Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
Arq. ciências saúde UNIPAR ; 26(2): 135-145, maio-ago. 2022.
Article in Portuguese | LILACS | ID: biblio-1372966

ABSTRACT

A meningite bacteriana é uma inflamação das leptomeninges que envolvem o Sistema Nervoso Central. Essa patologia, que possui diversos agentes etiológicos, apresenta-se na forma de síndrome, com quadro clínico grave. Entre as principais bactérias que causam a meningite, estão a Neisseria meningitis e Streptococcus pneumoniae. A transmissão ocorre através das vias aéreas por meio de gotículas, sendo a corrente sanguínea a principal rota para as bactérias chegarem à barreira hematoencefálica e, a partir dessa, até as meninges. Atualmente existem vários métodos de diagnóstico precisos, onde a cultura de líquido cefalorraquidiano (LCR) é o método padrão ouro. Ademais, a melhora na qualidade do tratamento com beta-lactâmicos e a maior possibilidade de prevenção, devido à elevação do número e da eficácia de vacinas, vem contribuindo para redução dos casos da doença e de sua gravidade. Porém, apesar desses avanços, ainda há um elevado número de mortalidades e sequelas causadas por essa síndrome.


Bacterial meningitis is an inflammation of the leptomeninges that surround the Central Nervous System. This pathology, which has several etiological agents, is presented as a syndrome with a severe clinical scenario. The main bacteria causing meningitis include Neisseria meningitis and Streptococcus pneumoniae. It can be transmitted by droplets through the airways, with the bacteria using the bloodstream as the main route to reach the blood-brain barrier, and from there to the meninges. There are currently several accurate diagnostic methods, with CSF culture being the gold standard. In addition, the improvement in the quality of beta-lactam treatment and the greater possibility of prevention due to the increased number and effectiveness of vaccines have contributed to reducing the number of cases and severity of the disease. Nevertheless, despite these advances, this syndrome still presents a high number of mortalities and sequelae.


Subject(s)
Pregnancy , Child, Preschool , Child , Aged , Cerebrospinal Fluid , Meningitis, Bacterial/diagnosis , Meningitis, Bacterial/therapy , Streptococcus pneumoniae/pathogenicity , Syndrome , Bacteria/classification , Meningitis, Bacterial/drug therapy , beta-Lactams/therapeutic use , Gram-Negative Bacteria , Gram-Positive Bacteria , Meningitis, Pneumococcal/drug therapy , Neisseria/pathogenicity
2.
PLoS Pathog ; 18(5): e1010497, 2022 05.
Article in English | MEDLINE | ID: mdl-35580146

ABSTRACT

The mechanisms used by human adapted commensal Neisseria to shape and maintain a niche in their host are poorly defined. These organisms are common members of the mucosal microbiota and share many putative host interaction factors with Neisseria meningitidis and Neisseria gonorrhoeae. Evaluating the role of these shared factors during host carriage may provide insight into bacterial mechanisms driving both commensalism and asymptomatic infection across the genus. We identified host interaction factors required for niche development and maintenance through in vivo screening of a transposon mutant library of Neisseria musculi, a commensal of wild-caught mice which persistently and asymptomatically colonizes the oral cavity and gut of CAST/EiJ and A/J mice. Approximately 500 candidate genes involved in long-term host interaction were identified. These included homologs of putative N. meningitidis and N. gonorrhoeae virulence factors which have been shown to modulate host interactions in vitro. Importantly, many candidate genes have no assigned function, illustrating how much remains to be learned about Neisseria persistence. Many genes of unknown function are conserved in human adapted Neisseria species; they are likely to provide a gateway for understanding the mechanisms allowing pathogenic and commensal Neisseria to establish and maintain a niche in their natural hosts. Validation of a subset of candidate genes confirmed a role for a polysaccharide capsule in N. musculi persistence but not colonization. Our findings highlight the potential utility of the Neisseria musculi-mouse model as a tool for studying the pathogenic Neisseria; our work represents a first step towards the identification of novel host interaction factors conserved across the genus.


Subject(s)
DNA Transposable Elements , Host Microbial Interactions , Neisseria , Animals , Carrier State/microbiology , Carrier State/physiopathology , DNA Transposable Elements/genetics , Gene Library , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Mice , Microbiota/genetics , Mucous Membrane/microbiology , Neisseria/genetics , Neisseria/pathogenicity , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/pathogenicity , Neisseria meningitidis/genetics , Neisseria meningitidis/pathogenicity , Symbiosis/genetics , Symbiosis/physiology , Virulence Factors/genetics
3.
PLoS One ; 15(11): e0242091, 2020.
Article in English | MEDLINE | ID: mdl-33166362

ABSTRACT

BACKGROUND: Perioperative oral management has been reported to be effective for preventing postoperative infectious complications. In addition, severe periodontal disease was identified as the significant risk factor for complications after gastrointestinal surgery. We investigated the bacteriological association between the periodontal pocket, stomach mucosa and drainage fluid to determine whether oral bacteria directly cause intra-abdominal infection after gastrectomy. METHODS: Patients who were scheduled to undergo surgery for gastric cancer were prospectively enrolled. We evaluated the similarity of bacterial strains in periodontal pocket, stomach mucosa and fluid from drainage tube. Gingival crevicular fluid and dental plaque were collected from the periodontal pocket and cultured to detect bacteria. Specimens from the resected stomach were collected and used for bacterial culturing. Drainage fluid from the abdominal cavity was also cultured. RESULTS: All of 52 patients were enrolled. In the periodontal pocket, α-Streptococcus spp., Neisseria sp., and Prevotella sp. were mainly detected. Bacterial cultures in the stomach mucosa were positive in 26 cases. In 20 cases (76.9%), the detected strains were the same as those in the periodontal pocket. Six patients had the postoperative intra-abdominal infection after gastrectomy, and the same bacterial strains was detected in both of drainage fluid and periodontal pocket in two patients with severe periodontal disease. CONCLUSIONS: We found the bacteriological association that same strain detected in periodontal pocket, stomach and in intra-abdominal drainage fluid after gastrectomy in patients with periodontal disease.


Subject(s)
Abdominal Abscess/epidemiology , Gastrectomy/adverse effects , Periodontitis/epidemiology , Postoperative Complications/epidemiology , Abdominal Abscess/microbiology , Aged , Female , Gastric Mucosa/microbiology , Humans , Male , Mouth Mucosa/microbiology , Neisseria/pathogenicity , Periodontitis/microbiology , Postoperative Complications/microbiology , Prevotella/pathogenicity , Streptococcus/pathogenicity
4.
Microb Genom ; 6(9)2020 09.
Article in English | MEDLINE | ID: mdl-32845827

ABSTRACT

Commensal non-pathogenic Neisseria spp. live within the human host alongside the pathogenic Neisseria meningitidis and Neisseria gonorrhoeae and due to natural competence, horizontal gene transfer within the genus is possible and has been observed. Four distinct Neisseria spp. isolates taken from the throats of two human volunteers have been assessed here using a combination of microbiological and bioinformatics techniques. Three of the isolates have been identified as Neisseria subflava biovar perflava and one as Neisseria cinerea. Specific gene clusters have been identified within these commensal isolate genome sequences that are believed to encode a Type VI Secretion System, a newly identified CRISPR system, a Type IV Secretion System unlike that in other Neisseria spp., a hemin transporter, and a haem acquisition and utilization system. This investigation is the first to investigate these systems in either the non-pathogenic or pathogenic Neisseria spp. In addition, the N. subflava biovar perflava possess previously unreported capsule loci and sequences have been identified in all four isolates that are similar to genes seen within the pathogens that are associated with virulence. These data from the four commensal isolates provide further evidence for a Neisseria spp. gene pool and highlight the presence of systems within the commensals with functions still to be explored.


Subject(s)
Bacterial Proteins/genetics , Neisseria/classification , Pharynx/microbiology , Whole Genome Sequencing/methods , Gene Transfer, Horizontal , Healthy Volunteers , High-Throughput Nucleotide Sequencing , Humans , Multigene Family , Neisseria/genetics , Neisseria/isolation & purification , Neisseria/pathogenicity , Phylogeny , Symbiosis , Type VI Secretion Systems/genetics , Virulence Factors/genetics
5.
BMC Infect Dis ; 20(1): 619, 2020 Aug 24.
Article in English | MEDLINE | ID: mdl-32831055

ABSTRACT

BACKGROUND: Neisseria macacae was discovered in the oral cavity of monkeys in 1983. In humans, it has been isolated from the upper respiratory tract of neutropenic patients. However, only two cases of N. macacae bacteremia have been reported in a 65-year-old man with infective endocarditis and a 5-month-old child with fever and petechiae. There are no reports of infections in cancer patients. Here, we present two cases of N. macacae bacteremia in cancer patients. CASE PRESENTATION: In the first case, a 42-year-old woman who underwent ovarian cancer surgery presented with duodenal invasion associated with multiple lymph node metastasis. N. macacae was isolated from her blood culture and identified using matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry (MS). In the second case, a 69-year-old woman with a long-standing history of esophagogastric junction cancer presented with fever. She had stage IVB cancer with lung, bone, and multiple lymph node metastasis. The last chemotherapy was administered 5 weeks before N. macacae was detected using MALDI-TOF MS and nitrate test negative. In both cases, transthoracic echography showed no vegetation. Antibiotics were administered for 14 and 13 days in the first and second cases, respectively. In both cases, fever alleviated on day 4 of antibiotic administration. Both patients were discharged after their conditions improved. CONCLUSIONS: This, to our knowledge, is the first report of N. macacae bacteremia in cancer patients. Both patients, mucosal damage was observed in the upper gastrointestinal tract. Therefore, exclusion diagnosis suggested that bacteremia invasion was caused by mucosal rupture in both cases. Both cases responded well to treatment with ß-lactam antibiotics and improved after 2 weeks. Modifying the treatment based on the source of the infection may shorten the treatment period. Therefore, further research on N. macacae bacteremia is necessary. Immunocompromised patients such as those with cancer are susceptible to mucosal damage by unusual bacterial species such as N. macacae despite not having contact with monkeys.


Subject(s)
Bacteremia/drug therapy , Bacteremia/microbiology , Neisseria/pathogenicity , Adult , Aged , Anti-Bacterial Agents/therapeutic use , Blood Culture/methods , Endocarditis, Bacterial/microbiology , Esophageal Neoplasms/microbiology , Esophagogastric Junction/pathology , Female , Humans , Male , Neisseria/genetics , Neisseria/isolation & purification , Ovarian Neoplasms/microbiology , RNA, Ribosomal, 16S , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
6.
Sci Rep ; 9(1): 14338, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31605019

ABSTRACT

Neisseria animaloris is considered to be a commensal of the canine and feline oral cavities. It is able to cause systemic infections in animals as well as humans, usually after a biting trauma has occurred. We recovered N. animaloris from chronically inflamed bite wounds on pectoral fins and tailstocks, from lungs and other internal organs of eight harbour porpoises. Gross and histopathological evidence suggest that fatal disseminated N. animaloris infections had occurred due to traumatic injury from grey seals. We therefore conclude that these porpoises survived a grey seal predatory attack, with the bite lesions representing the subsequent portal of entry for bacteria to infect the animals causing abscesses in multiple tissues, and eventually death. We demonstrate that forensic microbiology provides a useful tool for linking a perpetrator to its victim. Moreover, N. animaloris should be added to the list of potential zoonotic bacteria following interactions with seals, as the finding of systemic transfer to the lungs and other tissues of the harbour porpoises may suggest a potential to do likewise in humans.


Subject(s)
Forensic Genetics , Neisseria/pathogenicity , Seals, Earless/injuries , Wounds and Injuries/genetics , Animals , Animals, Wild/genetics , Animals, Wild/injuries , Animals, Wild/microbiology , Neisseria/genetics , Seals, Earless/genetics , Seals, Earless/microbiology , Wounds and Injuries/microbiology , Zoonoses/genetics , Zoonoses/microbiology
7.
Curr Opin Infect Dis ; 32(5): 490-496, 2019 10.
Article in English | MEDLINE | ID: mdl-31356239

ABSTRACT

PURPOSE OF REVIEW: Nonpathogenic commensal Neisseria are rarely considered in the clinical setting despite evidence that they can cause invasive opportunistic infections. In contrast, they may offer protection against pathogenic Neisseria, and such relationships are being actively explored in experimental studies. RECENT FINDINGS: Recent case reports are presented of invasive infection caused by nonpathogenic Neisseria in patients on novel biologic therapies. On the other hand, Neisseria lactamica, a nonpathogenic commensal, has been shown in human challenge studies to inhibit colonization by Neisseria meningitidis. Experimental mouse models have also explored the inhibitory effects of nonpathogenic Neisseria on Neisseria gonnhoreae infection. Cutting-edge advances in metagenomics and microbiomics are being used to understand the mechanisms underpinning these effects. SUMMARY: Clinicians should have increased awareness of nonpathogenic Neisseria. First, as new immunomodulating therapies become licenced, the interactions that maintain balance between commensals and their human hosts may be altered. Second, these bacteria are showing promise in their capacity to exclude pathogenic Neisseria species from their anatomical niches.


Subject(s)
Immunocompromised Host , Neisseria/immunology , Neisseria/pathogenicity , Neisseriaceae Infections/microbiology , Neisseriaceae Infections/prevention & control , Animals , Disease Models, Animal , Humans
8.
Cell Microbiol ; 21(8): e13035, 2019 08.
Article in English | MEDLINE | ID: mdl-31042331

ABSTRACT

We previously identified a Neisseria flavescens strain in the duodenum of celiac disease (CD) patients that induced immune inflammation in ex vivo duodenal mucosal explants and in CaCo-2 cells. We also found that vesicular trafficking was delayed after the CD-immunogenic P31-43 gliadin peptide-entered CaCo-2 cells and that Lactobacillus paracasei CBA L74 (L. paracasei-CBA) supernatant reduced peptide entry. In this study, we evaluated if metabolism and trafficking was altered in CD-N. flavescens-infected CaCo-2 cells and if any alteration could be mitigated by pretreating cells with L. paracasei-CBA supernatant, despite the presence of P31-43. We measured CaCo-2 bioenergetics by an extracellular flux analyser, N. flavescens and P31-43 intracellular trafficking by immunofluorescence, cellular stress by TBARS assay, and ATP by bioluminescence. We found that CD-N. flavescens colocalised more than control N. flavescens with early endocytic vesicles and more escaped autophagy thereby surviving longer in infected cells. P31-43 increased colocalisation of N. flavescens with early vesicles. Mitochondrial respiration was lower (P < .05) in CD-N. flavescens-infected cells versus not-treated CaCo-2 cells, whereas pretreatment with L. paracasei-CBA reduced CD-N. flavescens viability and improved cell bioenergetics and trafficking. In conclusion, CD-N. flavescens induces metabolic imbalance in CaCo-2 cells, and the L. paracasei-CBA probiotic could be used to correct CD-associated dysbiosis.


Subject(s)
Lacticaseibacillus paracasei/chemistry , Mitochondria/drug effects , Neisseria/drug effects , Oxidative Phosphorylation/drug effects , Probiotics/pharmacology , Adenosine Triphosphate/agonists , Adenosine Triphosphate/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/microbiology , Autophagy/drug effects , Autophagy/genetics , Caco-2 Cells , Celiac Disease/metabolism , Celiac Disease/microbiology , Celiac Disease/therapy , Culture Media, Conditioned/pharmacology , Dysbiosis/metabolism , Dysbiosis/microbiology , Dysbiosis/therapy , Gene Expression , Gliadin/antagonists & inhibitors , Gliadin/pharmacology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Lacticaseibacillus paracasei/physiology , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Neisseria/genetics , Neisseria/growth & development , Neisseria/pathogenicity , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/pharmacology , Thiobarbituric Acid Reactive Substances/metabolism , Transport Vesicles/drug effects , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
9.
Methods Mol Biol ; 1997: 403-412, 2019.
Article in English | MEDLINE | ID: mdl-31119636

ABSTRACT

We have developed a natural mouse model to study persistent colonization by commensal Neisseria. The system couples the ordinary lab mouse with Neisseria musculi (Nmus), a commensal in the oral cavity and gut of the wild mouse, Mus musculus. The pairing of Nmus with its natural reservoir circumvents host restriction barriers that have impeded previous studies of Neisseria in vivo behavior. The model allows, for the first time, for the dissection of host and neisserial determinants of asymptomatic colonization. Inoculation procedures are noninvasive and susceptibility to Nmus colonization varies with host genetic background. In colonized mice, bacterial burdens are detectable up to 1-year post inoculation, making it an ideal model for the study of persistence. As Nmus encodes several Neisseria gonorrhoeae (and Neisseria meningitidis) host interaction factors, the system can be used to query the in vivo functions of these commonly held genes and factors. Nmus also encodes many pathogenic Neisseria vaccine targets including a polysaccharide capsule, making the model potentially useful for vaccine development. The ease of genetic manipulation of Nmus enhances the feasibility of such studies.


Subject(s)
Disease Models, Animal , Gonorrhea/microbiology , Neisseria/pathogenicity , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gastrointestinal Microbiome/immunology , Gonorrhea/therapy , Humans , Mice/microbiology , Mouth Mucosa/immunology , Mouth Mucosa/microbiology , Neisseria/genetics , Neisseria/immunology , Symbiosis/immunology , Virulence Factors/genetics , Virulence Factors/metabolism
10.
J Mol Biol ; 431(16): 3010-3014, 2019 07 26.
Article in English | MEDLINE | ID: mdl-30986425

ABSTRACT

The 10 human-restricted Neisseria species all colonize mucosal surfaces, but show a spectrum of pathogenicity. The commensal Neisseria do not normally cause pathology, while the two pathogenic species, Neisseria meningitidis and Neisseria gonorrhoeae, straddle the border between commensalism and pathogenicity. Why the pathogenic Neisseria continue to mediate host damage after thousands of years of co-evolution with their human host, and why the commensal species have not acquired the ability to damage the host, if this capability provides a selective advantage, is not understood. One way the pathogenic species are different from the commensal species is by their ability to induce PMN inflammation, which is dependent on the site of colonization. I discuss how the site of colonization dictates whether copious inflammation occurs with both pathogenic species. I put forth a model that posits that an ancestor of both pathogenic species changed colonization site from the oral cavity to the genital tract of a human or humanoid and had to evolve multiple, new traits - to induce PMN inflammation and avoid adaptive immunity - to allow efficient sexual transmission. This model predicts that PMN inflammation produces the serious sequelae of gonorrhea and increases the probability that N. meningitidis might exit the oral cavity to produce systemic disease. In both cases, the pathology produced by these host-adapted species is an unintended by product of the inflammation but host damage does not provide any selective advantage for these organisms.


Subject(s)
Neisseria/immunology , Neisseria/pathogenicity , Neutrophils/metabolism , Host Specificity , Host-Pathogen Interactions , Humans , Neisseria/classification , Neisseria/physiology , Neisseria gonorrhoeae/immunology , Neisseria gonorrhoeae/pathogenicity , Neisseria meningitidis/immunology , Neisseria meningitidis/pathogenicity , Symbiosis
11.
Microbiology (Reading) ; 165(9): 917-928, 2019 09.
Article in English | MEDLINE | ID: mdl-30994440

ABSTRACT

Phase-variable DNA methyltransferases control the expression of multiple genes via epigenetic mechanisms in a wide variety of bacterial species. These systems are called phasevarions, for phase-variable regulons. Phasevarions regulate genes involved in pathogenesis, host adaptation and antibiotic resistance. Many human-adapted bacterial pathogens contain phasevarions. These include leading causes of morbidity and mortality worldwide, such as non-typeable Haemophilus influenzae, Streptococcus pneumoniae and Neisseria spp. Phase-variable methyltransferases and phasevarions have also been discovered in environmental organisms and veterinary pathogens. The existence of many different examples suggests that phasevarions have evolved multiple times as a contingency strategy in the bacterial domain, controlling phenotypes that are important in adapting to environmental change. Many of the organisms that contain phasevarions have existing or emerging drug resistance. Vaccines may therefore represent the best and most cost-effective tool to prevent disease caused by these organisms. However, many phasevarions also control the expression of current and putative vaccine candidates; variable expression of antigens could lead to immune evasion, meaning that vaccines designed using these targets become ineffective. It is therefore essential to characterize phasevarions in order to determine an organism's stably expressed antigenic repertoire, and rationally design broadly effective vaccines.


Subject(s)
Bacteria , DNA Restriction-Modification Enzymes/genetics , Epigenesis, Genetic , Methyltransferases , Bacteria/immunology , Bacteria/metabolism , Bacteria/pathogenicity , Bacterial Infections/drug therapy , Bacterial Infections/transmission , DNA Methylation , DNA Modification Methylases , DNA Restriction-Modification Enzymes/metabolism , Drug Resistance/genetics , Evolution, Molecular , Gene Expression Regulation, Bacterial , Haemophilus influenzae/genetics , Haemophilus influenzae/pathogenicity , Methyltransferases/genetics , Methyltransferases/metabolism , Mycoplasma/genetics , Mycoplasma/pathogenicity , Neisseria/genetics , Neisseria/pathogenicity , Neisseria meningitidis/genetics , Neisseria meningitidis/pathogenicity
12.
J Infect ; 78(2): 113-118, 2019 02.
Article in English | MEDLINE | ID: mdl-30408494

ABSTRACT

BACKGROUND: Non-meningococcal, non-gonococcal Neisseria spp. are typically commensal and rarely cause invasive disease. Eculizumab is a terminal complement inhibitor that increases susceptibility to meningococcal disease, but data on disease caused by typically-commensal Neisseria spp. are lacking. This series describes postmarketing reports of typically-commensal Neisseria spp. disease in patients receiving eculizumab. METHODS: We searched the FDA Adverse Event Reporting System (FAERS) and medical literature for reports of commensal Neisseria spp. disease in patients receiving eculizumab, from eculizumab U.S. approval (2007) through January 31, 2018. RESULTS: We identified seven FAERS reports (including one case also reported in the literature) of non-meningococcal, non-gonococcal Neisseria disease, including N. sicca (mucosa)/subflava (n = 2), N. cinerea (n = 2), N. sicca (mucosa) (n = 1), N. mucosa (n = 1, with concurrent alpha-hemolytic Streptococcus bacteremia), and N. flavescens (subflava) (n = 1). Four cases had sources of patient immunosuppression in addition to eculizumab. Three patients had sepsis (n = 2) or septic shock (n = 1). Five patients were bacteremic. All patients were hospitalized; the infections resolved with antibiotics. CONCLUSIONS: Our search identified seven cases of disease from typically commensal Neisseria spp. in eculizumab recipients. These findings suggest that any Neisseria spp. identified from a normally sterile site in an eculizumab recipient could represent true infection warranting prompt treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Bacteremia/chemically induced , Meningococcal Infections/chemically induced , Meningococcal Infections/microbiology , Neisseria/drug effects , Adolescent , Adult , Bacteremia/microbiology , Child , Child, Preschool , Female , Humans , Male , Meningococcal Infections/diagnosis , Neisseria/pathogenicity , Symbiosis
13.
PLoS One ; 13(5): e0196675, 2018.
Article in English | MEDLINE | ID: mdl-29763438

ABSTRACT

Pathogenic Neisseria are responsible for significantly higher levels of morbidity and mortality than their commensal relatives despite having similar genetic contents. Neisseria possess a disparate arsenal of surface determinants that facilitate host colonisation and evasion of the immune response during persistent carriage. Adaptation to rapid changes in these hostile host environments is enabled by phase variation (PV) involving high frequency, stochastic switches in expression of surface determinants. In this study, we analysed 89 complete and 79 partial genomes, from the NCBI and Neisseria PubMLST databases, representative of multiple pathogenic and commensal species of Neisseria using PhasomeIt, a new program that identifies putatively phase-variable genes and homology groups by the presence of simple sequence repeats (SSR). We detected a repertoire of 884 putative PV loci with maxima of 54 and 47 per genome in gonococcal and meningococcal isolates, respectively. Most commensal species encoded a lower number of PV genes (between 5 and 30) except N. lactamica wherein the potential for PV (36-82 loci) was higher, implying that PV is an adaptive mechanism for persistence in this species. We also characterised the repeat types and numbers in both pathogenic and commensal species. Conservation of SSR-mediated PV was frequently observed in outer membrane proteins or modifiers of outer membrane determinants. Intermittent and weak selection for evolution of SSR-mediated PV was suggested by poor conservation of tracts with novel PV genes often occurring in only one isolate. Finally, we describe core phasomes-the conserved repertoires of phase-variable genes-for each species that identify overlapping but distinctive adaptive strategies for the pathogenic and commensal members of the Neisseria genus.


Subject(s)
Adaptation, Biological/genetics , Bacterial Proteins/genetics , Genetic Variation/genetics , Neisseria/genetics , Neisseria/pathogenicity , Bacterial Outer Membrane Proteins/genetics , Genome, Bacterial/genetics , Microsatellite Repeats/genetics
14.
Infect Immun ; 86(5)2018 05.
Article in English | MEDLINE | ID: mdl-29440372

ABSTRACT

Commensals are important for the proper functioning of multicellular organisms. How a commensal establishes persistent colonization of its host is little understood. Studies of this aspect of microbe-host interactions are impeded by the absence of an animal model. We have developed a natural small animal model for identifying host and commensal determinants of colonization and of the elusive process of persistence. Our system couples a commensal bacterium of wild mice, Neisseria musculi, with the laboratory mouse. The pairing of a mouse commensal with its natural host circumvents issues of host restriction. Studies are performed in the absence of antibiotics, hormones, invasive procedures, or genetic manipulation of the host. A single dose of N. musculi, administered orally, leads to long-term colonization of the oral cavity and gut. All mice are healthy. Susceptibility to colonization is determined by host genetics and innate immunity. For N. musculi, colonization requires the type IV pilus. Reagents and powerful tools are readily available for manipulating the laboratory mouse, allowing easy dissection of host determinants controlling colonization resistance. N. musculi is genetically related to human-dwelling commensal and pathogenic Neisseria and encodes host interaction factors and vaccine antigens of pathogenic Neisseria Our system provides a natural approach for studying Neisseria-host interactions and is potentially useful for vaccine efficacy studies.


Subject(s)
Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/transmission , Host-Pathogen Interactions , Immunity, Innate , Mice/microbiology , Neisseria/pathogenicity , Symbiosis , Animals , Disease Models, Animal
15.
Infect Disord Drug Targets ; 17(1): 64-66, 2017.
Article in English | MEDLINE | ID: mdl-27889997

ABSTRACT

BACKGROUND: Neisseria are usually harmless inhabitants of otherwise asymptomatic persons' upper respiratory mucosal surfaces. METHOD: It is, therefore, expected that a disturbance in the physiology leads to nongonococcal, non-meningococcal Neisseria becoming pathogenic. RESULT: We report the case of a diabetic man who initially presented with nonspecific symptoms and was later found to have cystitis caused by N. oralis. CONCLUSION: We also review the pertinent literature and discuss available evidence on pathophysiological mechanisms of infection with such commensal bacteria.


Subject(s)
Cystitis/diagnosis , Cystitis/microbiology , Neisseria/isolation & purification , Neisseriaceae Infections/microbiology , Acute Disease , Cystitis/drug therapy , Cystitis/physiopathology , Diabetes Complications , Humans , Male , Middle Aged , Neisseria/drug effects , Neisseria/pathogenicity , Neisseriaceae Infections/diagnosis , Neisseriaceae Infections/drug therapy , Neisseriaceae Infections/physiopathology , Symbiosis
16.
Biochemistry ; 55(31): 4286-94, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27442026

ABSTRACT

Carcino-embryonic antigen-like cellular adhesion molecules (CEACAMs), members of the immunoglobulin superfamily, are responsible for cell-cell interactions and cellular signaling events. Extracellular interactions with CEACAMs have the potential to induce phagocytosis, as is the case with pathogenic Neisseria bacteria. Pathogenic Neisseria species express opacity-associated (Opa) proteins, which interact with a subset of CEACAMs on human cells, and initiate the engulfment of the bacterium. We demonstrate that recombinant Opa proteins reconstituted into liposomes retain the ability to recognize and interact with CEACAMs in vitro but do not maintain receptor specificity compared to that of Opa proteins natively expressed by Neisseria gonorrhoeae. We report that two Opa proteins interact with CEACAMs with nanomolar affinity, and we hypothesize that this high affinity is necessary to compete with the native CEACAM homo- and heterotypic interactions in the host. Understanding the mechanisms of Opa protein-receptor recognition and engulfment enhances our understanding of Neisserial pathogenesis. Additionally, these mechanisms provide insight into how human cells that are typically nonphagocytic can utilize CEACAM receptors to internalize exogenous matter, with implications for the targeted delivery of therapeutics and development of imaging agents.


Subject(s)
Antigens, CD/metabolism , Bacterial Outer Membrane Proteins/metabolism , Carcinoembryonic Antigen/metabolism , Cell Adhesion Molecules/metabolism , Neisseria/metabolism , Antigens, CD/chemistry , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Carcinoembryonic Antigen/chemistry , Cell Adhesion Molecules/chemistry , Host-Pathogen Interactions , Humans , Immunoglobulin Domains , Liposomes , Models, Molecular , Neisseria/genetics , Neisseria/pathogenicity , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/metabolism , Neisseria gonorrhoeae/pathogenicity , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
18.
J Clin Microbiol ; 54(8): 1949-55, 2016 08.
Article in English | MEDLINE | ID: mdl-27098959

ABSTRACT

Rapid low-cost whole-genome sequencing (WGS) is revolutionizing microbiology; however, complementary advances in accessible, reproducible, and rapid analysis techniques are required to realize the potential of these data. Here, investigations of the genus Neisseria illustrated the gene-by-gene conceptual approach to the organization and analysis of WGS data. Using the gene and its link to phenotype as a starting point, the BIGSdb database, which powers the PubMLST databases, enables the assembly of large open-access collections of annotated genomes that provide insight into the evolution of the Neisseria, the epidemiology of meningococcal and gonococcal disease, and mechanisms of Neisseria pathogenicity.


Subject(s)
Genomics/methods , Gonorrhea/microbiology , Meningococcal Infections/microbiology , Neisseria/genetics , Computational Biology/methods , Gonorrhea/epidemiology , Gonorrhea/pathology , High-Throughput Nucleotide Sequencing/methods , Humans , Meningococcal Infections/epidemiology , Meningococcal Infections/pathology , Neisseria/classification , Neisseria/pathogenicity
19.
Proc Natl Acad Sci U S A ; 112(51): E7128-37, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26621750

ABSTRACT

This paper reviews the developments that have occurred in the field of human genetics of infectious diseases from the second half of the 20th century onward. In particular, it stresses and explains the importance of the recently described monogenic inborn errors of immunity underlying resistance or susceptibility to specific infections. The monogenic component of the genetic theory provides a plausible explanation for the occurrence of severe infectious diseases during primary infection. Over the last 20 y, increasing numbers of life-threatening infectious diseases striking otherwise healthy children, adolescents, and even young adults have been attributed to single-gene inborn errors of immunity. These studies were inspired by seminal but neglected findings in plant and animal infections. Infectious diseases typically manifest as sporadic traits because human genotypes often display incomplete penetrance (most genetically predisposed individuals remain healthy) and variable expressivity (different infections can be allelic at the same locus). Infectious diseases of childhood, once thought to be archetypal environmental diseases, actually may be among the most genetically determined conditions of mankind. This nascent and testable notion has interesting medical and biological implications.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/immunology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Infections/genetics , Infections/immunology , Adolescent , Candidiasis, Chronic Mucocutaneous/genetics , Candidiasis, Chronic Mucocutaneous/immunology , Child , Complement System Proteins/genetics , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/immunology , Epidermodysplasia Verruciformis/genetics , Epidermodysplasia Verruciformis/immunology , Genetic Predisposition to Disease , Humans , Influenza, Human/genetics , Influenza, Human/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Malaria/genetics , Malaria/immunology , Models, Genetic , Models, Immunological , Mycobacterium Infections/genetics , Mycobacterium Infections/immunology , Neisseria/immunology , Neisseria/pathogenicity , Pneumococcal Infections/genetics , Pneumococcal Infections/immunology , Tinea/genetics , Tinea/immunology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...