Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 262
Filtrar
1.
Structure ; 32(10): 1725-1736.e4, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39032488

RESUMEN

Cag type IV secretion system (CagT4SS) translocates oncoprotein cytotoxin-associated gene A (CagA) into host cells and plays a key role in the pathogenesis of Helicobacter pylori. The structure of the outer membrane core complex (OMCC) in CagT4SS consists of CagX, CagY, CagM, CagT, and Cag3 in a stoichiometric ratio of 1:1:2:2:5 with 14-fold symmetry. However, the assembly pathway of OMCC remains elusive. Here, we report the crystal structures of CagT and Cag3-CagT complex, and the structural dynamics of Cag3 and CagT using hydrogen deuterium exchange-mass spectrometry (HDX-MS). The interwoven interaction of Cag3 and CagT involves conformational changes of CagT and ß strand swapping. In conjunction with biochemical and biophysical assays, we further demonstrate the different oligomerization states of Cag3 and Cag3-CagT complex. Additionally, the association with CagM requires the pre-formation of Cag3-CagT complex. These results demonstrate the generation of different intermediate sub-assemblies and their structural flexibility, potentially representing different building blocks for OMCC assembly.


Asunto(s)
Helicobacter pylori , Modelos Moleculares , Helicobacter pylori/metabolismo , Helicobacter pylori/química , Cristalografía por Rayos X , Multimerización de Proteína , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Tipo IV/metabolismo , Sistemas de Secreción Tipo IV/química , Unión Proteica , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Conformación Proteica
2.
J Mol Biol ; 436(4): 168432, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-38161000

RESUMEN

Helicobacter pylori colonizes the stomach in about half of the human population, leading to an increased risk of peptic ulcer disease and gastric cancer. H. pylori secretes an 88 kDa VacA toxin that contributes to pathogenesis. VacA assembles into oligomeric complexes in solution and forms anion-selective channels in cell membranes. Cryo-electron microscopy (cryo-EM) analyses of VacA oligomers in solution provided insights into VacA oligomerization but failed to reveal the structure of the hydrophobic N-terminal region predicted to be a pore-forming domain. In this study, we incubated VacA with liposomes and used single particle cryo-EM to analyze detergent-extracted VacA oligomers. A 3D structure of detergent-solubilized VacA hexamers revealed the presence of six α-helices extending from the center of the oligomers, a feature not observed in previous studies of water-soluble VacA oligomers. Cryo-electron tomography analysis and 2D averages of VacA associated with liposomes confirmed that central regions of the membrane-associated VacA oligomers can insert into the lipid bilayer. However, insertion is heterogenous, with some membrane-associated oligomers appearing only partially inserted and others sitting on top of the bilayer. These studies indicate that VacA undergoes a conformational change when contacting the membrane and reveal an α-helical region positioned to extend into the membrane. Although the reported VacA 3D structure does not represent a selective anion channel, our combined single particle 3D analysis, cryo-electron tomography, and modeling allow us to propose a model for the structural organization of the VacA N-terminus in the context of a hexamer as it inserts into the membrane.


Asunto(s)
Proteínas Bacterianas , Helicobacter pylori , Toxinas Biológicas , Canales Aniónicos Dependientes del Voltaje , Humanos , Proteínas Bacterianas/química , Microscopía por Crioelectrón/métodos , Detergentes , Helicobacter pylori/química , Liposomas/química , Toxinas Biológicas/química , Canales Aniónicos Dependientes del Voltaje/química , Multimerización de Proteína
3.
J Mol Recognit ; 36(9): e3049, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37553866

RESUMEN

Helicobacter pylori is the most common cause of gastric ulcers and is associated with gastric cancer. The enzyme HppA of class C nonspecific acid phosphohydrolases (NSAPs) of H. pylori plays a crucial role in the electron transport chain. Herein, we report an in silico homology model of HppA consisting of a monomeric α + ß model. A high throughput structure-based virtual screening approach yielded potential inhibitors against HppA with higher binding energies. Further analyses of molecular interaction maps and protein-ligand fingerprints, followed by molecular mechanics-generalized Born surface area (MM-GBSA) end point binding energy calculations of docked complexes, resulted in the detection of top binders/ligands. Our investigations identified potential substrate-competitive small molecule inhibitors of HppA, with admissible pharmacokinetic properties. These molecules may provide a starting point for developing novel therapeutic agents against H. pylori.


Asunto(s)
Fosfatasa Ácida , Helicobacter pylori , Fosfatasa Ácida/química , Fosfatasa Ácida/metabolismo , Helicobacter pylori/química , Helicobacter pylori/metabolismo , Simulación de Dinámica Molecular , Ensayos Analíticos de Alto Rendimiento , Simulación del Acoplamiento Molecular
4.
Comput Math Methods Med ; 2022: 7493834, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35069791

RESUMEN

Helicobacter pylori (H. pylori) is the most common risk factor for gastric cancer worldwide. The membrane proteins of the H. pylori are involved in bacterial adherence and play a vital role in the field of drug discovery. Thus, an accurate and cost-effective computational model is needed to predict the uncharacterized membrane proteins of H. pylori. In this study, a reliable benchmark dataset consisted of 114 membrane and 219 nonmembrane proteins was constructed based on UniProt. A support vector machine- (SVM-) based model was developed for discriminating H. pylori membrane proteins from nonmembrane proteins by using sequence information. Cross-validation showed that our method achieved good performance with an accuracy of 91.29%. It is anticipated that the proposed model will be useful for the annotation of H. pylori membrane proteins and the development of new anti-H. pylori agents.


Asunto(s)
Proteínas Bacterianas/genética , Helicobacter pylori/genética , Proteínas de la Membrana/genética , Secuencia de Aminoácidos , Aminoácidos/análisis , Proteínas Bacterianas/química , Biología Computacional , Bases de Datos de Proteínas/estadística & datos numéricos , Helicobacter pylori/química , Helicobacter pylori/patogenicidad , Interacciones Microbiota-Huesped , Humanos , Proteínas de la Membrana/química , Máquina de Vectores de Soporte
5.
Appl Biochem Biotechnol ; 194(1): 37-53, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34762267

RESUMEN

In the Asian region, Helicobacter pylori infects about 80% populations, which is most leading cause of peptic ulcers, and it is an asymptomatic infection. Studies reported that the particular bacteria carry specific virulence factors that leads to severe complications. These virulence factors can be used as a drug targets to inhibit their growth and pathogenicity. Chronic infection with H. pylori virulence factors are CagA, VacA and HtrA positive strains the risk factor of gastric cancer. In this study, we aimed to study the antagonistic interaction pattern between the potential eight algal peptides against the virulence factors of H. pylori through in silico analysis intended to treat peptic ulcer and prevent the further complications such as cancer. The proteins of virulent factors are docked using C-Docker algorithm and calculated the bind energy of the complexes. The results showed that the peptide derived from a green alga, Tetradesmus sp. are active against the three virulent factors such as cag-A, vac-A, and Htr-A with multiple hydrogen, vdW, electrostatic interactions, and mild π-hydrophobic bindings with the libdock energy score for CagA, VacA and HtrA are 175.625, 158.603 and 89.397 kcal/mol. These primes and the peptide lead to develop a better and potential inhibitors against H. pylori infection.


Asunto(s)
Proteínas Algáceas/química , Proteínas Bacterianas , Chlorophyta/química , Helicobacter pylori , Péptidos/química , Factores de Virulencia , Proteínas Algáceas/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Simulación por Computador , Helicobacter pylori/química , Helicobacter pylori/patogenicidad , Péptidos/farmacología , Factores de Virulencia/antagonistas & inhibidores , Factores de Virulencia/química
6.
Appl Biochem Biotechnol ; 194(1): 354-367, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34843077

RESUMEN

Gastric cancer is a pathological condition induced by the bacteria Helicobacter pylori. Targeting the key virulence factors of H. pylori causing gastric cancer is a promising method for treating gastric cancer. Recently, research has been focused on analyzing the adrenergic, cholinergic, and anti-cancer properties of their venom proteins. Testing the anti-cancer activity of the lethal proteins in the venom of P. volitans provides a bioactive compound for cancer treatment. Still, it is also helpful to eliminate the ecological imbalance caused by these fish in the marine environment. This study focuses on an in silico approach using Z-dock to analyze the bioactive prospective of the venom proteins of P. volitans against the essential virulence proteins of H. pylori responsible for inducing cancer. Our in silico docking study using a computational model of the venom proteins and H. pylori proteins has displayed the possible interactions between these proteins. The results revealed that P. volitans hyaluronidase and PV toxin's venom proteins effectively interact with H. pylori proteins Cag A, Cag L, GGT, Cag D, and urease that may be promising proteins in cancer therapy.


Asunto(s)
Proteínas Bacterianas/química , Proteínas de Peces/química , Venenos de los Peces/química , Helicobacter pylori/química , Simulación del Acoplamiento Molecular , Perciformes , Factores de Virulencia/química , Animales , Humanos , Neoplasias Gástricas
7.
Sci Rep ; 11(1): 24471, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34963695

RESUMEN

Helicobacter pylori is a Gram-negative bacterium that causes chronic inflammations in the stomach area and is involved in ulcers, which can develop into gastric malignancies. H. pylori attaches and colonizes to the human epithelium using some of their outer membrane proteins (OMPs). HomB and HomA are the most studied OMPs from H. pylori as they play a crucial role in adherence, hyper biofilm formation, antibiotic resistance and are also associated with severe gastric malignancies. The role of HomA and HomB in pathogenesis concerning their structure and function has not been evaluated yet. In the present study, we explored the structural aspect of HomA and HomB proteins using various computational, biophysical and small-angle X-ray scattering (SAXS) techniques. Interestingly, the in-silico analysis revealed that HomA/B consists of 8 discontinuous N and C terminal ß-strands forming a small ß-barrel, along with a large surface-exposed globular domain. Further, biophysical experiments suggested that HomA and HomB are dimeric and most likely the cysteine residues present on surface-exposed loops participate in protein-protein interactions. Our study provides essential structural information of unexplored proteins of the Hom family that can help in a better understanding of H. pylori pathogenesis.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/análisis , Infecciones por Helicobacter/microbiología , Helicobacter pylori/química , Secuencia de Aminoácidos , Humanos , Modelos Moleculares , Conformación Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X
8.
Cells ; 10(6)2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34199843

RESUMEN

BACKGROUND: Lipopolysaccharide (LPS) of Helicobacter pylori (Hp) bacteria causes disintegration of gastric tissue cells in vitro. It has been suggested that interleukin (IL)-33 is involved in healing gastric injury. AIM: To elucidate whether Hp LPS affects regeneration of gastric barrier initiated by IL-33. METHODS: Primary gastric epithelial cells or fibroblasts from Caviae porcellus were transfected with siRNA IL-33. Such cells, not exposed or treated with LPS Hp, were sub-cultured in the medium with or without exogenous IL-33. Then cell migration was assessed in conjunction with oxidative stress and apoptosis, activation of extracellular signal-regulated kinase (Erk), production of collagen I and soluble ST2 (IL-33 decoy). RESULTS: Control cells not treated with LPS Hp migrated in the presence of IL-33. The pro-regenerative activity of IL-33 was related to stimulation of cells to collagen I production. Wound healing by cells exposed to LPS Hp was inhibited even in the presence of IL-33. This could be due to increased oxidative stress and apoptosis in conjunction with Erk activation, sST2 elevation and modulation of collagen I production. CONCLUSIONS: The recovery of gastric barrier cells during Hp infection potentially can be affected due to downregulation of pro-regenerative activity of IL-33 by LPS Hp.


Asunto(s)
Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Mucosa Gástrica/fisiología , Helicobacter pylori/química , Interleucina-33/metabolismo , Lipopolisacáridos/farmacología , Regeneración/efectos de los fármacos , Animales , Cobayas , Lipopolisacáridos/química
9.
Biochemistry ; 60(24): 1933-1946, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34077175

RESUMEN

Helicobacter pylori is a Gram-negative bacterium that is responsible for gastric and duodenal ulcers. H. pylori uses the unusual mqn pathway with aminofutalosine (AFL) as an intermediate for menaquinone biosynthesis. Previous reports indicate that hydrolysis of AFL by 5'-methylthioadenosine nucleosidase (HpMTAN) is the direct path for producing downstream metabolites in the mqn pathway. However, genomic analysis indicates jhp0252 is a candidate for encoding AFL deaminase (AFLDA), an activity for deaminating aminofutolasine. The product, futalosine, is not a known substrate for bacterial MTANs. Recombinant jhp0252 was expressed and characterized as an AFL deaminase (HpAFLDA). Its catalytic specificity includes AFL, 5'-methylthioadenosine, 5'-deoxyadenosine, adenosine, and S-adenosylhomocysteine. The kcat/Km value for AFL is 6.8 × 104 M-1 s-1, 26-fold greater than that for adenosine. 5'-Methylthiocoformycin (MTCF) is a slow-onset inhibitor for HpAFLDA and demonstrated inhibitory effects on H. pylori growth. Supplementation with futalosine partially restored H. pylori growth under MTCF treatment, suggesting AFL deamination is significant for cell growth. The crystal structures of apo-HpAFLDA and with MTCF at the catalytic sites show a catalytic site Zn2+ or Fe2+ as the water-activating group. With bound MTCF, the metal ion is 2.0 Å from the sp3 hydroxyl group of the transition state analogue. Metabolomics analysis revealed that HpAFLDA has intracellular activity and is inhibited by MTCF. The mqn pathway in H. pylori bifurcates at aminofutalosine with HpMTAN producing adenine and depurinated futalosine and HpAFLDA producing futalosine. Inhibition of cellular HpMTAN or HpAFLDA decreased the cellular content of menaquinone-6, supporting roles for both enzymes in the pathway.


Asunto(s)
Helicobacter pylori/metabolismo , Nucleósidos/metabolismo , Vitamina K 2/metabolismo , Dominio Catalítico , Cristalografía por Rayos X/métodos , Desoxiadenosinas , Helicobacter pylori/química , Helicobacter pylori/enzimología , Modelos Moleculares , N-Glicosil Hidrolasas/química , N-Glicosil Hidrolasas/metabolismo , Nucleósidos/química , Purina-Nucleósido Fosforilasa/química , Especificidad por Sustrato , Tionucleósidos , Vitamina K 2/análogos & derivados
10.
J Microbiol ; 59(8): 763-770, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34061339

RESUMEN

Helicobacter pylori colonizes human gastric mucosa. Its infection is associated with gastric diseases including gastric cancer. CagA is one of the most important toxins produced by H. pylori. It is related to gastric cancer which can be injected into host cells via a type IV secretion system (T4SS). CagL is a structural component of T4SS apparatus, which triggers host cell signaling pathway. It has been reported that CagL polymorphisms may influence the severity of disease development. To explore the contribution of CagL polymorphisms between East Asian and Western H. pylori in pathogenesis, cagL gene in G27 H. pylori was swapped by K74 cagL which is identical to East Asian CagL consensus sequence and by Western 26695 H. pylori, resulting in G27 ΔcagL/cagLK74 and G27 ΔcagL/cagL26695, respectively. Intriguingly, G27 ΔcagL/cagLK74 showed significantly less ability of IL-8 induction than G27 ΔcagL/cagL26695 while displayed similar abilities of CagA phosphorylation, and cell elongation. Taken together, this study suggests that the CagL polymorphism may influence IL-8 induction, and K74 CagL has less ability to induce IL-8 secretion than G27 or 26695 CagL. Further research should address how the different capabilities of IL-8 induction between intraspecies-CagL are associated with the large differences of the incidence of gastric cancer between East Asian and Western countries.


Asunto(s)
Proteínas Bacterianas/genética , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/genética , Interleucina-8/metabolismo , Polimorfismo Genético , Secuencia de Aminoácidos , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Helicobacter pylori/química , Helicobacter pylori/metabolismo , Interacciones Huésped-Patógeno , Humanos , Interleucina-8/genética , República de Corea , Alineación de Secuencia
11.
Org Lett ; 22(22): 8780-8785, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33119312

RESUMEN

Helicobacter pylori, the most common cause of chronic gastritis, peptic ulcers, and gastric cancers, infects around half of the world's population. Although the drawbacks of antibiotic-based combination therapy are emerging, no effective vaccine is available to prevent H. pylori infections. Here, we describe the total synthesis of the unique α-(1→3)-linked tri-d-glycero-d-manno-heptose antigen from the lipopolysaccharide of H. pylori serogroups O3 and O6 and strains MO19, D2, D4, and D5 based on de novo synthesis of the differentially protected d-glycero-d-manno-heptosyl building blocks. Immunization of mice with the semisynthetic glycoconjugate elicited a very robust T-cell-dependent antigen-specific immune response, resulting in very high titers of IgG1 and IgG2b protective antibody isotypes. The postimmune sera recognized H. pylori NCTC 11637 and bound strongly to the surface of the intact bacteria.


Asunto(s)
Helicobacter pylori/inmunología , Heptosas/síntesis química , Lipopolisacáridos/química , Animales , Glicoconjugados/química , Helicobacter pylori/química , Heptosas/inmunología , Ratones , Estructura Molecular , Vacunas/inmunología
12.
Elife ; 92020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32876048

RESUMEN

The pathogenesis of Helicobacter pylori-associated gastric cancer is dependent on delivery of CagA into host cells through a type IV secretion system (T4SS). The H. pylori Cag T4SS includes a large membrane-spanning core complex containing five proteins, organized into an outer membrane cap (OMC), a periplasmic ring (PR) and a stalk. Here, we report cryo-EM reconstructions of a core complex lacking Cag3 and an improved map of the wild-type complex. We define the structures of two unique species-specific components (Cag3 and CagM) and show that Cag3 is structurally similar to CagT. Unexpectedly, components of the OMC are organized in a 1:1:2:2:5 molar ratio (CagY:CagX:CagT:CagM:Cag3). CagX and CagY are components of both the OMC and the PR and bridge the symmetry mismatch between these regions. These results reveal that assembly of the H. pylori T4SS core complex is dependent on incorporation of interwoven species-specific components.


Asunto(s)
Proteínas Bacterianas/ultraestructura , Helicobacter pylori/química , Sistemas de Secreción Tipo IV/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/clasificación , Microscopía por Crioelectrón , Modelos Moleculares , Conformación Proteica , Especificidad de la Especie , Sistemas de Secreción Tipo IV/química , Sistemas de Secreción Tipo IV/clasificación
13.
Biomolecules ; 10(7)2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32708696

RESUMEN

UreG is a P-loop GTP hydrolase involved in the maturation of nickel-containing urease, an essential enzyme found in plants, fungi, bacteria, and archaea. This protein couples the hydrolysis of GTP to the delivery of Ni(II) into the active site of apo-urease, interacting with other urease chaperones in a multi-protein complex necessary for enzyme activation. Whereas the conformation of Helicobacter pylori (Hp) UreG was solved by crystallography when it is in complex with two other chaperones, in solution the protein was found in a disordered and flexible form, defining it as an intrinsically disordered enzyme and indicating that the well-folded structure found in the crystal state does not fully reflect the behavior of the protein in solution. Here, isothermal titration calorimetry and site-directed spin labeling coupled to electron paramagnetic spectroscopy were successfully combined to investigate HpUreG structural dynamics in solution and the effect of Ni(II) and GTP on protein mobility. The results demonstrate that, although the protein maintains a flexible behavior in the metal and nucleotide bound forms, concomitant addition of Ni(II) and GTP exerts a structural change through the crosstalk of different protein regions.


Asunto(s)
Proteínas Bacterianas/metabolismo , Guanosina Trifosfato/metabolismo , Helicobacter pylori/metabolismo , Níquel/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Proteínas Bacterianas/química , Cristalografía por Rayos X , Infecciones por Helicobacter/microbiología , Helicobacter pylori/química , Humanos , Modelos Moleculares , Proteínas de Unión a Fosfato/química , Conformación Proteica
14.
Proteins ; 88(8): 1100-1109, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32181952

RESUMEN

Integration of template-based modeling, global sampling and precise scoring is crucial for the development of molecular docking programs with improved accuracy. We combined template-based modeling and ab-initio docking protocol as hybrid docking strategy called CoDock for the docking and scoring experiments of the seventh CAPRI edition. For CAPRI rounds 38-45, we obtained acceptable or better models in the top 10 submissions for eight out of the 16 evaluated targets as predictors, nine out of the 16 targets as scorers. Especially, we submitted acceptable models for all of the evaluated protein-oligosaccharide targets. For the CASP13-CAPRI experiment (round 46), we obtained acceptable or better models in the top 5 submissions for 10 out of the 20 evaluated targets as predictors, 11 out of the 20 targets as scorers. The failed cases for our group were mainly the difficult targets and the protein-peptide systems in CAPRI and CASP13-CAPRI experiments. In summary, this CAPRI edition showed that our hybrid docking strategy can be efficiently adapted to the increasing variety of challenges in the field of molecular interactions.


Asunto(s)
Simulación del Acoplamiento Molecular , Oligosacáridos/química , Péptidos/química , Proteínas/química , Programas Informáticos , Secuencia de Aminoácidos , Sitios de Unión , Helicobacter pylori/química , Helicobacter pylori/metabolismo , Humanos , Ligandos , Oligosacáridos/metabolismo , Péptidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Multimerización de Proteína , Proteínas/metabolismo , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/metabolismo , Proyectos de Investigación , Homología Estructural de Proteína , Termodinámica
15.
Biochemistry (Mosc) ; 85(2): 234-240, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32093599

RESUMEN

Helicobacter pylori is an important human pathogen that causes gastritis, gastric and duodenal ulcers, and gastric cancer. O-polysaccharides of H. pylori lipopolysaccharide (LPS) are composed of (ß1→3)-poly(N-acetyllactosamine) (polyLacNAc) decorated with multiple α-L-fucose residues. In many strains, their terminal LacNAc units are mono- or di-fucosylated to mimic Lewis X (Lex) and/or Lewis Y (Ley) oligosaccharides. The studies in rhesus macaques as a model of human infection by H. pylori showed that this bacterium adapts to the host during colonization by expressing host Lewis antigens. Here, we characterized LPS from H. pylori strains used in the previous study, including the parental J166 strain and the three derivatives (98-149, 98-169, and 98-181) isolated from rhesus macaques after long-term colonization. Chemical and NMR spectroscopic analyses of the LPS showed that the parent strain expressed Lex, Ley, and H type 1 terminal oligosaccharide units. The daughter strains were similar to the parental one in the presence of the same LPS core and fucosylated polyLacNAc chain of the same length but differed in the terminal oligosaccharide units. These were Lex in the isolates 98-149 and 98-169, which corresponded to the Lea phenotype of the host animals, and Ley was found in the 98-181 isolate from the macaque characterized by the Leb phenotype. As Lea and Leb are isomers of Lex and Ley, respectively, the observed correlation confirmed adaptation of the expression of terminal oligosaccharide units in H. pylori strains to the properties of the host gastric mucosa. The 98-181 strain also acquired glucosylation of the polyLacNAc chain and was distinguished by a lower expression of fucosylated internal LacNAc units (internal Lex) as a result of decoration of polyLacNAc with ß-glucopyranose, which may also play a role in the bacterial adaptation.


Asunto(s)
Helicobacter pylori/química , Lipopolisacáridos/química , Macaca mulatta/microbiología , Oligosacáridos/genética , Polisacáridos/metabolismo , Animales , Glicosilación , Helicobacter pylori/metabolismo , Lipopolisacáridos/aislamiento & purificación , Lipopolisacáridos/metabolismo , Oligosacáridos/análisis , Oligosacáridos/metabolismo , Fenotipo , Polisacáridos/química
16.
Biochim Biophys Acta Biomembr ; 1862(2): 183106, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31669571

RESUMEN

Gastric cancer is associated with high mortality and is preceded by an infection with Helicobacter pylori (H. pylori). H. pylori stimulates inflammation which involves the activation of Toll-like receptor 4 by lipopolysaccharide molecules from the H. pylori. This leads to chronic inflammation that can eventually lead to gastric cancer. Sox2 is a member of the high mobility group (HMG) box family of proteins, and recent studies have shown that HMG box proteins can modulate immune response by altering signaling to Toll-like receptors. Sox2 is overexpressed in most types of cancer with the exception of gastric cancer where expression of Sox2 is decreased. Here, we demonstrate that Sox2 can bind LPS and we investigated the thermodynamic drivers of the Sox2/LPS interaction.


Asunto(s)
Dominios HMG-Box , Lipopolisacáridos/química , Simulación del Acoplamiento Molecular , Factores de Transcripción SOXB1/química , Helicobacter pylori/química , Humanos , Lipopolisacáridos/metabolismo , Unión Proteica , Factores de Transcripción SOXB1/metabolismo
17.
Toxins (Basel) ; 11(11)2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31731531

RESUMEN

A key role in the carcinogenic action of Helicobacter pylori is played by the effector protein CagA, the first identified oncoprotein of the bacterial world. However, the present knowledge in regard to the bacterial injection of CagA into epithelial cells (through a type IV secretion system) and its intracellular fate is based primarily on experimental studies in vitro. Our study was aimed to investigate, in H. pylori-infected human gastric epithelium, CagA delivery and intracellular distribution in order to identify any in vivo counterpart of the cell injection mechanism described in vitro and any intracellular cytoplasmic site of preferential CagA distribution, thus shedding light on the natural history of CagA in vivo. By transmission electron microscopy and ultrastructural immunocytochemistry (which combine precise molecule localization with detailed analysis of bacterial-host cell interaction and epithelial cell ultrastructure), we investigated endoscopic biopsies of gastric antrum from H. pylori-infected dyspeptic patients. Our findings provide support for CagA direct injection into gastric epithelial cells at bacterial adhesion sites located on the lateral plasma membrane and for its cytosolic intracellular distribution with selective concentration inside peculiar proteasome-rich areas, which might be site not only of CagA degradation but also of CagA-promoted crucial events in gastric carcinogenesis.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Helicobacter pylori/química , Humanos
18.
Org Biomol Chem ; 17(21): 5269-5278, 2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31089638

RESUMEN

Mutasynthetic supplementation of the AHBA blocked mutant strain of S. hygroscopicus, the geldanamycin producer, with 21 aromatic and heteroaromatic amino acids provided new nonquinoid geldanamycin derivatives. Large scale (5 L) fermentation provided four new derivatives in sufficient quantity for full structural characterisation. Among these, the first thiophene derivative of reblastatin showed strong antiproliferative activity towards several human cancer cell lines. Additionally, inhibitory effects on human heat shock protein Hsp90α and bacterial heat shock protein from H. pylori HpHtpG were observed, revealing strong displacement properties for labelled ATP and demonstrating that the ATP-binding site of Hsps is the target site for the new geldanamycin derivatives.


Asunto(s)
Antineoplásicos/farmacología , Benzoquinonas/farmacología , Proteínas de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/farmacología , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Benzoquinonas/química , Benzoquinonas/aislamiento & purificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Proteínas de Choque Térmico/metabolismo , Helicobacter pylori/química , Humanos , Lactamas Macrocíclicas/química , Lactamas Macrocíclicas/aislamiento & purificación , Estructura Molecular , Streptomyces/química , Relación Estructura-Actividad
19.
J Comput Biol ; 26(10): 1177-1190, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31120321

RESUMEN

Helicobacter pylori is an infectious agent that colonizes the gastric mucosa of half of the population worldwide. This bacterium has been recognized as belonging to group 1 carcinogen by the World Health Organization for the role in development of gastritis, peptic ulcers, and cancer. Due to the increase in resistance to antibiotics used in the anti-H. pylori therapy, the development of an effective vaccine is an alternative of great interest, which remains a challenge. Therefore, a rational, strategic, and efficient vaccine design against H. pylori is necessary where the use of the most current bioinformatics tools could help achieve it. In this study, immunoinformatics approach was used to design a novel multiepitope oral vaccine against H. pylori. Our multiepitope vaccine is composed of cholera toxin subunit B (CTB) that is used as a mucosal adjuvant to enhance vaccine immunogenicity for oral immunization. CTB fused to 11 epitopes predicted of pathogenic (UreB170-189, VacA459-478, CagA1103-1122, GGT106-126, NapA30-44, and OipA211-230) and colonization (HpaA33-52, FlaA487-506, FecA437-456, BabA129-149, and SabA540-559) proteins from H. pylori. CKS9 peptide (CKSTHPLSC) targets epithelial microfold cells to enhance vaccine uptake from the gut barrier. All sequences were joined to each other by proper linkers. The vaccine was modeled and validated to achieve a high-quality three-dimensional structure. The vaccine design was evaluated as nonallergenic, antigenic, soluble, and with an appropriate molecular weight and isoelectric point. Our results suggest that our newly designed vaccine could serve as a promising anti-H. pylori vaccine candidate.


Asunto(s)
Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Epítopos/inmunología , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Administración Oral , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/química , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/química , Toxina del Cólera/administración & dosificación , Toxina del Cólera/inmunología , Biología Computacional , Epítopos/administración & dosificación , Infecciones por Helicobacter/inmunología , Helicobacter pylori/química , Humanos , Ratones , Modelos Moleculares , Estructura Secundaria de Proteína
20.
Mol Microbiol ; 112(2): 718-739, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31132312

RESUMEN

O-acetylserine sulfhydrylase (OASS) and cystathionine ß-synthase (CBS) are members of the PLP-II family, and involved in L-cysteine production. OASS produces L-cysteine via a de novo pathway while CBS participates in the reverse transsulfuration pathway. O-acetylserine-dependent CBS (OCBS) was previously identified as a new member of the PLP-II family, which are predominantly seen in bacteria. The bacterium Helicobacter pylori possess only one OASS (hp0107) gene and we showed that the protein coded by this gene actually functions as an OCBS and utilizes L-homocysteine and O-acetylserine (OAS) to produce cystathionine. HpOCBS did not show CBS activity with the substrate L-serine and required OAS exclusively. The HpOCBS structure in complex with methionine showed a closed cleft state, explaining the initial mode of substrate binding. Sequence and structural analyses showed differences between the active sites of OCBS and CBS, and explain their different substrate preferences. We identified three hydrophobic residues near the active site of OCBS, corresponding to one serine and two tyrosine residues in CBSs. Mutational studies were performed on HpOCBS and Saccharomyces cerevisiae CBS. A ScCBS double mutant (Y158F/Y226V) did not display activity with L-serine, indicating indispensability of these polar residues for selecting substrate L-serine, however, did show activity with OAS.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Cistationina betasintasa/química , Cistationina betasintasa/metabolismo , Helicobacter pylori/enzimología , Proteínas Bacterianas/genética , Sitios de Unión , Dominio Catalítico , Cistationina/metabolismo , Cistationina betasintasa/genética , Estabilidad de Enzimas , Helicobacter pylori/química , Helicobacter pylori/genética , Homocisteína/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Metionina/metabolismo , Serina/análogos & derivados , Serina/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA