the Sec pathway was successful. The transport of the peptide fragment was less efficient when SPYwbN was used instead of SPUsp45 (Fig. (Fig.2),2), which is in full agreement with previous data (12).Open in a separate windowFIG. 2.Amount of secreted peptides in the supernatant. Peptides from 2 ml of supernatant of induced cultures were applied to a gel. Lanes: M, kaleidoscopic marker (Bio-Rad); 1, NZ9000(pIL3BTC)(pNZazu); 2, NZ9000(pIL3BC)(pNG41azu); 3, NZ9000(pIL3BC)(pNG51azu).These data demonstrate for the first time that a peptide with an intramolecular thioether bridge can be translocated in vivo via the Sec pathway of L. lactis. A Sec system that has been studied in more detail is that of Escherichia coli. The Sec translocase in the membrane is composed of a highly conserved protein-conducting channel, SecYEG (6). The Sec translocase transports unfolded proteins, which is driven by the ATPase SecA. Homologues of SecYEG and SecA are also found in L. lactis (2, 9). In vitro studies demonstrated that the translocon SecYEG of E. coli can also translocate the polypeptide proOmpA with a disulfide-bridge, which can have a loop of 18 amino acids or smaller (22). Other in vitro data demonstrated that proOmpA labeled with bulky fluorescent probes, assessing up to 16 Å, was also transported (4). These in vitro data with the Sec system of E. coli suggested that the SecY translocon is not that rigid and could be used for other purposes, such as the in vivo translocation in L. lactis of peptides with thioether bridges. Interestingly, the efficiency of translocation in E. coli can be drastically enhanced by prlA (secY) mutations (6). Likely, in the near future such mutations in the SecY translocon of L. lactis can contribute to an even more successful application of the Sec pathway for transport of therapeutic peptides with thioether bridges.  相似文献   

16.
17.
The rates of pilin antigenic variation (Av) of two strains of Neisseria meningitidis were determined using an unbiased DNA sequencing assay. Strain MC58 underwent pilin Av at a rate similar to that of N. gonorrhoeae strain MS11 but lower than that of N. gonorrhoeae strain FA1090. Pilin Av was undetectable in strain FAM18.Neisseria meningitidis is a Gram-negative diplococcus that colonizes the nasopharynx of approximately 5 to 10% of the population and is usually nonpathogenic but can occasionally enter the bloodstream to cause septicemia and can eventually spread to the meninges, causing meningitis (15). Approximately 500,000 cases of meningococcal meningitis occur every year, with nearly 10% resulting in fatality (2).Type IV pili (TFP) are long filamentous structures protruding from the bacterial surface and are required for adherence of N. meningitidis to host cells (7). As with the TFP of the closely related pathogen Neisseria gonorrhoeae, the pili are able to undergo antigenic variation (Av). In N. gonorrhoeae, pilin Av occurs as a result of recombination between one of the multiple silent pilS copies and the expressed pilin gene (pilE). The pilS copies share significant regions of homology with pilE yet lack a promoter or ribosome-binding site and the initial 5′ coding sequence. Pilin Av relies on RecA and the RecF-like recombination pathway to catalyze gene conversion, resulting in an altered pilE sequence, carrying part of the pilS donor, and the original unaltered pilS sequence (8, 9).While the frequency of pilin Av has been measured in N. gonorrhoeae (5, 10, 12), this process has never been quantified in N. meningitidis. Two sequenced strains were picked to measure pilin Av: serogroup B strain MC58 (sequence type [ST-32] complex), isolated from an invasive infection (14), and serogroup C strain FAM18 (ST-11 complex), which was isolated from a patient with septicemia (1). In both strains, the native recA gene was replaced with the very highly conserved N. gonorrhoeae recA6 construct, which allows regulation of expression with IPTG (isopropyl-β-d-thiogalactopyranoside) (17). recA6 strains are RecA+ when grown with IPTG but are RecA when grown without IPTG (17). These phenotypes were confirmed by measuring the UV sensitivities and DNA transformation competence levels of both strains with or without IPTG, and both strains were shown to be piliated by transmission electron microscopy (data not shown). Bacteria were grown at 37°C with 5% CO2 on gonococcal medium base (GCB; Difco) plus Kellogg supplements I and II (11).The pilin Av sequencing assay was performed as described previously (5, 10, 12) with slight modifications. Briefly, FAM18 and MC58 were grown on solid GCB with 1 mM IPTG, allowing for the expression of RecA, for 22 h and 12.5 h, respectively, which was estimated to produce 20 generations. For FAM18, little or no pilin Av was expected since the G-quartet-forming sequence required for pilin Av is degenerate in this strain (3). Therefore, two random progenitor colonies were picked from IPTG-enriched medium and passaged on GCB without IPTG. Between 91 and 94 colonies were isolated from each FAM18 progenitor, and the sequence of the pilE gene was determined. For MC58, seven random progenitor colonies were picked and passaged on GCB without IPTG. Between 28 and 47 progeny colonies arising from each of the seven progenitors were isolated, and the pilE gene sequence was determined. In both MC58 and FAM18, the progeny colonies were passaged on GCB two times to ensure colony clonality. A single colony from each sample was isolated, and the pilE gene was PCR amplified as described previously (13).The primers used for amplification of MC58 pilE were McPilRBS (5′-GCATTTCCTTTCCAATTAGGAG) and MC58SP3A (5′-TTCCGTACGGATAGCTTCGTC). The primers used for amplification of FAM18 pilE were FAMFOR-2 (5′-ATTACGGGTTTACGTTTGCGG) and FAMREV-2 (5′-ACGCACCTACGCCTCACCCTAC). The DNA sequence was determined for each sample (SeqWright, Houston, TX, and the Genomics Core at Northwestern University) and analyzed (MacVector; Symantec Corp.). Colonies that showed pilE sequence changes were reanalyzed to confirm the Av event.The pilin Av frequency was determined for the progeny of each progenitor by dividing the total number of detected pilin Av events by the number of progeny of each set, resulting in two values for FAM18 and seven values for MC58 (Table (Table1).1). The pilin Av rate was determined by dividing the pilin Av frequency by the number of generations for each sample grown in the presence of IPTG, as determined by a colony assay at the time of harvest. After growth for the same time period, the total numbers of generations for MC58 and FAM18 grown under RecA induction were approximately 19 and 23, respectively.

TABLE 1.

Frequencies and rates of pilin Av in MC58 and FAM18a
Strain and progenitorNo. of progeny analyzedNo. of pilin Av events detectedPilin Av frequency (events/CFU)Pilin Av rate (events/CFU/ generation)
MC58
    A3060.21.0 × 10−2
    B45000
    C3120.063.1 × 10−3
    D4710.021.0 × 10−3
    E45000
    F3130.15.2 × 10−3
    G3110.031.6 × 10−3
FAM18
    A9400 (<0.01)0 (<4.6 × 10−4)
    B9100 (<0.01)0 (<4.7 × 10−4)
Open in a separate windowaValues in parentheses indicate the detection limit of pilin Av in this assay.MC58 possesses eight pilS copies in a single locus directly upstream of pilE, designated pilS1 to pilS8 (GenBank accession numbers NMB0019 to NMB0026, respectively) (16). FAM18 possesses two pilS sequences in a single locus, designated pilS1 and pilS2 (GenBank accession numbers NMC0002 and NMC0003, respectively) (1). All MC58 pilin Av events were confirmed by comparing the sequence of the altered pilE to the eight pilS sequences, and each was matched to a pilS sequence donor.As predicted, no pilin Av was detected in FAM18 within the 185 progeny colonies analyzed (Table (Table1).1). Therefore, the pilin Av frequency is less than the detection limit of 5.4 × 10−3 events/CFU and the rate is less than the detection limit of 2.3 × 10−4 events/CFU/generation. This result is consistent with observations from clinical ST-11 isolates which have a conserved pilE sequence (4).In contrast, pilin Av in MC58 was detected in five of the seven sets, with the highest frequency and rate belonging to set A, which had a frequency of 0.2 events/CFU and a rate of 1 × 10−2 events/CFU/generation (Table (Table1).1). The median frequency of pilin Av in MC58 was 0.03 events/CFU, and the median rate was 1.6 × 10−3 events/CFU/generation. Using the Wilcoxon rank sum test, the rates of pilin Av of MC58 were statistically reduced relative to the rates previously reported for N. gonorrhoeae strain FA1090 grown for 20 generations (10), with a P value of <0.05. In contrast, the rates of pilin Av of MC58 and gonococcal strain MS11, which was shown to have a reduced level of pilin Av relative to that of strain FA1090 from the same study (10), were not statistically different from each other.The pilS donor was determined for all 13 MC58 samples with a variant pilE gene. Five pilin Av events were the result of recombination with pilS3. One pilin Av event was the result of recombination with pilS1. One event was the result of recombination with pilS8. Five events could have been the result of recombination with pilS1, pilS2, or pilS3 in areas of these pilS copies that are identical. One event was the result of recombination with either pilS5 or pilS7. No recombination events with either pilS4 or pilS6 were detected. While these data suggest that there is a nonrandom distribution of donor pilS copies, which has been shown in N. gonorrhoeae (4, 9), further studies with more samples will have to be performed to verify this.These results definitively demonstrate that N. meningitidis strain MC58 undergoes pilin Av at a rate similar to that of N. gonorrhoeae strain MS11 yet reduced relative to that of N. gonorrhoeae strain FA1090. N. meningitidis strain FAM18, which lacks a well-defined G-4 quartet and encodes class II pilin (6, 18), undergoes pilin Av at a rate not detectable by the assay described here and is unlikely to undergo antigenic variation at all. These data clearly show that major differences in strain-to-strain pilin Av rates exist, an observation previously made for N. gonorrhoeae (10). The greatly reduced level of pilin Av in FAM18 suggests that this strain is not dependent upon pilin Av for its survival and spread from host to host and that other class II pilin gene-expressing strains (4) must have a relationship with the host immune system different from that of class I pilin-expressing strains. Whether there are differences in transmission, pilus function, or interactions with the host remains to be determined.  相似文献   

18.
The presence of Campylobacter spp. was investigated in 41 Antarctic fur seals (Arctocephalus gazella) and 9 Weddell seals (Leptonychotes weddellii) at Deception Island, Antarctica. Infections were encountered in six Antarctic fur seals. The isolates, the first reported from marine mammals in the Antarctic region, were identified as Campylobacter insulaenigrae and Campylobacter lari.The Antarctic and sub-Antarctic regions are often regarded as pristine landscapes, unaffected by human activity. A limited number of surveys have been carried out to investigate the possible occurrence of zoonotic enteropathogens and if certain bacteria could be used as tools for evaluating biological pollution in this area (4, 11). In the case of Campylobacter species, there have been only three reports in the literature, but in all of them Campylobacter was isolated from marine seabirds but not from marine mammals. Campylobacter jejuni was isolated in Antarctic and sub-Antarctic areas from Macaroni penguins (Eudyptes chrysolophus) (4), and Campylobacter lari was isolated from Brown skuas, South Polar skuas, and Adelie penguins (2, 11).Reports of Campylobacter species isolated from marine mammals are rare. Campylobacter insulaenigrae was isolated from three harbor seals (Phoca vitulina) and a harbor porpoise (Phocoena phocoena) in Scotland (7). The isolation of C. jejuni, C. lari, and an unknown Campylobacter species from juvenile northern elephant seals (Mirounga angustirostris) in California was also reported (22). Finally, 71 isolates of C. insulaenigrae and 1 isolate similar to but distinct from both Campylobacter upsaliensis and Campylobacter helveticus were isolated from northern elephant seals in California (23). In the South Georgia Archipelago, fecal swabs were taken from 206 Antarctic fur seal pups, but no isolates could be obtained (4). In this study, we successfully isolated C. lari from 7.3% of Antarctic fur seals (Arctocephalus gazella) sampled and C. insulaenigrae from a further 7.3%. On the other hand, Campylobacter was not detected in the nine Weddell seals (Leptonychotes weddellii) sampled. To our knowledge, this is the first report on the isolation of C. lari and C. insulaenigrae from marine mammals in the Antarctic region.Fieldwork was conducted at Deception Island (latitude of 62°58′S and longitude of 60°40′W), in the South Shetland Islands. During January to February 2007, Antarctic fur seals (Arctocephalus gazella) and Weddell seals (Leptonychotes weddellii) were captured and fecal samples were collected by insertion of sterile cotton wool swabs into the rectum of the marine mammals. A total of 41 Antarctic fur seals and 9 Weddell seals were sampled. The distribution by ages was of 7 adults (over 4 years of age with breeding activity), 19 subadults (2 to 4 years of age), and 15 juvenile Antarctic fur seals (less than 2 years of age), and 8 adult Weddell seals and 1 juvenile. All animals presented a good body condition and showed no symptoms at the time of sampling.Three swabs were taken from each animal and were placed in FBP medium (8) with 0.5% active charcoal (Sigma Ltd.), Amies transport medium with charcoal, and Cary Blair transport medium, respectively. All samples were kept at +4 to 8°C until culture in the lab. The number of days between sampling and cultivation varied from 96 to 124 days, with a median value of 105 days.Each swab was placed in 10 ml of Campylobacter enrichment broth (Lab M) with 5% laked horse blood and CAT supplement (cefoperazone [8 μg/ml], teicoplanin [4 μg/ml], and amphotericin B [10 μg/ml]) at 37°C. The broth was incubated at 37°C for 48 h and 5 days in 3.5-liter anaerobic containers using CampyGen sachets (Oxoid), before an aliquot of 100 μl was plated onto CAT agar and the plates were incubated at 37°C for 72 h in a microaerobic atmosphere. In addition, a 47-mm-diameter cellulose membrane with 0.60-μm pores was placed on the surface of an anaerobe agar base (Oxoid) with 5% laked horse blood. Eight to 10 drops of enrichment broth (200 μl) were placed onto the surface of the membrane. The membrane was left for 20 to 30 min on the agar surface at room temperature until all of the fluid had passed through (20). The plates were incubated as described above, but for 5 days to isolate the less common, slower growing species.Isolates were examined by dark-field microscopy to determine morphology and motility and tested to determine whether oxidase was produced. For each sample, five isolates from each of the solid media that had typical morphology and motility and for which the oxidase test was positive were frozen at −80°C in FBP medium (8) until they were tested by phenotypic and genotypic methods.Original Campylobacter identification was done by Gram staining, catalase activity, hippurate hydrolysis, ability to hydrolyze indoxyl acetate, urease activity, H2S production on triple-sugar iron slants, growth at 25°C and 42°C in a microaerophilc environment, growth at 37°C in an aerobic atmosphere, and agglutination with Microscreen latex (Microgen, Camberley, United Kingdom).No differences between the strains were observed in any of the phenotypic tests used. All isolates showed a Gram-negative, slender, curved, seagull wing-like morphology under light microscopy and positive reactions in the catalase test. They were negative for hippurate and indoxyl acetate hydrolysis and urease and did not show H2S production. In addition, they grew at 42°C but did not grow at 25°C or 37°C in an aerobic atmosphere. Finally, all of them were positive in the agglutination test.Because phenotypic results commonly lead to misidentification of Campylobacter species, it is recommended that a molecular method be included in the identification scheme for Campylobacter (5, 15). Identification of the isolates was performed using 16S rRNA gene PCR and sequence analysis (15, 21). Forward and reverse conserved 16S rRNA eubacterial primers 8F (5′-AGAGTTTGATCCTGGCTCAG-3′) and 1492R (5′-GGTTACCTTGTTACGACTT-3′) were used to amplify the 16S rRNA according to the protocol described by Jang et al. (9). Forward and reverse sequencing reactions were performed by the Laboratorio Central de Veterinaria''s DNA sequencing facility (LCV Algete, Madrid, Spain). Three strains were identified as C. lari and the other three as C. insulaenigrae based on both forward and reverse sequence analysis.Molecular characterization of strains was carried out using a combination of pulsed-field gel electrophoresis (PFGE) using KpnI enzyme and multilocus sequence typing (MLST). Preparation of intact Campylobacter DNA for PFGE was performed following the Pulsenet protocol (17, 24). PFGE for the restriction enzyme KpnI (Takara, Conda, Spain) was performed following the protocol described by Ribot et al. (17). DNA fragments were resolved on 0.9% Seakem Gold agarose gels (Iberlabo, Spain) with a Bio-Rad CHEF DRIII system (Bio-Rad, Spain) at 14°C and 6 V/cm. Electrophoresis was carried out for 22 h with pulse times ramping from 4 s to 20 s. The fingerprinting experiments were analyzed using the InfoQuest FP software (Bio-Rad, Spain), and the dendrogram was constructed using the unweighted-pair group method using average linkages (UPGMA).MLST of C. lari strains was performed as described by Miller et al. (13). In the case of C. insulaenigrae strains, MLST was performed following the protocol described by Stoddard et al. (23). All amplicons were sequenced by the Sequencing Service of the Instituto de Salud Carlos III (Madrid, Spain). Sequence data were collated, and alleles were assigned using the Campylobacter PubMLST database (http://pubmlst.org/campylobacter/). Novel alleles and sequence types were submitted for allele and sequence type (ST) designations when appropriate.Regarding the age distribution of animals, C. lari was isolated from 1 of 7 adult (14.3%), 1 of 19 subadult (5.3%), and 1 of 15 juvenile (6.6%) Antarctic fur seals. C. insulaenigrae was isolated from 1 of 7 adults (14.3%) and 2 of 19 of subadults (10.5%) but not from juvenile animals (Table (Table1).1). All strains were obtained from the swabs kept in FBP transport medium.

TABLE 1.

Source of Campylobacter isolates
AnimalAge category and sexDate (mo/day/yr) of:
Campylobacter sp. and isolate no.
SamplingCulture
L 06/56Adult male2/15/075/30/07C. insulaenigrae FR-07
L 06/78Subadult male2/15/075/30/07C. insulaenigrae FR-15
L 06/102Subadult male2/22/075/30/07C. lari FR-28
L 06/134Juvenile male2/21/075/30/07C. lari FR-36
L 06/146Subadult male2/22/075/30/07C. insulaenigrae FR-38
L 06/48Adult male2/22/075/30/07C. lari FR-48
Open in a separate windowCampylobacter is very sensitive to excessive amounts of oxygen and has little capacity to survive in the environment. It is therefore possible that the prevalence of Campylobacter species in Antarctic fur seals is greater than that obtained in our survey and that we have isolated more-resistant strains with a larger ability to survive a prolonged transport. Nevertheless, we think that the freezing medium described by Gorman and Adley (8) modified by the addition of 0.5% of activated charcoal is a very good transport medium since the bacteria remained viable for 3 months at refrigeration temperature, whereas they did not survive in the transport media routinely used for the preservation of fecal samples such as Amies and Cary Blair media.PFGE is a useful tool for conducting epidemiological studies of Campylobacter species. We used digestion with KpnI because it has been reported to have greater power of discrimination than digestion with SmaI (16). All isolates showed very different patterns (Fig. (Fig.1),1), indicating different sources of infection and circulation of different clones on Deception Island. These data were confirmed by the results of MLST, in which each strain belonged to a different ST, none of which had been previously reported. We submitted to the MLST database 12 new sequences of alleles tested for C. insulaenigrae and 10 new sequences of C. lari obtained (Table (Table22).Open in a separate windowFIG. 1.UPGMA dendrogram of PFGE profiles.

TABLE 2.

Alelle numbers and sequence types of Campylobacter isolates
Species and isolate no.STAllele no.a
aspA or adkatpAglnAglyApgipgmtkt
C. insulaenigrae
    FR-7412 (aspA)16*12*215*15*11*
    FR-15424 (aspA)1011*12*14*15*12*
    FR-38437 (aspA)17*11*13*14*313*
C. lari
    FR-281752* (adk)57*250*56*51*31*
    FR-361652* (adk)57*2256*52*31*
    FR-481853* (adk)58*1257*52*32*
Open in a separate windowaAsterisks indicate new alleles.The introduction of C. lari in the Antarctic fur seal colonies may have occurred through seabirds. C. lari has been isolated from Adelie penguins (Pygoscelis adeliae), kelp gull (Larus dominicanus), Brown skuas (Stercorarius antarctica lonnbergi), and South Polar skuas (Stercorarius maccormicki) in Hope Bay (11) and in the Antarctic Peninsula (2). Gulls can travel between South America and Antarctica and are potential carriers of enteric pathogens (1). Thus, C. lari has been isolated from kelp gulls in southern Chile (6). Also, South Polar skuas have been reported in Greenland and the Aleutian Islands and Brown skuas move around the Antarctic coast. Therefore, it is possible that these birds acquire infectious organisms when they move to areas with high levels of human activity. These birds have been reported on Deception Island (10), and it is common to find skuas and giant petrels on beaches where Antarctic fur seal colonies rest. The carrier birds could eliminate Campylobacter and pollute these areas. Alternatively, these birds could be occasional prey for Antarctic fur seals.C. insulaenigrae is a new Campylobacter species whose host range might be restricted to marine mammals (23). It could be hypothesized that C. insulaenigrae evolved from C. lari based on the presence of both species in sea lions and their sharing other characteristics such as the absence of the citrate synthase gene (23). In addition, considering that C. insulaenigrae has not been isolated from seabirds or shellfish and the migration ranges of sea lions are generally not very large, Antarctic fur seals could have been initially infected with C. lari, and subsequently this species has evolved, adapting to mammals. Alternatively both species could share an ancestor and have adapted to different hosts.The Antarctic fur seals captured showed no weight loss, diarrhea, or other symptoms at the time of sampling. However, due to the nature of our study, it is not possible to know whether the animal had been ill before the time of collection and was subsequently a carrier. Taking into account previous reports (7, 23) and our results, pinnipeds could possibly act as reservoir of C. insulaenigrae.The presence of Campylobacter in Antarctic fur seals could also be important due to the zoonotic potential of both species (5, 12, 18, 19). Therefore, researchers should continue to exercise caution when working with these animals. In addition, C. lari has been involved in waterborne outbreaks (3) and some reports have identified this species as the most frequently isolated from surface water (25). Most of the Antarctic stations'' catchwater from lakes generated by meltwater and the water treatment cannot be accomplished by chemical products to prevent marine pollution. In general, water is not treated or is treated only by filtration and UV light. Antarctic fur seals can nevertheless pollute the water of these lakes and/or infect other species such as penguins and other birds, which in turn could also act as a source of infection for humans. Furthermore, Obiri-Danso et al. (14) have reported that C. lari survives for longer in surface waters than C. jejuni and Campylobacter coli, so it would have a greater chance of surviving the water treatment. Finally, in case of infection, the therapy may be complicated because in many of the stations there are only basic medical services.In summary, we describe here the first isolation and characterization of two species of Campylobacter, C. lari and C. insulaenigrae, from Antarctic fur seals. Further studies are needed to determine the prevalence of Campylobacter spp. in Antarctic pinnipeds, the possible sources of infection and if the presence of Campylobacter in marine mammals could be a risk for human illness or could be a result of microbial pollution associated with human activity.  相似文献   

19.
The biological, serological, and genomic characterization of a paramyxovirus recently isolated from rockhopper penguins (Eudyptes chrysocome) suggested that this virus represented a new avian paramyxovirus (APMV) group, APMV10. This penguin virus resembled other APMVs by electron microscopy; however, its viral hemagglutination (HA) activity was not inhibited by antisera against any of the nine defined APMV serotypes. In addition, antiserum generated against this penguin virus did not inhibit the HA of representative viruses of the other APMV serotypes. Sequence data produced using random priming methods revealed a genomic structure typical of APMV. Phylogenetic evaluation of coding regions revealed that amino acid sequences of all six proteins were most closely related to APMV2 and APMV8. The calculation of evolutionary distances among proteins and distances at the nucleotide level confirmed that APMV2, APMV8, and the penguin virus all were sufficiently divergent from each other to be considered different serotypes. We propose that this isolate, named APMV10/penguin/Falkland Islands/324/2007, be the prototype virus for APMV10. Because of the known problems associated with serology, such as antiserum cross-reactivity and one-way immunogenicity, in addition to the reliance on the immune response to a single protein, the hemagglutinin-neuraminidase, as the sole base for viral classification, we suggest the need for new classification guidelines that incorporate genome sequence comparisons.Viruses from the Paramyxoviridae family have caused disease in humans and animals for centuries. Over the last 40 years, many paramyxoviruses isolated from animals and people have been newly described (16, 17, 22, 29, 31, 32, 36, 42, 44, 46, 49, 58, 59, 62-64). Viruses from this family are pleomorphic, enveloped, single-stranded, nonsegmented, negative-sense RNA viruses that demonstrate serological cross-reactivity with other paramyxoviruses related to them (30, 46). The subfamily Paramyxovirinae is divided into five genera: Respirovirus, Morbillivirus, Rubulavirus, Henipavirus, and Avulavirus (30). The Avulavirus genus contains nine distinct avian paramyxovirus (APMV) serotypes (Table (Table1),1), and information on the discovery of each has been reported elsewhere (4, 6, 7, 9, 12, 34, 41, 50, 51, 60, 68).

TABLE 1.

Characteristics of prototype viruses APMV1 to APMV9 and the penguin virus
StrainHostDiseaseDistributionFusion cleavagecGI accession no.
APMV1/Newcastle disease virus>250 speciesHigh mortalityWorldwideGRRQKRF45511218
InapparentWorldwideGGRQGRLa11545722
APMV2/Chicken/CA/Yucaipa/1956Turkey, chickens, psittacines, rails, passerinesDecrease in egg production and respiratory diseaseWorldwideDKPASRF169144527
APMV3/Turkey/WI/1968TurkeyMild respiratory disease and moderate egg decreaseWorldwidePRPSGRLa209484147
APMV3/Parakeet/Netherlands/449/1975Psittacines, passerines, flamingosNeurological, enteric, and respiratory diseaseWorldwideARPRGRLa171472314
APMV4/Duck/Hong Kong/D3/1975Duck, geese, chickensNone knownWorldwideVDIQPRF210076708
APMV5/Budgerigar/Japan/Kunitachi/1974Budgerigars, lorikeetsHigh mortality, enteric diseaseJapan, United Kingdom, AustraliaGKRKKRFa290563909
APMV6/Duck/Hong Kong/199/1977Ducks, geese, turkeysMild respiratory disease and increased mortality in turkeysWorldwidePAPEPRLb15081567
APMV7/Dove/TN/4/1975Pigeons, doves, turkeysMild respiratory disease in turkeysUnited States, England, JapanTLPSSRF224979458
APMV8/Goose/DE/1053/1976Ducks, geeseNone knownUnited States, JapanTYPQTRLa226343050
APMV9/Duck/NY/22/1978DucksNone knownWorldwideRIREGRIa217068693
APMV10/Penguin/Falkland Islands/324/2007Rockhopper penguinsNone KnownFalkland IslandsDKPSQRIa300432141
Open in a separate windowaRequires the addition of an exogenous protease.bProtease requirement depends on the isolate examined.cPutative.Six of these serotypes were classified in the latter half of the 1970s, when the most reliable assay available to classify paramyxoviruses was the hemagglutination inhibition (HI) assay (61). However, there are multiple problems associated with the use of serology, including the inability to classify some APMVs by comparing them to the sera of the nine defined APMVs alone (2, 8). In addition, one-way antigenicity and cross-reactivity between different serotypes have been documented for many years (4, 5, 14, 25, 29, 33, 34, 41, 51, 52, 60). The ability of APMVs, like other viruses, to show antigenic drift as it evolves over time (37, 43, 54) and the wide use and availability of precise molecular methods, such as PCR and genome sequencing, demonstrate the need for a more practical classification system.The genetic diversity of APMVs is still largely unexplored, as hundreds of avian species have never been surveyed for the presence of viruses that do not cause significant signs of disease or are not economically important. The emergence of H5N1 highly pathogenic avian influenza (HPAI) virus as the cause of the largest outbreak of a virulent virus in poultry in the past 100 years has spurred the development of surveillance programs to better understand the ecology of avian influenza (AI) viruses in aquatic birds around the globe, and in some instances it has provided opportunities for observing other viruses in wild bird populations (15, 53). In 2007, as part of a seabird health surveillance program in the Falkland Islands (Islas Malvinas), oral and cloacal swabs and serum were collected from rockhopper penguins (Eudyptes chrysocome) and environmental/fecal swab pools were collected from other seabirds.While AI virus has not yet been isolated from penguins in the sub-Antarctic and Antarctic areas, there have been two reports of serum antibodies positive to H7 and H10 from the Adélie species (11, 40). Rare isolations of APMV1, both virulent (45) and of low virulence (8), have been reported from Antarctic penguins. Sera positive for APMV1 and AMPV2 have also been reported (21, 24, 38, 40, 53). Since 1981, paramyxoviruses have been isolated from king penguins (Aptenodytes patagonicus), royal penguins (Eudyptes schlegeli), and Adélie penguins (Pygoscelis adeliae) from Antarctica and little blue penguins (Eudyptula minor) from Australia that cannot be identified as belonging to APMV1 to -9 and have not yet been classified (8, 11, 38-40). The morphology, biological and genomic characteristics, and antigenic relatedness of an APMV recently isolated from multiple penguin colonies on the Falkland Islands are reported here. Evidence that the virus belongs to a new serotype (APMV10) and a demonstration of the advantages of a whole genome system of analysis based on random sequencing followed by comparison of genetic distances are presented. Only after all APMVs are reported and classified will epidemiological information be known as to how the viruses are moving and spreading as the birds travel and interact with other avian species.  相似文献   

20.
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