Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Transl Med ; 22(1): 269, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38475767

RESUMO

BACKGROUND: Chemotherapy is a primary treatment for cancer, but its efficacy is often limited by cancer-associated bacteria (CAB) that impair tumor suppressor functions. Our previous research found that Mycoplasma fermentans DnaK, a chaperone protein, impairs p53 activities, which are essential for most anti-cancer chemotherapeutic responses. METHODS: To investigate the role of DnaK in chemotherapy, we treated cancer cell lines with M. fermentans DnaK and then with commonly used p53-dependent anti-cancer drugs (cisplatin and 5FU). We evaluated the cells' survival in the presence or absence of a DnaK-binding peptide (ARV-1502). We also validated our findings using primary tumor cells from a novel DnaK knock-in mouse model. To provide a broader context for the clinical significance of these findings, we investigated human primary cancer sequencing datasets from The Cancer Genome Atlas (TCGA). We identified F. nucleatum as a CAB carrying DnaK with an amino acid composition highly similar to M. fermentans DnaK. Therefore, we investigated the effect of F. nucleatum DnaK on the anti-cancer activity of cisplatin and 5FU. RESULTS: Our results show that both M. fermentans and F. nucleatum DnaKs reduce the effectiveness of cisplatin and 5FU. However, the use of ARV-1502 effectively restored the drugs' anti-cancer efficacy. CONCLUSIONS: Our findings offer a practical framework for designing and implementing novel personalized anti-cancer strategies by targeting specific bacterial DnaKs in patients with poor response to chemotherapy, underscoring the potential for microbiome-based personalized cancer therapies.


Assuntos
Antineoplásicos , Neoplasias , Animais , Camundongos , Humanos , Cisplatino , Proteína Supressora de Tumor p53 , Fluoruracila , Bactérias
2.
Front Immunol ; 15: 1334762, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38533492

RESUMO

Salmonella enterica serovar Typhi (S. Typhi), a human-restricted pathogen, invades the host through the gut to cause typhoid fever. Recent calculations of the typhoid fever burden estimated that more than 10 million new typhoid fever cases occur in low and middle-income countries, resulting in 65,400-187,700 deaths yearly. Interestingly, if not antibiotic-treated, upon the resolution of acute disease, 1%-5% of patients become asymptomatic chronic carriers. Chronically infected hosts are not only critical reservoirs of infection that transmit the disease to naive individuals but are also predisposed to developing gallbladder carcinoma. Nevertheless, the molecular mechanisms involved in the early interactions between gallbladder epithelial cells and S. Typhi remain largely unknown. Based on our previous studies showing that closely related S. Typhi strains elicit distinct innate immune responses, we hypothesized that host molecular pathways activated by S. Typhi strains derived from acutely and chronically infected patients would differ. To test this hypothesis, we used a novel human organoid-derived polarized gallbladder monolayer model, and S. Typhi strains derived from acutely and chronically infected patients. We found that S. Typhi strains derived from acutely and chronically infected patients differentially regulate host mitogen-activated protein kinase (MAPK) and S6 transcription factors. These variations might be attributed to differential cytokine signaling, predominantly via TNF-α and IL-6 production and appear to be influenced by the duration the isolate was subjected to selective pressures in the gallbladder. These findings represent a significant leap in understanding the complexities behind chronic S. Typhi infections in the gallbladder and may uncover potential intervention targets.


Assuntos
Salmonella typhi , Febre Tifoide , Humanos , Vesícula Biliar/patologia , Infecção Persistente , Imunidade
3.
JCI Insight ; 8(14)2023 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-37318849

RESUMO

Influenza A virus (IAV) infection is commonly complicated by secondary bacterial infections that lead to increased morbidity and mortality. Our recent work demonstrates that IAV disrupts airway homeostasis, leading to airway pathophysiology resembling cystic fibrosis disease through diminished cystic fibrosis transmembrane conductance regulator (CFTR) function. Here, we use human airway organotypic cultures to investigate how IAV alters the airway microenvironment to increase susceptibility to secondary infection with Streptococcus pneumoniae (Spn). We observed that IAV-induced CFTR dysfunction and airway surface liquid acidification is central to increasing susceptibility to Spn. Additionally, we observed that IAV induced profound transcriptional changes in the airway epithelium and proteomic changes in the airway surface liquid in both CFTR-dependent and -independent manners. These changes correspond to multiple diminished host defense pathways and altered airway epithelial function. Collectively, these findings highlight both the importance of CFTR function during infectious challenge and demonstrate a central role for the lung epithelium in secondary bacterial infections following IAV.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística , Influenza Humana , Humanos , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Streptococcus pneumoniae , Influenza Humana/complicações , Influenza Humana/metabolismo , Proteômica , Pulmão/metabolismo
4.
Virulence ; 14(1): 2215602, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37221835

RESUMO

BACKGROUND: Mycobacterium abscessus subspecies massiliense (M. massiliense) is increasingly recognized as an emerging bacterial pathogen, particularly in cystic fibrosis (CF) patients and CF centres' respiratory outbreaks. We characterized genomic and phenotypic changes in 15 serial isolates from two CF patients (1S and 2B) with chronic pulmonary M. massiliense infection leading to death, as well as four isolates from a CF centre outbreak in which patient 2B was the index case. RESULTS: Comparative genomic analysis revealed the mutations affecting growth rate, metabolism, transport, lipids (loss of glycopeptidolipids), antibiotic susceptibility (macrolides and aminoglycosides resistance), and virulence factors. Mutations in 23S rRNA, mmpL4, porin locus and tetR genes occurred in isolates from both CF patients. Interestingly, we identified two different spontaneous mutation events at the mycobacterial porin locus: a fusion of two tandem porin paralogs in patient 1S and a partial deletion of the first porin paralog in patient 2B. These genomic changes correlated with reduced porin protein expression, diminished 14C-glucose uptake, slower bacterial growth rates, and enhanced TNF-α induction in mycobacteria-infected THP-1 human cells. Porin gene complementation of porin mutants partly restored 14C-glucose uptake, growth rate and TNF-α levels to those of intact porin strains. CONCLUSIONS: We hypothesize that specific mutations accumulated and maintained over time in M. massiliense, including mutations shared among transmissible strains, collectively lead to more virulent, host adapted lineages in CF patients and other susceptible hosts.


Assuntos
Fibrose Cística , Mycobacterium abscessus , Mycobacterium , Humanos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Fibrose Cística/microbiologia , Genômica , Glucose , Pulmão , Mutação , Mycobacterium/genética , Mycobacterium abscessus/genética , Fator de Necrose Tumoral alfa/genética , Porinas/genética , Porinas/metabolismo
5.
Cell Rep ; 42(2): 112120, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36774550

RESUMO

Intestinal colonization of the oral bacterium Haemophilus parainfluenzae has been associated with Crohn's disease (CD) severity and progression. This study examines the role of periodontal disease (PD) as a modifier for colonization of H. parainfluenzae in patients with CD and explores the mechanisms behind H. parainfluenzae-mediated intestinal inflammation. Fifty subjects with and without CD were evaluated for the presence of PD, and their oral and fecal microbiomes were characterized. PD is associated with increased levels of H. parainfluenzae strains in subjects with CD. Oral inoculation of H. parainfluenzae elicits strain-dependent intestinal inflammation in murine models of inflammatory bowel disease, which is associated with increased intestinal interferon-γ (IFN-γ)+ CD4+ T cells and disruption of the host hypusination pathway. In summary, this study establishes a strain-specific pathogenic role of H. parainfluenzae in intestinal inflammation and highlights the potential effect of PD on intestinal colonization by pathogenic H. parainfluenzae strains in patients with CD.


Assuntos
Doença de Crohn , Doenças Periodontais , Humanos , Animais , Camundongos , Haemophilus parainfluenzae , Doença de Crohn/complicações , Doença de Crohn/metabolismo , Inflamação
6.
Int J Mol Sci ; 22(8)2021 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-33918708

RESUMO

Several species of mycoplasmas, including Mycoplasma fermentans, are associated with certain human cancers. We previously isolated and characterized in our laboratory a strain of human mycoplasma M. fermentans subtype incognitus (MF-I1) able to induce lymphoma in a Severe Combined Immuno-Deficient (SCID) mouse model, and we demonstrated that its chaperone protein, DnaK, binds and reduces functions of human poly-ADP ribose polymerase-1 (PARP1) and ubiquitin carboxyl-terminal hydrolase protein-10 (USP10), which are required for efficient DNA repair and proper p53 activities, respectively. We also showed that other bacteria associated with human cancers (including Mycoplasmapneumoniae, Helicobacterpylori, Fusobacteriumnucleatum, Chlamydiathrachomatis, and Chlamydia pneumoniae) have closely related DnaK proteins, indicating a potential common mechanism of cellular transformation. Here, we quantify dnaK mRNA copy number by RT-qPCR analysis in different cellular compartments following intracellular MF-I1 infection of HCT116 human colon carcinoma cells. DnaK protein expression in infected cells was also detected and quantified by Western blot. The amount of viable intracellular mycoplasma reached a steady state after an initial phase of growth and was mostly localized in the cytoplasm of the invaded cells, while we detected a logarithmically increased number of viable extracellular bacteria. Our data indicate that, after invasion, MF-I1 is able to establish a chronic intracellular infection. Extracellular replication was more efficient while MF-I1 cultured in cell-free axenic medium showed a markedly reduced growth rate. We also identified modifications of important regulatory regions and heterogeneous lengths of dnaK mRNA transcripts isolated from intracellular and extracellular MF-I1. Both characteristics were less evident in dnaK mRNA transcripts isolated from MF-I1 grown in cell-free axenic media. Taken together, our data indicate that MF-I1, after establishing a chronic infection in eukaryotic cells, accumulates different forms of dnaK with efficient RNA turnover.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Proteínas de Choque Térmico HSP70/genética , Mycoplasma fermentans/genética , Células Cultivadas , Células HCT116 , Humanos , Mutação , Infecções por Mycoplasma/microbiologia
7.
Proc Natl Acad Sci U S A ; 115(51): E12005-E12014, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30509983

RESUMO

We isolated a strain of human mycoplasma that promotes lymphomagenesis in SCID mice, pointing to a p53-dependent mechanism similar to lymphomagenesis in uninfected p53-/- SCID mice. Additionally, mycoplasma infection in vitro reduces p53 activity. Immunoprecipitation of p53 in mycoplasma-infected cells identified several mycoplasma proteins, including DnaK, a member of the Hsp70 chaperon family. We focused on DnaK because of its ability to interact with proteins. We demonstrate that mycoplasma DnaK interacts with and reduces the activities of human proteins involved in critical cellular pathways, including DNA-PK and PARP1, which are required for efficient DNA repair, and binds to USP10 (a key p53 regulator), impairing p53-dependent anticancer functions. This also reduced the efficacy of anticancer drugs that depend on p53 to exert their effect. mycoplasma was detected early in the infected mice, but only low copy numbers of mycoplasma DnaK DNA sequences were found in some primary and secondary tumors, pointing toward a hit-and-run/hide mechanism of transformation. Uninfected bystander cells took up exogenous DnaK, suggesting a possible paracrine function in promoting malignant transformation, over and above cells infected with the mycoplasma. Phylogenetic amino acid analysis shows that other bacteria associated with human cancers have similar DnaKs, consistent with a common mechanism of cellular transformation mediated through disruption of DNA-repair mechanisms, as well as p53 dysregulation, that also results in cancer-drug resistance. This suggests that the oncogenic properties of certain bacteria are DnaK-mediated.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Bactérias/genética , Carcinogênese/genética , Transformação Celular Neoplásica/genética , Chaperonas Moleculares/genética , Mycoplasma/genética , Adenosina Trifosfatases/classificação , Animais , Antineoplásicos/uso terapêutico , Proteínas de Bactérias/classificação , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Reparo do DNA , DNA Bacteriano/genética , Proteína Quinase Ativada por DNA/metabolismo , Modelos Animais de Doenças , Genes Bacterianos/genética , Células HCT116 , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Linfoma/genética , Linfoma/microbiologia , Linfoma/patologia , Camundongos , Camundongos SCID , Chaperonas Moleculares/classificação , Mycoplasma/patogenicidade , Infecções por Mycoplasma/microbiologia , Mycoplasma fermentans/genética , Mycoplasma fermentans/patogenicidade , Oncogenes , Filogenia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Análise de Sequência , Análise de Sequência de Proteína , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina Tiolesterase/metabolismo
8.
PLoS Pathog ; 13(8): e1006582, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28841717

RESUMO

For over 130 years, invasive pneumococcal disease has been associated with the presence of extracellular planktonic pneumococci, i.e. diplococci or short chains in affected tissues. Herein, we show that Streptococcus pneumoniae that invade the myocardium instead replicate within cellular vesicles and transition into non-purulent biofilms. Pneumococci within mature cardiac microlesions exhibited salient biofilm features including intrinsic resistance to antibiotic killing and the presence of an extracellular matrix. Dual RNA-seq and subsequent principal component analyses of heart- and blood-isolated pneumococci confirmed the biofilm phenotype in vivo and revealed stark anatomical site-specific differences in virulence gene expression; the latter having major implications on future vaccine antigen selection. Our RNA-seq approach also identified three genomic islands as exclusively expressed in vivo. Deletion of one such island, Region of Diversity 12, resulted in a biofilm-deficient and highly inflammogenic phenotype within the heart; indicating a possible link between the biofilm phenotype and a dampened host-response. We subsequently determined that biofilm pneumococci released greater amounts of the toxin pneumolysin than did planktonic or RD12 deficient pneumococci. This allowed heart-invaded wildtype pneumococci to kill resident cardiac macrophages and subsequently subvert cytokine/chemokine production and neutrophil infiltration into the myocardium. This is the first report for pneumococcal biofilm formation in an invasive disease setting. We show that biofilm pneumococci actively suppress the host response through pneumolysin-mediated immune cell killing. As such, our findings contradict the emerging notion that biofilm pneumococci are passively immunoquiescent.


Assuntos
Biofilmes , Macrófagos/imunologia , Miocardite/imunologia , Miocardite/microbiologia , Infecções Pneumocócicas/imunologia , Transcriptoma , Animais , Western Blotting , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Infecções Pneumocócicas/genética , Análise de Componente Principal , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/imunologia , Virulência/genética , Virulência/imunologia
9.
Microbes Infect ; 19(6): 323-333, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28408270

RESUMO

Pneumococcal serine-rich repeat protein (PsrP) is a glycoprotein that mediates Streptococcus pneumoniae attachment to lung cells and promotes biofilm formation. Herein, we investigated the transcriptional organization of psrP-secY2A2, the 37-kbp pathogenicity island encoding PsrP and its accessory genes. PCR amplification of cDNA and RNA-seq analysis found psrP-secY2A2 to be minimally composed of three operons: psrP-glyA, glyB, and glyC-asp5. Transcription of all three operons was greatest during biofilm growth and immunoblot analyses confirmed increased PsrP production by biofilm pneumococci. Using gas chromatography-mass spectrometry we identified monomeric N-acetylglucosamine as the primary glycoconjugate present on a recombinant intracellular version of PsrP, i.e. PsrP1-734. This finding was validated by immunoblot using lectins with known carbohydrate specificities. We subsequently deleted gtfA and gtfB, the GTFs thought to be responsible for addition of O-linked N-acetylglucosamine, and tested for PsrP and its associated virulence properties. These deletions negatively affected our ability to detect PsrP1-734 in bacterial whole cell lysates. Moreover, S. pneumoniae mutants lacking these genes pheno-copied the psrP mutant and were attenuated for: biofilm formation, adhesion to lung epithelial cells, and pneumonia in mice. Our studies identify the transcriptional organization of psrP-secY2A2 and show the indispensable role of GtfA and GtfB on PsrP-mediated pneumococcal virulence.


Assuntos
Proteínas de Bactérias/genética , Deleção de Genes , Streptococcus pneumoniae/genética , Fatores de Virulência/genética , Células A549 , Animais , Aderência Bacteriana , Biofilmes/crescimento & desenvolvimento , Adesão Celular , Células Epiteliais/microbiologia , Feminino , Genes Bacterianos , Ilhas Genômicas/genética , Humanos , Pulmão/citologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Óperon/genética , Streptococcus pneumoniae/patogenicidade
10.
Infect Immun ; 84(10): 2922-32, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27481242

RESUMO

Streptococcus pneumoniae is an opportunistic pathogen that colonizes the nasopharynx. Herein we show that carbon availability is distinct between the nasopharynx and bloodstream of adult humans: glucose is absent from the nasopharynx, whereas galactose is abundant. We demonstrate that pneumococcal neuraminidase A (NanA), which cleaves terminal sialic acid residues from host glycoproteins, exposed galactose on the surface of septal epithelial cells, thereby increasing its availability during colonization. We observed that S. pneumoniae mutants deficient in NanA and ß-galactosidase A (BgaA) failed to form biofilms in vivo despite normal biofilm-forming abilities in vitro Subsequently, we observed that glucose, sucrose, and fructose were inhibitory for biofilm formation, whereas galactose, lactose, and low concentrations of sialic acid were permissive. Together these findings suggested that the genes involved in biofilm formation were under some form of carbon catabolite repression (CCR), a regulatory network in which genes involved in the uptake and metabolism of less-preferred sugars are silenced during growth with preferred sugars. Supporting this notion, we observed that a mutant deficient in pyruvate oxidase, which converts pyruvate to acetyl-phosphate under non-CCR-inducing growth conditions, was unable to form biofilms. Subsequent comparative transcriptome sequencing (RNA-seq) analyses of planktonic and biofilm-grown pneumococci showed that metabolic pathways involving the conversion of pyruvate to acetyl-phosphate and subsequently leading to fatty acid biosynthesis were consistently upregulated during diverse biofilm growth conditions. We conclude that carbon availability in the nasopharynx impacts pneumococcal biofilm formation in vivo Additionally, biofilm formation involves metabolic pathways not previously appreciated to play an important role.


Assuntos
Biofilmes/crescimento & desenvolvimento , Metabolismo dos Carboidratos/fisiologia , Carboidratos/farmacologia , Galactose/farmacocinética , Neuraminidase/fisiologia , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/fisiologia , Análise de Variância , Animais , Biofilmes/efeitos dos fármacos , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Galactose/metabolismo , Galactose/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Ácido N-Acetilneuramínico/metabolismo , Líquido da Lavagem Nasal/química , Septo Nasal/metabolismo , Septo Nasal/microbiologia , Nasofaringe/metabolismo , Nasofaringe/microbiologia , Neuraminidase/metabolismo , Infecções Pneumocócicas/metabolismo , Streptococcus pneumoniae/efeitos dos fármacos , beta-Galactosidase/deficiência , beta-Galactosidase/metabolismo
11.
J Clin Microbiol ; 53(11): 3438-47, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26292297

RESUMO

The smooth-to-rough colony morphology shift in Mycobacterium abscessus has been implicated in loss of glycopeptidolipid (GPL), increased pathogenicity, and clinical decline in cystic fibrosis (CF) patients. However, the evolutionary phenotypic and genetic changes remain obscure. Serial isolates from nine non-CF patients with persistent M. abscessus infection were characterized by colony morphology, lipid profile via thin-layer chromatography and matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS), sequencing of eight genes in the GPL locus, and expression level of fadD23, a key gene involved in the biosynthesis of complex lipids. All 50 isolates were typed as M. abscessus subspecies abscessus and were clonally related within each patient. Rough isolates, all lacking GPL, predominated at later disease stages, some showing variation within rough morphology. While most (77%) rough isolates harbored detrimental mutations in mps1 and mps2, 13% displayed previously unreported mutations in mmpL4a and mmpS4, the latter yielding a putative GPL precursor. Two isolates showed no deleterious mutations in any of the eight genes sequenced. Mixed populations harboring different GPL locus mutations were detected in 5 patients, demonstrating clonal diversification, which was likely overlooked by conventional acid-fast bacillus (AFB) culture methods. Our work highlights applications of MALDI-TOF MS beyond identification, focusing on mycobacterial lipids relevant in virulence and adaptation. Later isolates displayed accumulation of triacylglycerol and reduced expression of fadD23, sometimes preceding rough colony onset. Our results indicate that clonal diversification and a shift in lipid metabolism, including the loss of GPL, occur during chronic lung infection with M. abscessus. GPL loss alone may not account for all traits associated with rough morphology.


Assuntos
Proteínas de Bactérias/genética , Ligases/genética , Metabolismo dos Lipídeos/genética , Infecções por Mycobacterium não Tuberculosas/microbiologia , Micobactérias não Tuberculosas/isolamento & purificação , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Bronquiectasia/microbiologia , Fibrose Cística/microbiologia , DNA Bacteriano/genética , Feminino , Dosagem de Genes/genética , Genoma Bacteriano/genética , Humanos , Lipídeos/genética , Masculino , Pessoa de Meia-Idade , Tipagem de Sequências Multilocus , Micobactérias não Tuberculosas/classificação , Micobactérias não Tuberculosas/genética , RNA Ribossômico 16S/genética , Análise de Sequência de DNA , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
12.
Am J Respir Crit Care Med ; 192(5): 618-28, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26038974

RESUMO

RATIONALE: The clinical features of patients infected with pulmonary nontuberculous mycobacteria (PNTM) are well described, but the genetic components of infection susceptibility are not. OBJECTIVES: To examine genetic variants in patients with PNTM, their unaffected family members, and a control group. METHODS: Whole-exome sequencing was done on 69 white patients with PNTM and 18 of their white unaffected family members. We performed a candidate gene analysis using immune, cystic fibrosis transmembrance conductance regulator (CFTR), cilia, and connective tissue gene sets. The numbers of patients, family members, and control subjects with variants in each category were compared, as was the average number of variants per person. MEASUREMENTS AND MAIN RESULTS: A significantly higher number of patients with PNTM than the other subjects had low-frequency, protein-affecting variants in immune, CFTR, cilia, and connective tissue categories (35, 26, 90, and 90%, respectively). Patients with PNTM also had significantly more cilia and connective tissue variants per person than did control subjects (2.47 and 2.55 compared with 1.38 and 1.40, respectively; P = 1.4 × 10(-6) and P = 2.7 × 10(-8), respectively). Patients with PNTM had an average of 5.26 variants across all categories (1.98 in control subjects; P = 2.8 × 10(-17)), and they were more likely than control subjects to have variants in multiple categories. We observed similar results for family members without PNTM infection, with the exception of the immune category. CONCLUSIONS: Patients with PNTM have more low-frequency, protein-affecting variants in immune, CFTR, cilia, and connective tissue genes than their unaffected family members and control subjects. We propose that PNTM infection is a multigenic disease in which combinations of variants across gene categories, plus environmental exposures, increase susceptibility to the infection.


Assuntos
Cílios/genética , Tecido Conjuntivo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Imunidade/genética , Infecções por Mycobacterium não Tuberculosas/genética , Tuberculose Pulmonar/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Causalidade , Estudos de Coortes , Exoma , Família , Feminino , Predisposição Genética para Doença , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Componente Principal , Análise de Sequência de DNA
13.
Emerg Infect Dis ; 20(3): 364-71, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24565502

RESUMO

Three recently sequenced strains isolated from patients during an outbreak of Mycobacterium abscessus subsp. massiliense infections at a cystic fibrosis center in the United States were compared with 6 strains from an outbreak at a cystic fibrosis center in the United Kingdom and worldwide strains. Strains from the 2 cystic fibrosis outbreaks showed high-level relatedness with each other and major-level relatedness with strains that caused soft tissue infections during an epidemic in Brazil. We identified unique single-nucleotide polymorphisms in cystic fibrosis and soft tissue outbreak strains, separate single-nucleotide polymorphisms only in cystic fibrosis outbreak strains, and unique genomic traits for each subset of isolates. Our findings highlight the necessity of identifying M. abscessus to the subspecies level and screening all cystic fibrosis isolates for relatedness to these outbreak strains. We propose 2 diagnostic strategies that use partial sequencing of rpoB and secA1 genes and a multilocus sequence typing protocol.


Assuntos
Surtos de Doenças , Infecções por Mycobacterium/epidemiologia , Mycobacterium/isolamento & purificação , Brasil , Fibrose Cística/complicações , Genoma Bacteriano , Humanos , Tipagem de Sequências Multilocus , Mycobacterium/classificação , Mycobacterium/genética , Infecções por Mycobacterium/complicações , Infecções por Mycobacterium/diagnóstico , Infecções por Mycobacterium/microbiologia , Filogenia , Polimorfismo de Nucleotídeo Único , Reino Unido , Estados Unidos
14.
BMC Genomics ; 14: 378, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23742656

RESUMO

BACKGROUND: Viral upper respiratory tract infections are associated with increased colonization by Streptococcus pneumoniae but the mechanisms underlying this relationship are unclear. The objective of this study is to describe a comprehensive picture of the cellular interaction between the adhering bacteria and host cells in the presence or absence of a viral co-infection. RESULTS: Gene expression profiles of Detroit-562 pharyngeal cells, which were either mock-infected or infected with human respiratory syncytial virus (RSV) or human parainfluenza virus 3 (HPIV3), were analyzed using human microarrays. Transcription response of S. pneumoniae strain TIGR4 (serotype 4) in the presence of either mock- or viral-infected cells was analyzed by pneumococcal microarray. Significantly regulated genes were identified by both significance analysis of microarray (SAM) and a ≥ 2-fold change ratio cut-off. The adherence of S. pneumoniae to human pharyngeal cells was significantly augmented in the presence of RSV or HPIV3 infection. Global gene expression profiling of the host cells during infection with RSV or HPIV3 revealed increased transcription of carcinoembryonic antigen-related cell adhesion molecules (CEACAM1), CD47, fibronectin, interferon-stimulated genes and many other host cell adhesion molecules. Pneumococci increased transcription of several genes involved in adhesive functions (psaA, pilus islet), choline uptake and incorporation (lic operon), as well as transport and binding. CONCLUSIONS: We have identified a core transcriptome that represents the basic machinery required for adherence of pneumococci to D562 cells infected or not infected with a virus. These bacterial genes and cell adhesion molecules can potentially be used to control pneumococcal adherence occurring secondary to a viral infection.


Assuntos
Adaptação Fisiológica/genética , Vírus da Parainfluenza 3 Humana/fisiologia , Faringe/citologia , Vírus Sinciciais Respiratórios/fisiologia , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/fisiologia , Transcrição Gênica , Aderência Bacteriana/genética , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Regulação Bacteriana da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Faringe/metabolismo , Faringe/microbiologia , Faringe/virologia
15.
BMC Genomics ; 14: 383, 2013 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-23758733

RESUMO

BACKGROUND: Streptococcus pneumoniae is a leading cause of childhood morbidity and mortality worldwide, despite the availability of effective pneumococcal vaccines. Understanding the molecular interactions between the bacterium and the host will contribute to the control and prevention of pneumococcal disease. RESULTS: We used a combination of adherence assays, mutagenesis and functional genomics to identify novel factors involved in adherence. By contrasting these processes in two pneumococcal strains, TIGR4 and G54, we showed that adherence and invasion capacities vary markedly by strain. Electron microscopy showed more adherent bacteria in association with membranous pseudopodia in the TIGR4 strain. Operons for cell wall phosphorylcholine incorporation (lic), manganese transport (psa) and phosphate utilization (phn) were up-regulated in both strains on exposure to epithelial cells. Pneumolysin, pili, stress protection genes (adhC-czcD) and genes of the type II fatty acid synthesis pathway were highly expressed in the naturally more invasive strain, TIGR4. Deletion mutagenesis of five gene regions identified as regulated in this study revealed attenuation in adherence. Most strikingly, ∆SP_1922 which was predicted to contain a B-cell epitope and revealed significant attenuation in adherence, appeared to be expressed as a part of an operon that includes the gene encoding the cytoplasmic pore-forming toxin and vaccine candidate, pneumolysin. CONCLUSION: This work identifies a list of novel potential pneumococcal adherence determinants.


Assuntos
Perfilação da Expressão Gênica , Genômica , Faringe/citologia , Fenótipo , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/fisiologia , Transcrição Gênica/genética , Aderência Bacteriana/genética , Linhagem Celular Tumoral , Técnicas de Inativação de Genes , Genes Bacterianos/genética , Humanos , Mutagênese , Análise de Sequência com Séries de Oligonucleotídeos , Faringe/microbiologia , Deleção de Sequência , Especificidade da Espécie
16.
PLoS One ; 3(7): e2683, 2008 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-18628950

RESUMO

Streptococcus pneumoniae is a member of the Mitis group of streptococci which, according to 16S rRNA-sequence based phylogenetic reconstruction, includes 12 species. While other species of this group are considered prototypes of commensal bacteria, S. pneumoniae is among the most frequent microbial killers worldwide. Population genetic analysis of 118 strains, supported by demonstration of a distinct cell wall carbohydrate structure and competence pheromone sequence signature, shows that S. pneumoniae is one of several hundred evolutionary lineages forming a cluster separate from Streptococcus oralis and Streptococcus infantis. The remaining lineages of this distinct cluster are commensals previously collectively referred to as Streptococcus mitis and each represent separate species by traditional taxonomic standard. Virulence genes including the operon for capsule polysaccharide synthesis and genes encoding IgA1 protease, pneumolysin, and autolysin were randomly distributed among S. mitis lineages. Estimates of the evolutionary age of the lineages, the identical location of remnants of virulence genes in the genomes of commensal strains, the pattern of genome reductions, and the proportion of unique genes and their origin support the model that the entire cluster of S. pneumoniae, S. pseudopneumoniae, and S. mitis lineages evolved from pneumococcus-like bacteria presumably pathogenic to the common immediate ancestor of hominoids. During their adaptation to a commensal life style, most of the lineages gradually lost the majority of genes determining virulence and became genetically distinct due to sexual isolation in their respective hosts.


Assuntos
Streptococcus pneumoniae/genética , Streptococcus pneumoniae/fisiologia , Algoritmos , Proteínas de Bactérias/genética , Linhagem da Célula , Análise por Conglomerados , DNA/metabolismo , Evolução Molecular , Genética Populacional , Genoma Bacteriano , Modelos Genéticos , Peptídeos/química , Filogenia , Polimorfismo Genético , RNA Ribossômico 16S/química , Recombinação Genética
17.
Genome Res ; 16(11): 1334-8, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17065606

RESUMO

Comparative studies require knowledge of the evolutionary relationships between taxa. However, neither morphological nor paleontological data have been able to unequivocally resolve the major groups of holometabolous insects so far. Here, we utilize emerging genome projects to assemble and analyze a data set of 185 nuclear genes, resulting in a fully resolved phylogeny of the major insect model species. Contrary to the most widely accepted phylogenetic hypothesis, bees and wasps (Hymenoptera) are basal to the other major holometabolous orders, beetles (Coleoptera), moths (Lepidoptera), and flies (Diptera). We validate our results by meticulous examination of potential confounding factors. Phylogenomic approaches are thus able to resolve long-standing questions about the phylogeny of insects.


Assuntos
Abelhas/genética , Insetos/genética , Vespas/genética , Animais , Abelhas/classificação , Evolução Biológica , Genoma de Inseto , Genômica , Proteínas de Insetos/genética , Insetos/classificação , Modelos Genéticos , Filogenia , Proteômica , Especificidade da Espécie , Vespas/classificação
19.
PLoS Biol ; 2(10): e303, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15383840

RESUMO

Methanotrophs are ubiquitous bacteria that can use the greenhouse gas methane as a sole carbon and energy source for growth, thus playing major roles in global carbon cycles, and in particular, substantially reducing emissions of biologically generated methane to the atmosphere. Despite their importance, and in contrast to organisms that play roles in other major parts of the carbon cycle such as photosynthesis, no genome-level studies have been published on the biology of methanotrophs. We report the first complete genome sequence to our knowledge from an obligate methanotroph, Methylococcus capsulatus (Bath), obtained by the shotgun sequencing approach. Analysis revealed a 3.3-Mb genome highly specialized for a methanotrophic lifestyle, including redundant pathways predicted to be involved in methanotrophy and duplicated genes for essential enzymes such as the methane monooxygenases. We used phylogenomic analysis, gene order information, and comparative analysis with the partially sequenced methylotroph Methylobacterium extorquens to detect genes of unknown function likely to be involved in methanotrophy and methylotrophy. Genome analysis suggests the ability of M. capsulatus to scavenge copper (including a previously unreported nonribosomal peptide synthetase) and to use copper in regulation of methanotrophy, but the exact regulatory mechanisms remain unclear. One of the most surprising outcomes of the project is evidence suggesting the existence of previously unsuspected metabolic flexibility in M. capsulatus, including an ability to grow on sugars, oxidize chemolithotrophic hydrogen and sulfur, and live under reduced oxygen tension, all of which have implications for methanotroph ecology. The availability of the complete genome of M. capsulatus (Bath) deepens our understanding of methanotroph biology and its relationship to global carbon cycles. We have gained evidence for greater metabolic flexibility than was previously known, and for genetic components that may have biotechnological potential.


Assuntos
Regulação Bacteriana da Expressão Gênica , Genoma , Metano/metabolismo , Methylococcus capsulatus/genética , Proteínas de Bactérias/química , Carbono/química , Transporte de Elétrons , Ácidos Graxos/química , Genoma Bacteriano , Genômica/métodos , Metano/química , Modelos Biológicos , Dados de Sequência Molecular , Nitrogênio/química , Oxigênio/química , Oxigênio/metabolismo , Peptídeos/química , Filogenia , Análise de Sequência de DNA
20.
PLoS Biol ; 2(3): E69, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15024419

RESUMO

The complete sequence of the 1,267,782 bp genome of Wolbachia pipientis wMel, an obligate intracellular bacteria of Drosophila melanogaster, has been determined. Wolbachia, which are found in a variety of invertebrate species, are of great interest due to their diverse interactions with different hosts, which range from many forms of reproductive parasitism to mutualistic symbioses. Analysis of the wMel genome, in particular phylogenomic comparisons with other intracellular bacteria, has revealed many insights into the biology and evolution of wMel and Wolbachia in general. For example, the wMel genome is unique among sequenced obligate intracellular species in both being highly streamlined and containing very high levels of repetitive DNA and mobile DNA elements. This observation, coupled with multiple evolutionary reconstructions, suggests that natural selection is somewhat inefficient in wMel, most likely owing to the occurrence of repeated population bottlenecks. Genome analysis predicts many metabolic differences with the closely related Rickettsia species, including the presence of intact glycolysis and purine synthesis, which may compensate for an inability to obtain ATP directly from its host, as Rickettsia can. Other discoveries include the apparent inability of wMel to synthesize lipopolysaccharide and the presence of the most genes encoding proteins with ankyrin repeat domains of any prokaryotic genome yet sequenced. Despite the ability of wMel to infect the germline of its host, we find no evidence for either recent lateral gene transfer between wMel and D. melanogaster or older transfers between Wolbachia and any host. Evolutionary analysis further supports the hypothesis that mitochondria share a common ancestor with the alpha-Proteobacteria, but shows little support for the grouping of mitochondria with species in the order Rickettsiales. With the availability of the complete genomes of both species and excellent genetic tools for the host, the wMel-D. melanogaster symbiosis is now an ideal system for studying the biology and evolution of Wolbachia infections.


Assuntos
Genômica/métodos , Wolbachia/genética , Trifosfato de Adenosina/química , Animais , Linhagem da Célula , DNA/química , DNA/genética , Primers do DNA/química , Drosophila melanogaster/microbiologia , Evolução Molecular , Deleção de Genes , Duplicação Gênica , Biblioteca Gênica , Genes Bacterianos , Genoma , Genoma Bacteriano , Glicólise , Sequências Repetitivas Dispersas , Modelos Genéticos , Dados de Sequência Molecular , Fases de Leitura Aberta , Parasitos , Filogenia , Reação em Cadeia da Polimerase , Estrutura Terciária de Proteína , Purinas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA