Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.595
Filtrar
Mais filtros

Eixos temáticos
Intervalo de ano de publicação
1.
Nat Immunol ; 20(6): 687-700, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31061528

RESUMO

Most tissue-resident macrophage populations develop during embryogenesis, self-renew in the steady state and expand during type 2 immunity. Whether shared mechanisms regulate the proliferation of macrophages in homeostasis and disease is unclear. Here we found that the transcription factor Bhlhe40 was required in a cell-intrinsic manner for the self-renewal and maintenance of large peritoneal macrophages (LPMs), but not that of other tissue-resident macrophages. Bhlhe40 was necessary for the proliferation, but not the polarization, of LPMs in response to the cytokine IL-4. During infection with the helminth Heligmosomoides polygyrus bakeri, Bhlhe40 was required for cell cycling of LPMs. Bhlhe40 repressed the expression of genes encoding the transcription factors c-Maf and Mafb and directly promoted expression of transcripts encoding cell cycle-related proteins to enable the proliferation of LPMs. In LPMs, Bhlhe40 bound to genomic sites co-bound by the macrophage lineage-determining factor PU.1 and to unique sites, including Maf and loci encoding cell-cycle-related proteins. Our findings demonstrate a tissue-specific control mechanism that regulates the proliferation of resident macrophages in homeostasis and type 2 immunity.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Proteínas de Homeodomínio/genética , Homeostase/genética , Homeostase/imunologia , Imunidade/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores , Ciclo Celular/genética , Ciclo Celular/imunologia , Proliferação de Células , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Infecções por Helicobacter/genética , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Proteínas de Homeodomínio/metabolismo , Imunofenotipagem , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Transgênicos , Monócitos/imunologia , Monócitos/metabolismo , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Transcriptoma
2.
Immunity ; 52(4): 573-575, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32294401

RESUMO

Little is known about host-microbiota interactions regulating anti-microbial immunity in the stomach. In this issue, Satoh-Takayama et al. describe an additional immune mechanism involving innate lymphoid cells type 2 (ILC2), which controls infection with Helicobacter pylori, a bacterium associated with inflammation and cancer.


Assuntos
Helicobacter pylori , Linfócitos , Helicobacter pylori/imunologia , Imunidade Inata , Imunoglobulina A , Estômago
3.
Immunity ; 52(4): 635-649.e4, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32240600

RESUMO

The intestinal microbiota shapes and directs immune development locally and systemically, but little is known about whether commensal microbes in the stomach can impact their immunological microenvironment. Here, we report that group 2 innate lymphoid cells (ILC2s) were the predominant ILC subset in the stomach and show that their homeostasis and effector functions were regulated by local commensal communities. Microbes elicited interleukin-7 (IL-7) and IL-33 production in the stomach, which in turn triggered the propagation and activation of ILC2. Stomach ILC2s were also rapidly induced following infection with Helicobacter pylori. ILC2-derived IL-5 resulted in the production of IgA, which coated stomach bacteria in both specific pathogen-free (SPF) and H. pylori-infected mice. Our study thus identifies ILC2-dependent IgA response that is regulated by the commensal microbiota, which is implicated in stomach protection by eliminating IgA-coated bacteria including pathogenic H. pylori.


Assuntos
Microbioma Gastrointestinal/imunologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/patogenicidade , Imunoglobulina A/biossíntese , Interleucina-5/imunologia , Estômago/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Feminino , Regulação da Expressão Gênica , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/imunologia , Imunidade Humoral , Imunidade Inata , Interleucina-33/genética , Interleucina-33/imunologia , Interleucina-5/genética , Interleucina-7/genética , Interleucina-7/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cultura Primária de Células , Transdução de Sinais , Estômago/microbiologia , Simbiose/imunologia , Subpopulações de Linfócitos T/classificação
4.
Cell Immunol ; 399-400: 104811, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38518686

RESUMO

Helicobacter pylori-associated stomach infection is a leading cause of gastric ulcer and related cancer. H. pylori modulates the functions of infiltrated immune cells to survive the killing by reactive oxygen and nitrogen species (ROS and RNS) produced by these cells. Uncontrolled immune responses further produce excess ROS and RNS which lead to mucosal damage. The persistent oxidative stress is a major cause of gastric cancer. H. pylori regulates nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs), nitric oxide synthase 2 (NOS2), and polyamines to control ROS and RNS release through lesser-known mechanisms. ROS and RNS produced by these pathways differentiate macrophages and T cells from protective to inflammatory phenotype. Pathogens-associated molecular patterns (PAMPs) induced ROS activates nuclear oligomerization domain (NOD), leucine rich repeats (LRR) and pyrin domain-containing protein 3 (NLRP3) inflammasome for the release of pro-inflammatory cytokines. This study evaluates the role of H. pylori secreted concentrated proteins (HPSCP) related oxidative stress role in NLRP3 inflammasome activation and macrophage differentiation. To perceive the role of ROS/RNS, THP-1 and AGS cells were treated with 10 µM diphenyleneiodonium (DPI), 50 µM salicyl hydroxamic acid (SHX), 5 µM Carbonyl cyanide-4-(trifluoromethoxy) phenylhydrazone (FCCP), which are specific inhibitors of NADPH oxidase (NOX), Myeloperoxidase (MPO), and mitochondrial oxidative phosphorylation respectively. Cells were also treated with 10 µM of NOS2 inhibitor l-NMMA and 10 µM of N-acetyl cysteine (NAC), a free radical scavenger·H2O2 (100 µM) treated and untreated cells were used as positive controls and negative control respectively. The expression of gp91phox (NOX2), NOS2, NLRP3, CD86 and CD163 was analyzed through fluorescent microscopy. THP-1 macrophages growth was unaffected whereas the gastric epithelial AGS cells proliferated in response to higher concentration of HPSCP. ROS and myeloperoxidase (MPO) level increased in THP-1 cells and nitric oxide (NO) and lipid peroxidation significantly decreased in AGS cells. gp91phox expression was unchanged, whereas NOS2 and NLRP3 downregulated in response to HPSCP, but increased after inhibition of NO, ROS and MPO in THP-1 cells. HPSCP upregulated the expression of M1 and M2 macrophage markers, CD86 and CD163 respectively, which was decreased after the inhibition of ROS. This study concludes that there are multiple pathways which are generating ROS during H. pylori infection which further regulates other cellular processes. NO is closely associated with MPO and inhibition of NLRP3 inflammasome. The low levels of NO and MPO regulates gastrointestinal tract homeostasis and overcomes the inflammatory response of NLRP3. The ROS also plays crucial role in macrophage polarization hence alter the immune responses duing H. pylori pathogenesis.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Inflamassomos , Macrófagos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Estresse Oxidativo , Espécies Reativas de Oxigênio , Humanos , Helicobacter pylori/imunologia , Espécies Reativas de Oxigênio/metabolismo , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/metabolismo , Inflamassomos/metabolismo , Inflamassomos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Macrófagos/metabolismo , Macrófagos/imunologia , Proteínas de Bactérias/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Células THP-1 , NADPH Oxidases/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Diferenciação Celular/imunologia
5.
Helicobacter ; 29(4): e13119, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39108210

RESUMO

Helicobacter pylori infection causes chronic gastritis, ulcers, and gastric cancer, making it a threat to human health. Despite the use of antibiotic therapy, the global prevalence of H. pylori infection remains high, necessitating early eradication measures. Immunotherapy, especially vaccine development, is a promising solution in this direction, albeit the selection of an appropriate animal model is critical in efficient vaccine production. Accordingly, we conducted a literature, search and summarized the commonly used H. pylori strains, H. pylori infection-related animal models, and models for evaluating H. pylori vaccines. Based on factors such as the ability to replicate human diseases, strain compatibility, vaccine types, and eliciting of immune responses, we systematically compared the advantages and disadvantages of different animal models, to obtain the informed recommendations. In addition, we have proposed novel perspectives on H. pylori-related animal models to advance research and vaccine evaluation for the prevention and treatment of diseases such as gastric cancer.


Assuntos
Vacinas Bacterianas , Modelos Animais de Doenças , Infecções por Helicobacter , Helicobacter pylori , Infecções por Helicobacter/prevenção & controle , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/imunologia , Animais , Vacinas Bacterianas/imunologia , Helicobacter pylori/imunologia , Humanos
6.
Helicobacter ; 29(2): e13067, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38514932

RESUMO

BACKGROUND: Helicobacter pylori (H. pylori) is a rod-shaped, gram-negative, microaerophilic bacterium that can be identified by gram staining. Its relationship with cancer is significant since it is involved in approximately 80% of gastric cancers and 5.5% of all malignant cancers. Two lines of treatment have been defined for H. pylori, but almost 40% of patients do not respond to the first line. Recent trials have investigated oral Immunotherapy as a new treatment method. The aim of this systematic review was to investigate the potential effects of oral Immunotherapy on eradication rate of H. pylori in human studies. METHODS: The systematic review was performed according to PRISMA guidelines. We searched online databases, including Scopus, PubMed, and Web of Science (ISI). Our search strategy was limited to English articles and studies on human populations that use oral immunotherapy for H. pylori. RESULTS: The total number of primary research records in different databases was 2775. After removing duplicate articles (n = 870), we excluded 1829 for reasons including non-human studies, irrelevance to our study objective, non-English language, or lack of information. Of the remaining 76 articles, only seven had sufficient information, and the rest were excluded. The studies were divided into two groups: those that used bovine antibody and those that used immunoglobulin Y to eradicate H. pylori. CONCLUSION: In the group of Immunoglobulin Y, three out of four studies suggest that using Immunoglobulin Y for the treatment of H. pylori infection is significant. However, the group using bovine antibody for the treatment of H. pylori infection has various results, as two out of three studies concluded that bovine antibody therapy is not significant.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Imunoterapia , Humanos , Infecções por Helicobacter/terapia , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Imunoterapia/métodos , Administração Oral , Animais
7.
Helicobacter ; 29(4): e13128, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39177204

RESUMO

BACKGROUND: Infection with Helicobacter pylori (Hp) mostly occurs during childhood, and persistent infection may lead to severe gastric diseases and even gastric cancer. Currently, the primary method for eradicating Hp is through antibiotic treatment. However, the increasing multidrug resistance in Hp strains has diminished the effectiveness of antibiotic treatments. Vaccination could potentially serve as an effective intervention to resolve this issue. AIMS: Through extensive research and analysis of the vital protein characteristics involved in Hp infection, we aim to provide references for subsequent vaccine antigen selection. Additionally, we summarize the current research and development of Hp vaccines in order to provide assistance for future research. MATERIALS AND METHODS: Utilizing the databases PubMed and the Web of Science, a comprehensive search was conducted to compile articles pertaining to Hp antigens and vaccines. The salient aspects of these articles were then summarized to provide a detailed overview of the current research landscape in this field. RESULTS: Several potential antigens have been identified and introduced through a thorough understanding of the infection process and pathogenic mechanisms of Hp. The conserved and widely distributed candidate antigens in Hp, such as UreB, HpaA, GGT, and NAP, are discussed. Proteins such as CagA and VacA, which have significant virulence effects but relatively poor conservatism, require further evaluation. Emerging antigens like HtrA and dupA have significant research value. In addition, vaccines based on these candidate antigens have been compiled and summarized. CONCLUSIONS: Vaccines are a promising method for preventing and treating Hp. While some Hp vaccines have achieved promising results, mature products are not yet available on the market. Great efforts have been directed toward developing various types of vaccines, underscoring the need for developers to select appropriate antigens and vaccine formulations to improve success rates.


Assuntos
Antígenos de Bactérias , Vacinas Bacterianas , Infecções por Helicobacter , Helicobacter pylori , Desenvolvimento de Vacinas , Helicobacter pylori/imunologia , Vacinas Bacterianas/imunologia , Infecções por Helicobacter/prevenção & controle , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Humanos , Antígenos de Bactérias/imunologia , Animais
8.
Helicobacter ; 29(3): e13105, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38924222

RESUMO

Helicobacter pylori (H. pylori) colonizes the stomach and leads to the secretion of a vast range of cytokines by infiltrated leukocytes directing immune/inflammatory response against the bacterium. To regulate immune/inflammatory responses, suppressors of cytokine signaling (SOCS) proteins bind to multiple signaling components located downstream of cytokine receptors, such as Janus kinase (JAK), signal transducers and activators of transcription (STAT). Dysfunctional SOCS proteins in immune cells may facilitate the immune evasion of H. pylori, allowing the bacteria to induce chronic inflammation. Dysregulation of SOCS expression and function can contribute to the sustained H. pylori-mediated gastric inflammation which can lead to gastric cancer (GC) development. Among SOCS molecules, dysregulated expression of SOCS1, SOCS2, SOCS3, and SOCS6 were indicated in H. pylori-infected individuals as well as in GC tissues and cells. H. pylori-induced SOCS1, SOCS2, SOCS3, and SOCS6 dysregulation can contribute to the GC development. The expression of SOCS molecules can be influenced by various factors, such as epigenetic DNA methylation, noncoding RNAs, and gene polymorphisms. Modulation of the expression of SOCS molecules in gastric epithelial cells and immune cells can be considered to control gastric carcinogenesis as well as regulate antitumor immune responses, respectively. This review aimed to explain the interplay between H. pylori and SOCS molecules in GC development and immune response induction as well as to provide insights regarding potential therapeutic strategies modulating SOCS molecules.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Proteínas Supressoras da Sinalização de Citocina , Humanos , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/microbiologia , Helicobacter pylori/imunologia , Helicobacter pylori/patogenicidade , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética , Interações Hospedeiro-Patógeno/imunologia , Transdução de Sinais
9.
Helicobacter ; 29(3): e13097, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38819071

RESUMO

BACKGROUND: Helicobacter pylori (H. pylori) can evade the host's immune response and persist for a long time on the gastric mucosa. T helper (Th) cells appear to be involved in the control of H. pylori bacteria but promote mucosal inflammation. In contrast, regulatory T cells (Tregs) may reduce inflammation but promote H. pylori persistence. CC motif chemokine receptor 6 (CCR6) is involved in the migration of various cells into inflamed gastric mucosa. In this study, we examined CCR6+ Th cells and CCR6+ Tregs during H. pylori infection in humans. MATERIALS AND METHODS: Isolation of cells from blood and mucosal biopsies, magnetic separation of В cells, CD4+ and CD4+CCR6+CD45RO+ T cells, antigen-specific activation, B cell response in vitro, flow cytometry, determination of CD4+CD25hiFoxP3+ Tregs and various groups of Th cells. RESULTS: CD4+CCR6+ blood lymphocytes from healthy donors included Th cells and Tregs. These CCR6+ Th cells produced proinflammatory cytokines and also stimulated plasma cell maturation and antibody production in vitro. H. pylori gastritis and peptic ulcer disease were associated with an increase in the number of circulate CD4+CCR6+CD45RO+ cells and the percentage of Th1, Th17 and Th1/17 cells in this lymphocyte subgroup. In H. pylori-positive patients, circulating CD4+CCR6+ cells contained a higher proportion of H. pylori-specific cells compared with their CD4+CCR6- counterparts. H. pylori infection strongly increased the content of CD4+ lymphocytes in the inflamed gastric mucosa, with the majority of these CD4+ lymphocytes expressing CCR6. CD4+CCR6+ lymphocytes from H. pylori-infected stomach included Tregs and in vivo activated T cells, some of which produced interferon-γ without ex vivo stimulation. CONCLUSION: H. pylori infection causes an increase in the number of mature CD4+CCR6+ lymphocytes in the blood, with a pro-inflammatory shift in their composition and enrichment of the gastric mucosa with CD4+CCR6+ lymphocytes, including CCR6+ Th1 cells and Tregs.


Assuntos
Mucosa Gástrica , Infecções por Helicobacter , Helicobacter pylori , Receptores CCR6 , Linfócitos T Reguladores , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Citometria de Fluxo , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/imunologia , Receptores CCR6/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia
10.
Helicobacter ; 29(3): e13096, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38790093

RESUMO

BACKGROUND: Helicobacter pylori (H. pylori) is a common bacterial infection which predominately drives upper gastrointestinal pathology. We carried out a nationwide serological survey in response to the deficiency of robust African data on H. pylori prevalence, age of acquisition, socio-geographic determinants, and impact on gastric physiology. MATERIALS AND METHODS: This was a cross-sectional study of archival plasma samples collected during the Zambia Population-based HIV impact Assessment (ZAMPHIA) 2016 survey. ZAMPHIA used a two-stage door-to-door stratified cluster sample approach to collect samples from adults and children from age 0 to 59 years (n = 24,266). We randomly retrieved one fifth of these samples from each of Zambia's 10 provinces and used ELISA to test for H. pylori IgG antibodies, pepsinogen 1 and 2 and gastrin-17. A pepsinogen 1:2 ratio of <3 was used to define gastric atrophy. RESULTS: The analysis of 4050 plasma samples (30% <16 years, 53% females) revealed an overall H. pylori seroprevalence of 79%. By the age of 10 years, more than 75% of the children had H. pylori. Urban residence was associated with increased odds (OR 1.8, 95% CI 1.5-2.2, p < 0.001) and HIV infection was associated with reduced odds (OR 0.7, 95% CI 0.5-0.9, p = 0.02) of H. pylori seropositivity. Gastric atrophy was detected in 6% of H. pylori seropositive adults below 45 years of age and 9% in those between 45 and 59 years. CONCLUSIONS: We have confirmed a high prevalence of H. pylori seropositivity in Zambia, predominantly in urban settings. The prevalence of gastric atrophy is broadly consistent with other populations around the globe, but our sample did not include adults over 60 years.


Assuntos
Anticorpos Antibacterianos , Infecções por Helicobacter , Helicobacter pylori , Humanos , Zâmbia/epidemiologia , Infecções por Helicobacter/epidemiologia , Infecções por Helicobacter/microbiologia , Feminino , Masculino , Adulto , Estudos Transversais , Adolescente , Pessoa de Meia-Idade , Adulto Jovem , Helicobacter pylori/isolamento & purificação , Helicobacter pylori/imunologia , Criança , Pré-Escolar , Anticorpos Antibacterianos/sangue , Estudos Soroepidemiológicos , Lactente , Prevalência , Recém-Nascido , Imunoglobulina G/sangue , Gastrite Atrófica/epidemiologia , Gastrite Atrófica/microbiologia
11.
Helicobacter ; 29(3): e13104, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38923222

RESUMO

Helicobacter pylori (H. pylori) strain is the most genetically diverse pathogenic bacterium and now alarming serious human health concern ranging from chronic gastritis to gastric cancer and human death all over the world. Currently, the majority of commercially available diagnostic assays for H. pylori is a challenging task due to the heterogeneity of virulence factors in various geographical regions. In this concern, designing of universal multi-epitope immunogenic biomarker targeted for all H. pylori strains would be crucial to successfully immunodiagnosis assay and vaccine development for H. pylori infection. Hence, the present study aimed to explore the potential immunogenic epitopes of PSA D15 and Cag11 proteins of H. pylori, using immunoinformatics web tools in order to design novel immune-reactive multi-epitope antigens for enhanced immunodiagnosis in humans. Through an in silico immunoinformatics approach, high-ranked B-cell, MHC-I, and MHC-II epitopes of PSA D15 and Cag11 proteins were predicted, screened, and selected. Subsequently, a novel multi-epitope PSA D15 and Cag11 antigens were designed by fused the high-ranked B-cell, MHC-I, and MHC-II epitopes and 50S ribosomal protein L7/L12 adjuvant using linkers. The antigenicity, solubility, physicochemical properties, secondary and tertiary structures, 3D model refinement, and validations were carried. Furthermore, the designed multi-epitope antigens were subjected to codon adaptation and in silico cloning, immune response simulation, and molecular docking with receptor molecules. A novel, stable multi-epitope PSA D15 and Cag11 H. pylori antigens were developed and immune simulation of the designed antigens showed desirable levels of immunological response. Molecular docking of designed antigens with immune receptors (B-cell, MHC-I, MHC-II, and TLR-2/4) revealed robust interactions and stable binding affinity to the receptors. The codon optimized and in silico cloned showed that the designed antigens were successfully expressed (CAI value of 0.95 for PSA D15 and 1.0 for Cag11) after inserted into pET-32ba (+) plasmid of the E. coli K12 strain. In conclusion, this study revealed that the designed multi-epitope antigens have a huge immunological potential candidate biomarker and useful in developing immunodiagnostic assays and vaccines for H. pylori infection.


Assuntos
Antígenos de Bactérias , Biologia Computacional , Helicobacter pylori , Helicobacter pylori/imunologia , Helicobacter pylori/genética , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/química , Humanos , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Epitopos/imunologia , Testes Imunológicos/métodos , Simulação de Acoplamento Molecular , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/genética , Imunoinformática
12.
Helicobacter ; 29(3): e13107, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38943311

RESUMO

BACKGROUND: The "hygiene hypothesis" states that reduced exposure to microbial antigens due to an excessively hygienic environment can increase the risk of developing autoimmune diseases, including atopic disorders and asthma. In recent decades, there has been a progressive decline in the prevalence of numerous microorganisms following improved hygienic-sanitary conditions. More specifically, several studies reported an inverse association between the reduction in Helicobacter pylori infection and the rise of asthma and allergic disorders. AIM: To evaluate the prevalence of atopic disorders in a pediatric population in relation to seropositivity against H. pylori. METHODS: Children from Northern Sardinia, Italy, referred to the local Children's Hospital for any reason, were investigated to identify risk factors, especially H. pylori infection, associated with atopic disorders. A validated questionnaire, including demographics, house size, history of breastfeeding, residence, school or daycare center attendance, exposure to animals, and a defined diagnosis of atopy-including asthma-was filled out by a trained pediatrician according to parents' answers and child records. A blood sample was collected from each participant and immunoglobulin G against H. pylori was assessed by a locally validated ELISA test. RESULTS: The seroprevalence of H. pylori infection was 11.7% among 492 children (240 females). Thirty-two children had a confirmed diagnosis of asthma and 12 of allergy. No one child showed both conditions. Statistically significant differences in H. pylori seropositivity were not detected between children with or without atopy (8.4% vs. 12.6; p = 0.233). Although atopic disorders were more frequent in children exposed to traditional atopic risk factors, none of them showed to be significant after adjusting for all covariates. CONCLUSIONS: Serologically assessed H. pylori infection was not significantly associated with a reduced risk of atopic diseases in children.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Hipersensibilidade , Humanos , Itália/epidemiologia , Infecções por Helicobacter/epidemiologia , Infecções por Helicobacter/microbiologia , Feminino , Masculino , Criança , Helicobacter pylori/imunologia , Pré-Escolar , Hipersensibilidade/epidemiologia , Adolescente , Prevalência , Estudos Soroepidemiológicos , Anticorpos Antibacterianos/sangue , Estudos de Coortes , Fatores de Risco , Lactente , Inquéritos e Questionários , Imunoglobulina G/sangue , Asma/epidemiologia , Asma/imunologia
13.
Sensors (Basel) ; 24(16)2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39204849

RESUMO

Helicobacter pylori (H. pylori) is a common pathogen with a high prevalence of infection in human populations. The diagnosis of H. pylori infection is critical for its treatment, eradication, and prognosis. Biosensors have been demonstrated to be powerful for the rapid onsite detection of pathogens, particularly for point-of-care test (POCT) scenarios. In this work, we propose a novel optical biosensor, based on nanomaterial porous silicon (PSi) and photonic surface state Tamm Plasmon Polariton (TPP), for the detection of cytotoxin-associated antigen A (CagA) of H. pylori bacterium. We fabricated the PSi TPP biosensor, analyzed its optical characteristics, and demonstrated through experiments, with the sensing of the CagA antigen, that the TPP biosensor has a sensitivity of 100 pm/(ng/mL), a limit of detection of 0.05 ng/mL, and specificity in terms of positive-to-negative ratio that is greater than six. From these performance factors, it can be concluded that the TPP biosensor can serve as an effective tool for the diagnosis of H. pylori infection, either in analytical labs or in POCT applications.


Assuntos
Antígenos de Bactérias , Proteínas de Bactérias , Técnicas Biossensoriais , Helicobacter pylori , Silício , Técnicas Biossensoriais/métodos , Silício/química , Helicobacter pylori/imunologia , Helicobacter pylori/isolamento & purificação , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/análise , Proteínas de Bactérias/imunologia , Porosidade , Humanos , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/microbiologia
14.
Int J Mol Sci ; 25(11)2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38892046

RESUMO

Trained immunity is a concept in immunology in which innate immune cells, such as monocytes and macrophages, exhibit enhanced responsiveness and memory-like characteristics following initial contact with a pathogenic stimulus that may promote a more effective immune defense following subsequent contact with the same pathogen. Helicobacter pylori, a bacterium that colonizes the stomach lining, is etiologically associated with various gastrointestinal diseases, including gastritis, peptic ulcer, gastric adenocarcinoma, MALT lymphoma, and extra gastric disorders. It has been demonstrated that repeated exposure to H. pylori can induce trained immunity in the innate immune cells of the gastric mucosa, which become more responsive and better able to respond to subsequent H. pylori infections. However, interactions between H. pylori and trained immunity are intricate and produce both beneficial and detrimental effects. H. pylori infection is characterized histologically as the presence of both an acute and chronic inflammatory response called acute-on-chronic inflammation, or gastritis. The clinical outcomes of ongoing inflammation include intestinal metaplasia, gastric atrophy, and dysplasia. These same mechanisms may also reduce immunotolerance and trigger autoimmune pathologies in the host. This review focuses on the relationship between trained immunity and H. pylori and underscores the dynamic interplay between the immune system and the pathogen in the context of gastric colonization and inflammation.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Tolerância Imunológica , Imunidade Inata , Humanos , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/imunologia , Animais , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Gastrite/imunologia , Gastrite/microbiologia , Memória Imunológica , Imunidade Treinada
15.
Immunity ; 41(3): 451-464, 2014 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-25220211

RESUMO

Innate immune responses are critical for mucosal immunity. Here we describe an innate lymphocyte population, iCD8α cells, characterized by expression of CD8α homodimers. iCD8α cells exhibit innate functional characteristics such as the capacity to engulf and kill bacteria. Development of iCD8α cells depends on expression of interleukin-2 receptor γ chain (IL-2Rγc), IL-15, and the major histocompatibility complex (MHC) class Ib protein H2-T3, also known as the thymus leukemia antigen or TL. While lineage tracking experiments indicated that iCD8α cells have a lymphoid origin, their development was independent of the transcriptional suppressor Id2, suggesting that these cells do not belong to the family of innate lymphoid cells. Finally, we identified cells with a similar phenotype in humans, which were profoundly depleted in newborns with necrotizing enterocolitis. These findings suggest a critical role of iCD8α cells in immune responses associated with the intestinal epithelium.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos CD8/biossíntese , Imunidade nas Mucosas/imunologia , Mucosa Intestinal/citologia , Linfócitos/imunologia , Animais , Citrobacter rodentium/imunologia , Citocalasina D/farmacologia , Enterocolite Necrosante , Helicobacter pylori/imunologia , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Proteína 2 Inibidora de Diferenciação/genética , Subunidade gama Comum de Receptores de Interleucina/biossíntese , Interleucina-15/biossíntese , Interleucina-2/biossíntese , Interleucina-7/biossíntese , Mucosa Intestinal/imunologia , Ativação Linfocitária/imunologia , Linfócitos/classificação , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia
16.
J Immunol ; 206(6): 1161-1170, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33568397

RESUMO

Helicobacter pylori is the major etiological agent for most gastric cancer. CagA has been reported to be an important virulence factor of H. pylori, but its effect on the immune response is not yet clear. In this study, wild-type C57BL/6 mice and Ptpn6me-v/me-v mice were randomly assigned for infection with H. pylori We demonstrated that CagA suppressed H. pylori-stimulated expression of proinflammatory cytokines in vivo. Besides, we infected mouse peritoneal macrophages RAW264.7 and AGS with H. pylori Our results showed that CagA suppressed expression of proinflammatory cytokines through inhibiting the MAPKs and NF-κB pathways activation in vitro. Mechanistically, we found that CagA interacted with the host cellular tyrosine phosphatase SHP-1, which facilitated the recruitment of SHP-1 to TRAF6 and inhibited the K63-linked ubiquitination of TRAF6, which obstructed the transmission of signal downstream. Taken together, these findings reveal a previously unknown mechanism by which CagA negatively regulates the posttranslational modification of TRAF6 in innate antibacterial immune response and provide molecular basis for new therapeutics to treat microbial infection.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Animais , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Células HEK293 , Células HeLa , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/metabolismo , Humanos , Imunidade Inata , Lisina/metabolismo , Macrófagos Peritoneais , Masculino , Camundongos , Camundongos Transgênicos , Cultura Primária de Células , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/imunologia , Transfecção , Ubiquitinação/imunologia
17.
Semin Cell Dev Biol ; 101: 59-67, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32033828

RESUMO

Helicobacter pylori colonizes human stomach mucosa and its infection causes gastrointestinal diseases with variable severity. Bacterial infection stimulates autophagy, which is a part of innate immunity used to eliminate intracellular pathogens. Several intracellular bacteria have evolved multipronged strategies to circumvent this conserved system and thereby enhance their chance of intracellular survival. Nonetheless, studies on H. pylori have produced inconsistent results, showing either elevated or reduced clearance efficiency of intracellular bacteria through autophagy. In this review, we summarize recent studies on the mechanisms involved in autophagy induced by H. pylori and the fate of intracellular bacteria.


Assuntos
Mucosa Gástrica/imunologia , Helicobacter pylori/imunologia , Interações Hospedeiro-Patógeno/imunologia , Mucosa Gástrica/microbiologia , Humanos , Evasão da Resposta Imune
18.
Gastroenterology ; 160(4): 1256-1268.e9, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33189701

RESUMO

BACKGROUND & AIMS: Inflammation in the gastrointestinal tract may lead to the development of cancer. Dicarbonyl electrophiles, such as isolevuglandins (isoLGs), are generated from lipid peroxidation during the inflammatory response and form covalent adducts with amine-containing macromolecules. Thus, we sought to determine the role of dicarbonyl electrophiles in inflammation-associated carcinogenesis. METHODS: The formation of isoLG adducts was analyzed in the gastric tissues of patients infected with Helicobacter pylori from gastritis to precancerous intestinal metaplasia, in human gastric organoids, and in patients with colitis and colitis-associated carcinoma (CAC). The effect on cancer development of a potent scavenger of dicarbonyl electrophiles, 5-ethyl-2-hydroxybenzylamine (EtHOBA), was determined in transgenic FVB/N insulin-gastrin (INS-GAS) mice and Mongolian gerbils as models of H pylori-induced carcinogenesis and in C57BL/6 mice treated with azoxymethane-dextran sulfate sodium as a model of CAC. The effect of EtHOBA on mutations in gastric epithelial cells of H pylori-infected INS-GAS mice was assessed by whole-exome sequencing. RESULTS: We show increased isoLG adducts in gastric epithelial cell nuclei in patients with gastritis and intestinal metaplasia and in human gastric organoids infected with H pylori. EtHOBA inhibited gastric carcinoma in infected INS-GAS mice and gerbils and attenuated isoLG adducts, DNA damage, and somatic mutation frequency. Additionally, isoLG adducts were elevated in tissues from patients with colitis, colitis-associated dysplasia, and CAC as well as in dysplastic tumors of C57BL/6 mice treated with azoxymethane-dextran sulfate sodium. In this model, EtHOBA significantly reduced adduct formation, tumorigenesis, and dysplasia severity. CONCLUSIONS: Dicarbonyl electrophiles represent a link between inflammation and somatic genomic alterations and are thus key targets for cancer chemoprevention.


Assuntos
Transformação Celular Neoplásica/imunologia , Neoplasias Associadas a Colite/imunologia , Lipídeos/imunologia , Lesões Pré-Cancerosas/imunologia , Neoplasias Gástricas/imunologia , Animais , Benzilaminas/farmacologia , Benzilaminas/uso terapêutico , Núcleo Celular/metabolismo , Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias Associadas a Colite/microbiologia , Neoplasias Associadas a Colite/patologia , Neoplasias Associadas a Colite/prevenção & controle , Modelos Animais de Doenças , Células Epiteliais , Mucosa Gástrica/citologia , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/imunologia , Mucosa Gástrica/patologia , Gastrite/imunologia , Gastrite/microbiologia , Gastrite/patologia , Gerbillinae , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/imunologia , Helicobacter pylori/isolamento & purificação , Humanos , Lipídeos/antagonistas & inibidores , Metaplasia/imunologia , Metaplasia/microbiologia , Metaplasia/patologia , Camundongos , Camundongos Transgênicos , Organoides , Lesões Pré-Cancerosas/tratamento farmacológico , Lesões Pré-Cancerosas/microbiologia , Lesões Pré-Cancerosas/patologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/prevenção & controle
19.
Eur J Immunol ; 51(11): 2641-2650, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34398472

RESUMO

Helicobacter pylori is a Gram-negative bacterium found on the luminal surface of the gastric mucosa in at least 50% of the world's human population. The protective effect of breastfeeding against H. pylori infection has been extensively reported; however, the mechanisms behind this protection remain poorly understood. Human IgA from colostrum has reactivity against H. pylori antigens. Despite that IgA1 and IgA2 display structural and functional differences, their reactivity against H. pylori had not been previously determined. We attested titers and reactivity of human colostrum-IgA subclasses by ELISA, immunoblot, and flow cytometry. Colostrum samples from healthy mothers had higher titers of IgA; and IgA1 mostly recognized H. pylori antigens. Moreover, we found a correlation between IgA1 reactivity and their neutralizing effect determined by inhibition of cytoskeletal changes in AGS cells infected with H. pylori. In conclusion, colostrum-IgA reduces H. pylori infection of epithelial gastric cells, suggesting an important role in preventing the bacteria establishment during the first months of life. As a whole, these results suggest that IgA1 from human colostrum provides protection that may help in the development of the mucosal immune system of newborn children.


Assuntos
Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Colostro/imunologia , Helicobacter pylori/imunologia , Imunoglobulina A Secretora/imunologia , Citoesqueleto , Células Epiteliais , Feminino , Mucosa Gástrica/imunologia , Infecções por Helicobacter/imunologia , Humanos , Gravidez
20.
Int Immunol ; 33(9): 491-504, 2021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34297096

RESUMO

As a key virulence factor for persistent colonization, urease B subunit (UreB) is considered to be an ideal vaccine antigen against Helicobacter pylori infection. However, the role and molecular mechanisms of UreB involved in immune microenvironment dysregulation still remain largely unknown. In the present study, we evaluated the effects of UreB on macrophage activation and found that UreB induced PD-L1 accumulation on bone marrow-derived macrophages (BMDMs). Co-culture assays further revealed that UreB-induced PD-L1 expression on BMDMs significantly decreased the proliferation and secretion of cytolytic molecules (granzyme B and perforin) of splenic CD8+ T cells isolated from inactivated H. pylori-immunized mice. More importantly, using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and co-immunoprecipitation techniques, it has been confirmed that myosin heavy chain 9 (Myh9) is a direct membrane receptor for UreB and is required for PD-L1 up-regulation on BMDMs. Molecular studies further demonstrated that the interaction between UreB and Myh9 decreased GCN2 autophosphorylation and enhanced the intracellular pool of amino acids, leading to the up-regulation of S6K phosphorylation, a commonly used marker for monitoring activation of mTORC1 signaling activity. Furthermore, blocking mTORC1 activation with its inhibitor Temsirolimus reversed the UreB-induced PD-L1 up-regulation and the subsequent inhibitory effects of BMDMs on activation of cytotoxic CD8+ T-cell responses. Overall, our data unveil a novel immunosuppressive mechanism of UreB during H. pylori infection, which may provide valuable clues for the optimization of H. pylori vaccine.


Assuntos
Antígeno B7-H1/imunologia , Proteínas de Bactérias/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Cadeias Pesadas de Miosina/imunologia , Urease/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7 , Células THP-1
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa