Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Mil Med ; 189(7-8): e1753-e1759, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38243767

RESUMO

INTRODUCTION: Military trainees are at increased risk for infectious disease outbreaks because of the unique circumstances of the training environment (e.g., close proximity areas and physiologic/psychologic stress). Standard medical countermeasures in military training settings include routine immunization (e.g., influenza and adenovirus) as well as chemoprophylaxis [e.g., benzathine penicillin G (Bicillin) for the prevention of group A streptococcal disease] for pathogens associated with outbreaks in these settings. In a population of U.S. Army Infantry trainees, we evaluated changes in the oral microbiome during a 14-week military training cycle. MATERIALS AND METHODS: Trainees were enrolled in an observational cohort study in 2015-2016. In 2015, Bicillin was administered to trainees to ameliorate the risk of group A Streptococcus outbreaks, whereas in 2016, trainees did not receive a Bicillin inoculation. Oropharyngeal swabs were collected from participants at days 0, 7, 14, 28, 56, and 90 of training. Swabs were collected, flash frozen, and stored. DNA was extracted from swabs, and amplicon sequencing of the 16s rRNA gene was performed. Microbiome dynamics were evaluated using the QIIME 2 workflow along with DADA2, SINA with SILVA, and an additional processing in R. RESULTS: We observed that microbiome samples from the baseline (day 0) visit were distinct from one another, whereas samples collected on day 14 exhibited significant microbiome convergence. Day 14 convergence was coincident with an increase in DNA sequences associated with Streptococcus, though there was not a significant difference between Streptococcus abundance over time between 2015 and 2016 (P = .07), suggesting that Bicillin prophylaxis did not significantly impact overall Streptococcus abundance. CONCLUSIONS: The temporary convergence of microbiomes is coincident with a rise in communicable infections in this population. The dynamic response of microbiomes during initial military training supports similar observations in the literature of transient convergence of the human microbiome under cohabitation in the time frame including in this experiment. This population and the associated longitudinal studies allow for controlled studies of human microbiome under diverse conditions.


Assuntos
Microbiota , Militares , Humanos , Microbiota/fisiologia , Masculino , Militares/estatística & dados numéricos , Feminino , Estudos de Coortes , Georgia/epidemiologia , Boca/microbiologia , RNA Ribossômico 16S/análise
2.
bioRxiv ; 2023 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-37292721

RESUMO

The majority of the world population carry the gastric pathogen Helicobacter pylori. Fortunately, most individuals experience only low-grade or no symptoms, but in many cases the chronic inflammatory infection develops into severe gastric disease, including duodenal ulcer disease and gastric cancer. Here we report on a protective mechanism where H. pylori attachment and accompanying chronic mucosal inflammation can be reduced by antibodies that are present in a vast majority of H. pylori carriers. These antibodies block binding of the H. pylori attachment protein BabA by mimicking BabA's binding to the ABO blood group glycans in the gastric mucosa. However, many individuals demonstrate low titers of BabA blocking antibodies, which is associated with an increased risk for duodenal ulceration, suggesting a role for these antibodies in preventing gastric disease.

3.
Curr Oncol ; 29(5): 3576-3584, 2022 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-35621679

RESUMO

Pseudomyxoma peritonei (PMP) is a rare clinical syndrome. It originates from neoplasms of the appendix and leads to the formation of peritoneal implants and the accumulation of mucinous ascites. PMP represents a spectrum of low to high-grade disease. Despite aggressive management, many PMP patients recur, leading to debilitating symptoms and few treatment options. Therefore, scientists have continued to look for ways to improve treatment and further understand disease pathogenesis. Microorganisms were previously hypothesized to play a role in PMP progression and development. Hence, antibacterial treatment was suggested by some authors, but the data were limited. In this paper, we review the current data on the role of bacteria in PMP, discuss the significance, and suggest possible solutions to the inherent challenges in these studies. Given the limitations of the discussed studies, we remain skeptical about introducing novel antibacterial treatment into clinical practice at this time; however, the available data are valuable and indicate that more research into the molecular mechanisms of PMP is needed.


Assuntos
Neoplasias do Apêndice , Apêndice , Neoplasias Peritoneais , Pseudomixoma Peritoneal , Neoplasias do Apêndice/complicações , Neoplasias do Apêndice/diagnóstico , Neoplasias do Apêndice/patologia , Apêndice/patologia , Humanos , Recidiva Local de Neoplasia , Neoplasias Peritoneais/terapia , Pseudomixoma Peritoneal/diagnóstico , Pseudomixoma Peritoneal/patologia , Pseudomixoma Peritoneal/terapia
4.
J Microbiol ; 60(2): 207-214, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34757586

RESUMO

The polymorphic bacterial oncoprotein, CagA shows geography-dependent variation in the C-terminal Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs; East-Asian H. pylori isolates carry the ABD type while Western isolates carry the ABC type. In Western isolates, the EPIYA-C motif is sometimes found in multi-copy and this genotype is associated with disease severity. Interestingly, a small number of East-Asian H. pylori isolates have been found to carry Western ABC-type CagA. To gain a better understanding of these unusual isolates, the genomes of four Korean H. pylori clinical isolates carrying ABC-type CagA were sequenced via third generation (Pac-Bio SMRT) sequencing technology. The obtained data were utilized for phylogenetic analysis as well as comparison of additional virulence factors that are known to show geographic-dependent polymorphisms. Three of four isolates indeed belonged to the hpEastAsia group and showed typical East-Asian polymorphism in virulence factors such as homA/B/C, babA/B/C, and oipA. One isolate grouped to HpAfrica and showed typical Western polymorphism of virulence factors such as cagA, homA/B/C, and oipA. To understand the occurrence of the multi-copy EPIYA-C motif genotype in an East-Asian H. pylori background, the Korean clinical isolate, K154 was analyzed; this strain belonged to hpEastAsia but encoded CagA EPIYA-ABCCCC. Based on DNA sequence homology within the CagA multimerization (CM) sequence that flanked the EPIYA-C motifs, we predicted that the number of C motifs might change via homologous recombination. To test this hypothesis, K154 was cultured for one generation and 287 single colonies were analyzed for the number of EPIYA-C motifs using PCR-based screening and DNA sequencing verification. Three out of 284 (1%) single colony isolates showed changes in the number of EPIYA-C motifs in vitro; one isolate increased to five EPIYA-C motifs, one decreased to three EPIYA-C motifs, and one completely deleted the EPIYA-C motifs. The capacity for dynamic changes in the number of EPIYA-C repeats of CagA may play a role in generating important intraspecies diversity in East-Asian H. pylori.


Assuntos
Motivos de Aminoácidos , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Helicobacter pylori/classificação , Helicobacter pylori/genética , Fatores de Virulência/genética , Técnicas de Tipagem Bacteriana/métodos , DNA Bacteriano , Ásia Oriental , Genoma Bacteriano , Genótipo , Infecções por Helicobacter/microbiologia , Helicobacter pylori/metabolismo , Humanos , Filogenia , Polimorfismo Genético , República da Coreia
5.
Sci Rep ; 9(1): 11203, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31371778

RESUMO

Infection with CagA+ Helicobacter pylori strains is linked to an increased risk for gastric diseases, including gastric cancer. Recent evidence indicates that dynamic expansion and contraction of cagA copy number may serve as a novel mechanism to enhance disease development. Herein, comparative genomic analysis divided hpEurope into two groups: hpEurope/type-A and type-B. Only hpEurope/type-B displayed the multi-cagA genotype. Further analysis showed that cagPAI appears to have been independently introduced into two different H. pylori types, termed pre-type-A and pre-type-B, which consequently evolved to cagPAI type-A and type-B, respectively; importantly, all multi-cagA genotype strains displayed cagPAI type-B. Two direct cagA-flanking repeats of a genetic element termed CHA-ud were essential for the multi-cagA genotype in strain PMSS1 (hpEurope/type-B and cagPAI type-B). Furthermore, introduction of this genetic element into strain G27 (hpEurope/type-A and cagPAI type-A) was sufficient to generate the multi-cagA genotype. The critical steps in the evolution of the multi-cagA genotype involved creation of CHA-ud at cagA upstream in cagPAI type-B strains followed by its duplication to cagA downstream. En masse, elucidation of the mechanism by which H. pylori evolved to carry multiple copies of cagA helps to provide a better understanding of how this ancient pathogen interacts with its host.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Variações do Número de Cópias de DNA , Evolução Molecular , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Coevolução Biológica , DNA Intergênico/genética , Duplicação Gênica , Genes Bacterianos/genética , Genômica , Helicobacter pylori/patogenicidade , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Tipagem Molecular , Virulência/genética
6.
Adv Exp Med Biol ; 1149: 173-194, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31016629

RESUMO

Infecting half of the world's population, Helicobacter pylori is a medically important bacterium that induces a variety of gastric diseases, including gastritis, peptic ulcer disease, and gastric cancer. Sequencing of almost 1000 H. pylori isolates has revealed a diverse genome that contains abundant polymorphic genetic elements; many of these lie in factors likely to be associated with virulence. To ascertain the effect of these varying genetic elements, numerous epidemiological studies have investigated the contribution of the various polymorphisms to gastric disease development; particular focus has been placed on polymorphisms in the outer membrane proteins (OMPs), an effector protein, and a toxin produced by H. pylori. These studies have revealed geographic variation in the prevalence of various polymorphisms as well as in the associations between particular polymorphisms and gastric disease development. Furthermore, researchers have identified polymorphisms in multiple genes that frequently occur in combination. Though no single polymorphic genetic factor alone can fully account for gastric disease development in a population, the evaluation of multiple polymorphisms in a colonizing H. pylori strain can aid in the assessment of the pathogenic potential of the strain. Here we review specific H. pylori genetic polymorphisms (Bab proteins, Hom proteins, HopQ, SabA, SabB, OipA, IceA, VacA and CagA) that have been linked to disease outcome and discuss how geographic location and virulence factor polymorphisms together contribute to H. pylori-induced disease.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Polimorfismo Genético , Proteínas de Bactérias/genética , Genoma Bacteriano/genética , Genótipo , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Humanos
7.
Eur J Surg Oncol ; 45(9): 1723-1726, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30770164

RESUMO

Pseudomyxoma peritonei (PMP) is a subtype of peritoneal carcinomatosis that is traditionally treated by cytoreductive surgery (CRS) followed by hyperthermic intraperitoneal chemotherapy (HIPEC). A growing body of evidence suggests that microbes are associated with various tumor types and have been found in organs and cavities that were once considered sterile. Prior and ongoing research from our consortium of PMP researchers strongly suggests that bacteria are associated with PMP tumors. While the significance of this association is unclear, in our opinion, further research is warranted to understand whether these bacteria contribute to the development, maintenance and/or progression of PMP. Elucidation of a possible causal role for bacteria in PMP could suggest a benefit for supplementation of antibiotics to current treatment protocols.


Assuntos
Antibacterianos/uso terapêutico , Procedimentos Cirúrgicos de Citorredução , Hipertermia Induzida , Pseudomixoma Peritoneal/microbiologia , Pseudomixoma Peritoneal/terapia , Terapia Combinada , Feminino , Humanos , Masculino
8.
J Ethnopharmacol ; 232: 236-243, 2019 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-30578933

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: The neem tree (Azadirachta indica A.Juss), of the Meliaceae family, has been used in India for millennia in traditional medicine. Parts of the tree are used to treat problems with the gastrointestinal tract, urinary tract, and hair; to combat infections of smallpox and plasmodium; and to treat ulcers, diabetes, blood pressure, headache, and heartburn. Natural products and extracts from the tree have been reported to have antimicrobial, antifungal, and antiparasitic activities. AIM OF THE STUDY: Antibiotic resistance in the gastric pathogen Helicobacter pylori is increasing, and novel therapeutics to eradicate this bacterium are needed. Given the growing interest in the use of natural products as antimicrobials, this study was designed to examine the bactericidal effects of an extract of neem oil against H. pylori. MATERIALS AND METHODS: Neem oil was obtained from a commercial source and subjected to liquid-liquid extraction with diethyl ether and aqueous methanol; the methanol-soluble fraction was retained. The minimum inhibitory (MIC) and bactericidal (MBC) concentrations were determined against nine strains of H. pylori. Additionally, specific properties of the extract were characterized using H. pylori strain G27: bactericidal kinetics, reversibility, and effectiveness under growth arrest conditions and at low pH. The hemolytic activity of the extract was measured in vitro. RESULTS: The MIC and MBC of the extract against the H. pylori strains were between 25 and 51 µg/mL and 43-68 µg/mL, respectively. The bactericidal activity was time- and concentration-dependent, and at the highest concentrations (75-105 µg/mL), no detectable bacteria were present by 6 h. The activity of the extract was reversible, independent of H. pylori growth, and increased at low pH. The extract exhibited no appreciable hemolytic activity. CONCLUSIONS: Neem oil extract has significant bactericidal activity against H. pylori. The extract has several favorable pharmacological properties, including ability to kill non-growing bacteria, increased activity at low pH, and no hemolytic activity. The compound(s) present in the extract could potentially be used as a future treatment for H. pylori infection.


Assuntos
Antibacterianos/farmacologia , Glicerídeos/farmacologia , Helicobacter pylori/efeitos dos fármacos , Terpenos/farmacologia , Animais , Eritrócitos/efeitos dos fármacos , Helicobacter pylori/crescimento & desenvolvimento , Cavalos , Concentração de Íons de Hidrogênio , Testes de Sensibilidade Microbiana
9.
J Biol Inorg Chem ; 23(8): 1309-1330, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30264175

RESUMO

Helicobacter pylori HypA (HpHypA) is a metallochaperone necessary for maturation of [Ni,Fe]-hydrogenase and urease, the enzymes required for colonization and survival of H. pylori in the gastric mucosa. HpHypA contains a structural Zn(II) site and a unique Ni(II) binding site at the N-terminus. X-ray absorption spectra suggested that the Zn(II) coordination depends on pH and on the presence of Ni(II). This study was performed to investigate the structural properties of HpHypA as a function of pH and Ni(II) binding, using NMR spectroscopy combined with DFT and molecular dynamics calculations. The solution structure of apo,Zn-HpHypA, containing Zn(II) but devoid of Ni(II), was determined using 2D, 3D and 4D NMR spectroscopy. The structure suggests that a Ni-binding and a Zn-binding domain, joined through a short linker, could undergo mutual reorientation. This flexibility has no physiological effect on acid viability or urease maturation in H. pylori. Atomistic molecular dynamics simulations suggest that Ni(II) binding is important for the conformational stability of the N-terminal helix. NMR chemical shift perturbation analysis indicates that no structural changes occur in the Zn-binding domain upon addition of Ni(II) in the pH 6.3-7.2 range. The structure of the Ni(II) binding site was probed using 1H NMR spectroscopy experiments tailored to reveal hyperfine-shifted signals around the paramagnetic metal ion. On this basis, two possible models were derived using quantum-mechanical DFT calculations. The results provide a comprehensive picture of the Ni(II) mode to HpHypA, important to rationalize, at the molecular level, the functional interactions of this chaperone with its protein partners.


Assuntos
Proteínas de Bactérias/metabolismo , Helicobacter pylori/química , Metalochaperonas/metabolismo , Níquel/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Teoria da Densidade Funcional , Escherichia coli/genética , Glicina/genética , Concentração de Íons de Hidrogênio , Metalochaperonas/química , Metalochaperonas/genética , Modelos Químicos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Mutação , Níquel/química , Ressonância Magnética Nuclear Biomolecular/métodos , Ligação Proteica , Conformação Proteica em alfa-Hélice , Domínios Proteicos , Zinco/química , Zinco/metabolismo
10.
Front Microbiol ; 9: 1497, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30116222

RESUMO

One elusive area in the Helicobacter pylori field is an understanding of why some infections result in gastric cancer, yet others persist asymptomatically for the life-span of the individual. Even before the genomic era, the high level of intraspecies diversity of H. pylori was well recognized and became an intriguing area of investigation with respect to disease progression. Of interest in this regard is the unique repertoire of over 60 outer membrane proteins (OMPs), several of which have been associated with disease outcome. Of these OMPs, the association between HomB and disease outcome varies based on the population being studied. While the molecular roles for some of the disease-associated OMPs have been evaluated, little is known about the role that HomB plays in the H. pylori lifecycle. Thus, herein we investigated homB expression, regulation, and contribution to biofilm formation. We found that in H. pylori strain G27, homB was expressed at a relatively low level until stationary phase. Furthermore, homB expression was suppressed at low pH in an ArsRS-dependent manner; mutation of arsRS resulted in increased homB transcript at all tested time-points. ArsRS regulation of homB appeared to be direct as purified ArsR was able to specifically bind to the homB promoter. This regulation, combined with our previous finding that ArsRS mutations lead to enhanced biofilm formation, led us to test the hypothesis that homB contributes to biofilm formation by H. pylori. Indeed, subsequent biofilm analysis using a crystal-violet quantification assay and scanning electron microscopy (SEM) revealed that loss of homB from hyper-biofilm forming strains resulted in reversion to a biofilm phenotype that mimicked wild-type. Furthermore, expression of homB in trans from a promoter that negated ArsRS regulation led to enhanced biofilm formation even in strains in which the chromosomal copy of homB had been deleted. Thus, homB is necessary for hyper-biofilm formation of ArsRS mutant strains and aberrant regulation of this gene is sufficient to induce a hyper-biofilm phenotype. In summary, these data suggest that the ArsRS-dependent regulation of OMPs such as HomB may be one mechanism by which ArsRS dictates biofilm development in a pH responsive manner.

11.
Microbiol Mol Biol Rev ; 82(2)2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29743338

RESUMO

Despite decades of effort, Helicobacter pylori infections remain difficult to treat. Over half of the world's population is infected by H. pylori, which is a major cause of duodenal and gastric ulcers as well as gastric cancer. During chronic infection, H. pylori localizes within the gastric mucosal layer, including deep within invaginations called glands; thanks to its impressive ability to survive despite the harsh acidic environment, it can persist for the host's lifetime. This ability to survive and persist in the stomach is associated with urease production, chemotactic motility, and the ability to adapt to the fluctuating environment. Additionally, biofilm formation has recently been suggested to play a role in colonization. Biofilms are surface-associated communities of bacteria that are embedded in a hydrated matrix of extracellular polymeric substances. Biofilms pose a substantial health risk and are key contributors to many chronic and recurrent infections. This link between biofilm-associated bacteria and chronic infections likely results from an increased tolerance to conventional antibiotic treatments as well as immune system action. The role of this biofilm mode in antimicrobial treatment failure and H. pylori survival has yet to be determined. Furthermore, relatively little is known about the H. pylori biofilm structure or the genes associated with this mode of growth. In this review, therefore, we aim to highlight recent findings concerning H. pylori biofilms and the molecular mechanism of their formation. Additionally, we discuss the potential roles of biofilms in the failure of antibiotic treatment and in infection recurrence.


Assuntos
Antibacterianos/uso terapêutico , Biofilmes/crescimento & desenvolvimento , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Animais , Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Doença Crônica , Modelos Animais de Doenças , Resistência Microbiana a Medicamentos/genética , Infecções por Helicobacter/patologia , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/genética , Humanos , Recidiva
12.
Sci Rep ; 7(1): 14479, 2017 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-29101342

RESUMO

L-Asparaginases (ASNases) have been used as first line drugs for paediatric Acute Lymphoblastic Leukaemia (ALL) treatment for more than 40 years. Both the Escherichia coli (EcAII) and Erwinia chrysanthemi (ErAII) type II ASNases currently used in the clinics are characterized by high in vivo instability, short half-life and the requirement of several administrations to obtain a pharmacologically active concentration. Moreover, they are sensitive to proteases (cathepsin B and asparagine endopeptidase) that are over-expressed by resistant leukaemia lymphoblasts, thereby impairing drug activity and pharmacokinetics. Herein, we present the biochemical, structural and in vitro antiproliferative characterization of a new EcAII variant, N24S. The mutant shows completely preserved asparaginase and glutaminase activities, long-term storage stability, improved thermal parameters, and outstanding resistance to proteases derived from leukaemia cells. Structural analysis demonstrates a modification in the hydrogen bond network related to residue 24, while Normal Mode-based geometric Simulation and Molecular Dynamics predict a general rigidification of the monomer as compared to wild-type. These improved features render N24S a potential alternative treatment to reduce the number of drug administrations in vivo and to successfully address one of the major current challenges of ALL treatment: spontaneous, protease-dependent and immunological inactivation of ASNase.


Assuntos
Antineoplásicos/farmacologia , Asparaginase/metabolismo , Asparaginase/farmacologia , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/farmacologia , Escherichia coli/enzimologia , Antineoplásicos/química , Asparaginase/química , Asparaginase/genética , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Armazenamento de Medicamentos , Estabilidade Enzimática , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Humanos , Ligação de Hidrogênio , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Peptídeo Hidrolases/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Conformação Proteica , Temperatura
13.
Sci Rep ; 7(1): 11057, 2017 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-28887533

RESUMO

The polymorphic CagA toxin is associated with Helicobacter pylori-induced disease. Previous data generated using non-isogenic strains and transfection models suggest that variation surrounding the C-terminal Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs as well as the number of EPIYA motifs influence disease outcome. To investigate potential CagA-mediated effects on host cell signaling, we constructed and characterized a large panel of isogenic H. pylori strains that differ primarily in the CagA EPIYA region. The number of EPIYA-C motifs or the presence of an EPIYA-D motif impacted early changes in host cell elongation; however, the degree of elongation was comparable across all strains at later time points. In contrast, the strain carrying the EPIYA-D motif induced more IL-8 secretion than any other EPIYA type, and a single EPIYA-C motif induced comparable IL-8 secretion as isolates carrying multiple EPIYA-C alleles. Similar levels of ERK1/2 activation were induced by all strains carrying a functional CagA allele. Together, our data suggest that polymorphism in the CagA C-terminus is responsible for differential alterations in some, but not all, host cell signaling pathways. Notably, our results differ from non-isogenic strain studies, thus highlighting the importance of using isogenic strains to study the role of CagA toxin polymorphism in gastric cancer development.


Assuntos
Motivos de Aminoácidos , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Células Epiteliais/microbiologia , Helicobacter pylori/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Proteínas Mutantes/metabolismo , Transdução de Sinais , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Linhagem Celular , Células Epiteliais/fisiologia , Helicobacter pylori/genética , Humanos , Proteínas Mutantes/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
14.
mBio ; 8(1)2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28223454

RESUMO

Infection with Helicobacter pylori is a major risk factor for development of gastric disease, including gastric cancer. Patients infected with H. pylori strains that express CagA are at even greater risk of gastric carcinoma. Given the importance of CagA, this report describes a new molecular mechanism by which the cagA copy number dynamically expands and contracts in H. pylori Analysis of strain PMSS1 revealed a heterogeneous population in terms of numbers of cagA copies; strains carried from zero to four copies of cagA that were arranged as direct repeats within the chromosome. Each of the multiple copies of cagA was expressed and encoded functional CagA; strains with more cagA repeats exhibited higher levels of CagA expression and increased levels of delivery and phosphorylation of CagA within host cells. This concomitantly resulted in more virulent phenotypes as measured by cell elongation and interleukin-8 (IL-8) induction. Sequence analysis of the repeat region revealed three cagA homologous areas (CHAs) within the cagA repeats. Of these, CHA-ud flanked each of the cagA copies and is likely important for the dynamic variation of cagA copy numbers. Analysis of a large panel of clinical isolates showed that 7.5% of H. pylori strains isolated in the United States harbored multiple cagA repeats, while none of the tested Korean isolates carried more than one copy of cagA Finally, H. pylori strains carrying multiple cagA copies were differentially associated with gastric disease. Thus, the dynamic expansion and contraction of cagA copy numbers may serve as a novel mechanism by which H. pylori modulates gastric disease development.IMPORTANCE Severity of H. pylori-associated disease is directly associated with carriage of the CagA toxin. Though the sequences of the CagA protein can differ across strains, previous analyses showed that virtually all H. pylori strains carry one or no copies of cagA This study showed that H. pylori can carry multiple tandem copies of cagA that can change dynamically. Isolates harboring more cagA copies produced more CagA, thus enhancing toxicity to host cells. Analysis of 314 H. pylori clinical strains isolated from patients in South Korea and the United States showed that 7.5% of clinical strains in the United States carried multiple cagA copies whereas none of the South Korean strains did. This study demonstrated a novel molecular mechanism by which H. pylori dynamically modulates cagA copy number, which affects CagA expression and activity and may impact downstream development of gastric disease.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Dosagem de Genes , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/genética , Gastropatias/microbiologia , Gastropatias/patologia , Perfilação da Expressão Gênica , Helicobacter pylori/patogenicidade , Humanos , Coreia (Geográfico) , Sequências Repetitivas de Ácido Nucleico , Análise de Sequência de DNA , Homologia de Sequência , Estados Unidos
15.
Biochemistry ; 56(8): 1105-1116, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28177601

RESUMO

The human pathogen Helicobacter pylori requires nickel for colonization of the acidic environment of the stomach. HypA, a Ni metallochaperone that is typically associated with hydrogenase maturation, is also required for urease maturation and acid survival of H. pylori. There are two proposed Ni site structures for HypA; one is a paramagnetic six-coordinate site characterized by X-ray absorption spectroscopy (XAS) in unmodified HypA, while another is a diamagnetic four-coordinate planar site characterized by solution nuclear magnetic resonance in an N-terminally modified HypA construct. To determine the role of the N-terminal amine in Ni binding of HypA, an N-terminal extension variant, L2*-HypA, in which a leucine residue was inserted into the second position of the amino acid sequence in the proposed Ni-binding motif, was characterized in vitro and in vivo. Structural characterization of the Ni site using XAS showed a coordination change from six-coordinate in wild-type HypA (WT-HypA) to five-coordinate pyramidal in L2*-HypA, which was accompanied by the loss of two N/O donor protein ligands and the addition of an exogenous bromide ligand from the buffer. The magnetic properties of the Ni sites in WT-HypA compared to those of the Ni sites in L2*-HypA confirmed that a spin-state change from high to low spin accompanied this change in structure. The L2*-HypA H. pylori strain was shown to be acid sensitive and deficient in urease activity in vivo. In vitro characterization showed that L2*-HypA did not disrupt the HypA-UreE interaction that is essential for urease maturation but was at least 20-fold weaker in Ni binding than WT-HypA. Characterization of the L2*-HypA variant clearly demonstrates that the N-terminal amine of HypA is involved in proper Ni coordination and is necessary for urease activity and acid survival.


Assuntos
Aminas/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Helicobacter pylori/enzimologia , Níquel/metabolismo , Urease/metabolismo , Proteínas de Bactérias/genética , Proteínas de Transporte/genética , Metalochaperonas , Modelos Moleculares , Mutação , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína
16.
Fertil Steril ; 107(3): 813-820.e1, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28069180

RESUMO

OBJECTIVE: To conduct a pilot study to investigate the possible presence of bacteria throughout the female reproductive tract and to make a preliminary assessment of whether there are differences in the composition of the microbial communities between these body sites and/or between patients. DESIGN: Prospective pilot study followed by 16S amplification and high-throughput sequencing. SETTING: Tertiary care military hospital. PATIENT(S): A total of 10 women underwent a total hysterectomy with bilateral salpingo-oopherectomy; tissue samples were collected from the vagina, resected cervix, uterus, fallopian tubes, and ovaries. INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Microbial composition of samples within patients and between body sites. RESULT(S): The microbial composition of each sample was characterized by amplification and sequencing of the V1-V3 region of the 16S rRNA gene. Bacteria were identified in 95% of the samples; the remaining 5% of samples showed no evidence of bacterial 16S rRNA. The microbial communities present at each anatomical location were highly related across the samples and across the patients. The Firmicutes phylum was highly abundant as was the Lactobacillus genus. CONCLUSION(S): This study is the first global evaluation of the distribution of bacteria throughout the female reproductive tract in its entirety. Bacteria were detected by 16S sequencing from anatomical sites including the fallopian tubes and ovaries. The microbial profiles were closely related regardless of which body site or patient the samples originated from. The results of this trial will serve as the basis for future work correlating the colonization of the female reproductive tract with both obstetric and gynecologic conditions.


Assuntos
Bactérias/isolamento & purificação , Tubas Uterinas/microbiologia , Tubas Uterinas/cirurgia , Histerectomia , Ovariectomia , Ovário/microbiologia , Ovário/cirurgia , Salpingectomia , Útero/microbiologia , Útero/cirurgia , Adulto , Bactérias/classificação , Bactérias/genética , Feminino , Hospitais Militares , Humanos , Maryland , Pessoa de Meia-Idade , Projetos Piloto , Estudos Prospectivos , Ribotipagem
17.
Artigo em Inglês | MEDLINE | ID: mdl-29379775

RESUMO

Helicobacter pylori is associated with hypergastrinemia, which has been linked to the development of gastric diseases. Although the molecular mechanism is not fully understood, H. pylori is known to modulate the Erk pathway for induction of gastrin expression. Herein we found that an epidermal growth factor (EGF) receptor kinase inhibitor significantly blocked H. pylori-induced gastrin promoter activity, suggesting involvement of EGF receptor ligands. Indeed, H. pylori induced mRNA expression of EGF family members such as amphiregulin, EGF, heparin-binding EGF-like growth factor (HB-EGF), and transforming growth factor-α. Of these, specific siRNA targeting of HB-EGF significantly blocked H. pylori-induced gastrin expression. Moreover, H. pylori induced HB-EGF ectodomain shedding, which we found to be a critical process for H. pylori-induced gastrin expression. Thus, we demonstrate a novel role for human mature HB-EGF in stimulating gastrin promoter activity during H. pylori infection. Further investigation using specific siRNAs targeting each isoform of Raf, Mek, and Erk elucidated that the mechanism underlying H. pylori-induced gastrin expression can be delineated as the sequential activation of HB-EGF, the EGF receptor, C-Raf, Mek1, and the Erk2 molecules in the MAPK pathway. Surprisingly, whereas Erk2 acts as a potent activator of gastrin expression, siRNA knockdown of Erk1 induced gastrin promoter activity, suggesting that Erk1 typically acts as a repressor of gastrin expression. Elucidation of the mechanism of gastrin modulation by HB-EGF-mediated EGF receptor transactivation should facilitate the development of therapeutic strategies against H. pylori-related hypergastrinemia and consequently gastric disease development, including gastric cancers.


Assuntos
Gastrinas/genética , Regulação da Expressão Gênica , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Helicobacter pylori/fisiologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Transdução de Sinais , Proteínas de Ligação a DNA/metabolismo , Receptores ErbB/metabolismo , Gastrinas/metabolismo , Genes Reporter , Infecções por Helicobacter/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Mutação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-raf/metabolismo , RNA Mensageiro/genética , Fatores de Transcrição/metabolismo
18.
Curr Opin Infect Dis ; 29(3): 304-10, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26779778

RESUMO

PURPOSE OF REVIEW: Infection with the Gram-negative, microaerophilic pathogen Helicobacter pylori results in gastric cancer in a subset of infected individuals. As such, H. pylori is the only WHO classified bacterial class I carcinogen. Numerous studies have identified mechanisms by which H. pylori alters host cell signaling pathways to cause disease. The purpose of this review is to highlight recent studies that explore mechanisms associated with induction of gastric cancer. RECENT FINDINGS: Over the last year and a half, new mechanisms contributing to the etiology of H. pylori-associated gastric cancer development have been discovered. In addition to utilizing the oncogenic CagA toxin to alter host cell signaling pathways, H. pylori also induces host DNA damage and alters DNA methylation to perturb downstream signaling. Furthermore, H. pylori activates numerous host cell pathways and proteins that result in epithelial-to-mesenchymal transition and induction of cell survival and proliferation. SUMMARY: Mounting evidence suggests that H. pylori promotes gastric carcinogenesis using a multifactorial approach. Intriguingly, many of the targeted pathways and mechanisms show commonality with diverse forms of cancer.


Assuntos
Dano ao DNA , Transição Epitelial-Mesenquimal , Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Metilação de DNA , Progressão da Doença , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Humanos , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/microbiologia
19.
Genome Announc ; 3(6)2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-26564036

RESUMO

Peritoneal tumors from a rare malignancy, pseudomyxoma peritonei, frequently contain bacteria. Evidence suggests that tumor-associated bacteria contribute to pseudomyxoma peritonei development and/or progression. One unique isolate (PMP191F) was characterized via whole-genome sequencing using the Illumina MiSeq platform. PMP191F shows similarities to the Chitinophaga, Niastella, and Flavitalea genera.

20.
Int J Med Microbiol ; 305(3): 392-403, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25804332

RESUMO

Helicobacter pylori from different individuals exhibits substantial genetic diversity. However, the kinetics of bacterial diversification after infection with a single strain is poorly understood. We investigated evolution of H. pylori following long-term infection in the primate stomach; Rhesus macaques were infected with H. pylori strain USU101 and then followed for 10 years. H. pylori was regularly cultured from biopsies, and single colony isolates were analyzed. At 1-year, DNA fingerprinting showed that all output isolates were identical to the input strain; however, at 5-years, different H. pylori fingerprints were observed. Microarray-based comparative genomic hybridization revealed that long term persistence of USU101 in the macaque stomach was associated with specific whole gene changes. Further detailed investigation showed that levels of the BabA protein were dramatically reduced within weeks of infection. The molecular mechanisms behind this reduction were shown to include phase variation and gene loss via intragenomic rearrangement, suggesting strong selective pressure against BabA expression in the macaque model. Notably, although there is apparently strong selective pressure against babA, babA is required for establishment of infection in this model as a strain in which babA was deleted was unable to colonize experimentally infected macaques.


Assuntos
Variação Genética , Infecções por Helicobacter/microbiologia , Helicobacter pylori/genética , Helicobacter pylori/isolamento & purificação , Adesinas Bacterianas/genética , Animais , Biópsia , Hibridização Genômica Comparativa , Impressões Digitais de DNA , DNA Bacteriano/genética , Modelos Animais de Doenças , Rearranjo Gênico , Estudos Longitudinais , Macaca mulatta , Análise em Microsséries , Seleção Genética , Estômago/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA