Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.155
Filtrar
1.
Food Chem ; 448: 139157, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38569411

RESUMEN

About half of the world's population is infected with the bacterium Helicobacter pylori. For colonization, the bacterium neutralizes the low gastric pH and recruits immune cells to the stomach. The immune cells secrete cytokines, i.e., the pro-inflammatory IL-17A, which directly or indirectly damage surface epithelial cells. Since (I) dietary proteins are known to be digested into bitter tasting peptides in the gastric lumen, and (II) bitter tasting compounds have been demonstrated to reduce the release of pro-inflammatory cytokines through functional involvement of bitter taste receptors (TAS2Rs), we hypothesized that the sweet-tasting plant protein thaumatin would be cleaved into anti-inflammatory bitter peptides during gastric digestion. Using immortalized human parietal cells (HGT-1 cells), we demonstrated a bitter taste receptor TAS2R16-dependent reduction of a H. pylori-evoked IL-17A release by up to 89.7 ± 21.9% (p ≤ 0.01). Functional involvement of TAS2R16 was demonstrated by the study of specific antagonists and siRNA knock-down experiments.


Asunto(s)
Helicobacter pylori , Interleucina-17 , Proteínas de Plantas , Receptores Acoplados a Proteínas G , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Interleucina-17/metabolismo , Interleucina-17/genética , Interleucina-17/inmunología , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/química , Gusto , Digestión , Péptidos/farmacología , Péptidos/química , Péptidos/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/inmunología , Línea Celular
2.
Nature ; 628(8009): 854-862, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38570678

RESUMEN

The intestinal immune system is highly adapted to maintaining tolerance to the commensal microbiota and self-antigens while defending against invading pathogens1,2. Recognizing how the diverse network of local cells establish homeostasis and maintains it in the complex immune environment of the gut is critical to understanding how tolerance can be re-established following dysfunction, such as in inflammatory disorders. Although cell and molecular interactions that control T regulatory (Treg) cell development and function have been identified3,4, less is known about the cellular neighbourhoods and spatial compartmentalization that shapes microorganism-reactive Treg cell function. Here we used in vivo live imaging, photo-activation-guided single-cell RNA sequencing5-7 and spatial transcriptomics to follow the natural history of T cells that are reactive towards Helicobacter hepaticus through space and time in the settings of tolerance and inflammation. Although antigen stimulation can occur anywhere in the tissue, the lamina propria-but not embedded lymphoid aggregates-is the key microniche that supports effector Treg (eTreg) cell function. eTreg cells are stable once their niche is established; however, unleashing inflammation breaks down compartmentalization, leading to dominance of CD103+SIRPα+ dendritic cells in the lamina propria. We identify and validate the putative tolerogenic interaction between CD206+ macrophages and eTreg cells in the lamina propria and identify receptor-ligand pairs that are likely to govern the interaction. Our results reveal a spatial mechanism of tolerance in the lamina propria and demonstrate how knowledge of local interactions may contribute to the next generation of tolerance-inducing therapies.


Asunto(s)
Helicobacter hepaticus , Tolerancia Inmunológica , Cadenas alfa de Integrinas , Análisis de la Célula Individual , Linfocitos T Reguladores , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/citología , Animales , Ratones , Cadenas alfa de Integrinas/metabolismo , Helicobacter hepaticus/inmunología , Tolerancia Inmunológica/inmunología , Femenino , Masculino , Antígenos CD/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/inmunología , Inflamación/inmunología , Inflamación/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/citología , Intestinos/inmunología , Intestinos/citología , Ratones Endogámicos C57BL , RNA-Seq , Transcriptoma , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Células Dendríticas/inmunología
3.
Immunohorizons ; 8(4): 339-353, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38639570

RESUMEN

Helicobacter pylori is a Gram-negative pathogen that colonizes the stomach, induces inflammation, and drives pathological changes in the stomach tissue, including gastric cancer. As the principal cytokine produced by Th17 cells, IL-17 mediates protective immunity against pathogens by inducing the activation and mobilization of neutrophils. Whereas IL-17A is largely produced by lymphocytes, the IL-17 receptor is expressed in epithelial cells, fibroblasts, and hematopoietic cells. Loss of the IL-17RA in mice results in impaired antimicrobial responses to extracellular bacteria. In the context of H. pylori infection, this is compounded by extensive inflammation in Il17ra-/- mice. In this study, Foxa3creIl17rafl/fl (Il17raΔGI-Epi) and Il17rafl/fl (control) mice were used to test the hypothesis that IL-17RA signaling, specifically in epithelial cells, protects against severe inflammation after H. pylori infection. The data indicate that Il17raΔGI-Epi mice develop increased inflammation compared with controls. Despite reduced Pigr expression, levels of IgA increased in the gastric wash, suggesting significant increase in Ag-specific activation of the T follicular helper/B cell axis. Gene expression analysis of stomach tissues indicate that both acute and chronic responses are significantly increased in Il17raΔGI-Epi mice compared with controls. These data suggest that a deficiency of IL-17RA in epithelial cells is sufficient to drive chronic inflammation and hyperactivation of the Th17/T follicular helper/B cell axis but is not required for recruitment of polymorphonuclear neutrophils. Furthermore, the data suggest that fibroblasts can produce chemokines in response to IL-17 and may contribute to H. pylori-induced inflammation through this pathway.


Asunto(s)
Infecciones por Helicobacter , Receptores de Interleucina-17 , Animales , Ratones , Células Epiteliales/metabolismo , Infecciones por Helicobacter/inmunología , Helicobacter pylori , Inflamación/metabolismo , Interleucina-17/metabolismo , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/metabolismo
4.
J Gastroenterol Hepatol ; 38(4): 625-633, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36740832

RESUMEN

BACKGROUND: Helicobacter pylori, a gram-negative bacterium persisting on the gastric mucosa, is involved in the pathogenesis of a variety of gastric diseases. Leukocyte cell-derived chemotaxin 2 (LECT2) treatment increased the phagocytic capacity of lymphocytes and improved immune function in bacterial infection. Whether the immune cells infected with H. pylori are affected by LECT2 is unclear. METHODS: Bone marrow-derived dendritic cells (BMDCs) from wild-type C57BL/6 mice, CD209a knockout mice, or LECT2 knockout mice were exposed to H. pylori at a multiplicity of infection of 10 for 24 h. The maturity of DCs and the cytokines secreted by DCs were analyzed by flow cytometry, western blot, and real-time PCR. The signaling pathway underlying CD209a activation after LECT2 treatment were also detected. RESULTS: LECT2 treatment promoted H. pylori-induced BMDC maturation and produced a high level of anti-inflammatory cytokine (IL-10) but a low level of pro-inflammatory cytokine (IL-23p40). Moreover, LECT2-pretreated DCs shifted the development of pro-inflammatory Th1/Th17 cells to Treg cells. CD209a mediated LECT2-induced maturation and secretion of DC in H. pylori-primed BMDCs. LECT2 was further confirmed to induce the secretion of certain cytokines via CD209a-JNK/P38 MAPK pathway. CONCLUSION: This study reveals that LECT2 modulated the functions of H. pylori-primed DCs in a CD209a-dependent manner, which might hinder the clearance of H. pylori and contribute to its colonization.


Asunto(s)
Células Dendríticas , Infecciones por Helicobacter , Helicobacter pylori , Péptidos y Proteínas de Señalización Intercelular , Receptores de Superficie Celular , Animales , Ratones , Citocinas/metabolismo , Células Dendríticas/inmunología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo
5.
J Immunol Res ; 2022: 3861518, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36317079

RESUMEN

Helicobacter pylori colonization and persistence could precede gastric adenocarcinoma. Elucidating immune recognition strategies of H. pylori is therefore imperative to curb chronic persistence in the human host. Toll-like receptor 7 (TLR7) and TLR8 are widely known as viral single-stranded RNA (ssRNA) sensors yet less studied in the bacteria context. Here, we investigated the involvement of these receptors in the immunity to H. pylori. Human THP-1 monocytic cells were infected with H. pylori, and the expression levels of human Toll-like receptors (TLRs) were examined. The roles of TLR7 and TLR8 in response to H. pylori infection were further investigated using receptor antagonists. Among all TLR transcripts examined, TLR8 exhibited the most prominent upregulation, followed by TLR7 in the THP-1 cells infected with H. pylori J99 or SS1 strains. H. pylori infection-mediated IFN-α and IFN-ß transactivation was significantly abrogated by the TLR7/8 (but not TLR7) antagonist. Additionally, TLR7/8 antagonist treatment reduced H. pylori infection-mediated phosphorylation of interferon regulatory factor 7 (IRF7). Our study suggests a novel role of TLR8 signaling in host immunity against H. pylori through sensing live bacteria to elicit the production of type I interferon.


Asunto(s)
Infecciones por Helicobacter , Interferón Tipo I , Monocitos , Receptor Toll-Like 8 , Humanos , Infecciones por Helicobacter/inmunología , Helicobacter pylori/metabolismo , Interferón Tipo I/metabolismo , Monocitos/inmunología , Receptor Toll-Like 8/metabolismo
6.
Int J Mol Sci ; 23(5)2022 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-35269560

RESUMEN

Gastric cancer is a leading cause of cancer-related death, and a large proportion of cases are inseparably linked to infections with the bacterial pathogen and type I carcinogen Helicobacter pylori. The development of gastric cancer follows a cascade of transformative tissue events in an inflammatory environment. Proteases of host origin as well as H. pylori-derived proteases contribute to disease progression at every stage, from chronic gastritis to gastric cancer. In the present article, we discuss the importance of (metallo-)proteases in colonization, epithelial inflammation, and barrier disruption in tissue transformation, deregulation of cell proliferation and cell death, as well as tumor metastasis and neoangiogenesis. Proteases of the matrix metalloproteinase (MMP) and a disintegrin and metalloproteinase domain-containing protein (ADAM) families, caspases, calpain, and the H. pylori proteases HtrA, Hp1012, and Hp0169 cleave substrates including extracellular matrix molecules, chemokines, and cytokines, as well as their cognate receptors, and thus shape the pathogenic microenvironment. This review aims to summarize the current understanding of how proteases contribute to disease progression in the gastric compartment.


Asunto(s)
Infecciones por Helicobacter/inmunología , Helicobacter pylori/patogenicidad , Péptido Hidrolasas/metabolismo , Neoplasias Gástricas/patología , Proteínas Bacterianas/metabolismo , Progresión de la Enfermedad , Regulación de la Expresión Génica , Infecciones por Helicobacter/complicaciones , Helicobacter pylori/inmunología , Humanos , Metaloproteasas/metabolismo , Proteolisis , Serina Proteasas/metabolismo , Neoplasias Gástricas/microbiología
7.
Mediators Inflamm ; 2022: 2944156, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35300405

RESUMEN

Helicobacter pylori is a spiral-shaped gram-negative bacterium. Its infection is mainly transmitted via oral-oral and fecal-oral routes usually during early childhood. It can achieve persistent colonization by manipulating the host immune responses, which also causes mucosal damage and inflammation. H. pylori gastritis is an infectious disease and results in chronic gastritis of different severity in near all patients with infection. It may develop from acute/chronic inflammation, chronic atrophic gastritis, intestinal metaplasia, dysplasia, and intraepithelial neoplasia, eventually to gastric cancer. This review attempts to cover recent studies which provide important insights into how H. pylori causes chronic inflammation and what the characteristic is, which will immunologically explain H. pylori gastritis.


Asunto(s)
Gastritis Atrófica , Gastritis , Infecciones por Helicobacter , Helicobacter pylori , Gastritis/inmunología , Gastritis/microbiología , Gastritis Atrófica/inmunología , Gastritis Atrófica/microbiología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Humanos , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/microbiología
8.
Gut Microbes ; 14(1): 2044721, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35289715

RESUMEN

Helicobacter pylori is the major risk factor for gastric cancer. H. pylori harboring the type IV secretion system (T4SS) and its effector CagA encoded on the cag pathogenicity Island (cagPAI) increases the risk. H. pylori PMSS1 has a multi-cagA genotype, modulating cagA copy number dynamically from zero to four copies. To examine the effect of the immune response on cagA copy number change, we utilized a mouse model with different immune status. PMSS1 recovered from Rag1-/- mice, lacking functional T or B cells, retained more cagA copies. PMSS1 recovered from Il10-/- mice, showing intense inflammation, had fewer cagA copies compared to those recovered from wild-type mice. Moreover, cagA copy number of PMSS1 recovered from wild-type and Il10-/- mice was positively correlated with the capacity to induce IL-8 secretion at four weeks of infection. Since recombination in cagY influences T4SS function, including CagA translocation and IL-8 induction, we constructed a multiple linear regression model to predict H. pylori-induced IL-8 expression based on cagA copy number and cagY recombination status; H. pylori induces more IL-8 secretion when the strain has more cagA copies and intact cagY. This study shows that H. pylori PMSS1 in mice with less intense immune response possess higher cagA copy number than those infected in mice with more intense immune response and thus the multi-cagA genotype, along with cagY recombination, functions as an immune-sensitive regulator of H. pylori virulence.


Asunto(s)
Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Microbioma Gastrointestinal , Infecciones por Helicobacter , Helicobacter pylori , Animales , Proteínas Bacterianas/metabolismo , Variaciones en el Número de Copia de ADN , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Inmunidad , Interleucina-10/genética , Interleucina-8/metabolismo , Ratones , Virulencia
9.
Cell Rep ; 38(6): 110359, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35139377

RESUMEN

The two human pathogens Helicobacter pylori and Mycobacterium tuberculosis (Mtb) co-exist in many geographical areas of the world. Here, using a co-infection model of H. pylori and the Mtb relative M. bovis bacillus Calmette-Guérin (BCG), we show that both bacteria affect the colonization and immune control of the respective other pathogen. Co-occurring M. bovis boosts gastric Th1 responses and H. pylori control and aggravates gastric immunopathology. H. pylori in the stomach compromises immune control of M. bovis in the liver and spleen. Prior antibiotic H. pylori eradication or M. bovis-specific immunization reverses the effects of H. pylori. Mechanistically, the mutual effects can be attributed to the redirection of regulatory T cells (Treg cells) to sites of M. bovis infection. Reversal of Treg cell redirection by CXCR3 blockade restores M. bovis control. In conclusion, the simultaneous presence of both pathogens exacerbates the problems associated with each individual infection alone and should possibly be factored into treatment decisions.


Asunto(s)
Helicobacter pylori/patogenicidad , Infecciones por Mycobacterium/microbiología , Mycobacterium tuberculosis/patogenicidad , Linfocitos T Reguladores/microbiología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Ratones Endogámicos C57BL , Mycobacterium bovis/patogenicidad , Mycobacterium tuberculosis/inmunología
10.
Front Immunol ; 13: 789379, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154106

RESUMEN

Autoimmunity prevalence, as measured by antinuclear antibodies (ANA), is increasing in U.S. adolescents. Improved hygiene and cleaner environments in childhood may reduce exposure to infections and other immune challenges, resulting in improper immune responses to later-life exposures. We examined associations of hygiene hypothesis indicators, including asthma, allergies, and antibodies to infectious agents, with ANA prevalence, measured by HEp-2 immunofluorescence, in adolescents (aged 12-19 years) over a 25-year time span in the National Health and Nutrition Examination Survey (NHANES) (N=2,709), adjusting for age, sex, race/ethnicity, body mass index, education and survey cycle, overall and within individual time periods, using logistic regression. Prevalence of ANA in adolescents increased from 5.0% in 1988-1991 to 12.8% in 2011-2012. ANA were positively associated with diagnosis of asthma in early childhood (OR: 2.07, CI: 1.09-3.99) and the effect estimate for current hay fever was elevated but not statistically significant (OR: 1.55, CI: 0.85-2.84). Fewer than 2% of those with ANA in 1988-1991 had been diagnosed with asthma, compared with 18% in 1999-2000, and 27% in 2003-2004 and 2011-2012. ANA trended negatively with Helicobacter pylori antibodies (OR: 0.49, CI: 0.24-0.99). ANA may be useful as an additional indicator of inadequate immune education in adolescence, a critical period of growth and development.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Asma/epidemiología , Asma/inmunología , Autoinmunidad , Hipótesis de la Higiene , Higiene , Adolescente , Asma/diagnóstico , Niño , Estudios Transversales , Femenino , Infecciones por Helicobacter/epidemiología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Herpes Simple/epidemiología , Herpes Simple/inmunología , Herpes Simple/virología , Herpesvirus Humano 1/inmunología , Humanos , Masculino , Prevalencia , Autoinforme , Toxoplasma/inmunología , Toxoplasmosis/epidemiología , Toxoplasmosis/inmunología , Toxoplasmosis/parasitología , Estados Unidos/epidemiología , Adulto Joven
11.
Helicobacter ; 27(2): e12875, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35092634

RESUMEN

BACKGROUND: Helicobacter pylori (H. pylori) colonizes the human gastric mucosa with a high worldwide prevalence. Currently, H. pylori is eradicated by the use of antibiotics. However, elevated antibiotic resistance suggests new therapeutic strategies need to be envisioned: one approach being prophylactic vaccination. Pre-clinical and clinical data show that a urease-based vaccine is efficient in decreasing H. pylori infection through the mobilization of T helper (Th) cells, especially Th17 cells. Th17 cells produce interleukins such as IL-22 and IL-17, among others, and are key players in vaccine efficacy. Recently, granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing Th17 cells have been identified. AIM: This study explores the possibility that GM-CSF plays a role in the reduction of H. pylori infection following vaccination. RESULTS: We demonstrate that GM-CSF+ IL-17+ Th17 cells accumulate in the stomach mucosa of H. pylori infected mice during the vaccine-induced reduction of H. pylori infection. Secondly, we provide evidence that vaccinated GM-CSF deficient mice only modestly reduce H. pylori infection. Conversely, we observe that an increase in GM-CSF availability reduces H. pylori burden in chronically infected mice. Thirdly, we show that GM-CSF, by acting on gastric epithelial cells, promotes the production of ßdefensin3, which exhibits H. pylori bactericidal activities. CONCLUSION: Taken together, we demonstrate a key role of GM-CSF, most probably originating from Th17 cells, in the vaccine-induced reduction of H. pylori infection.


Asunto(s)
Vacunas Bacterianas , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Infecciones por Helicobacter , Helicobacter pylori , Animales , Vacunas Bacterianas/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/prevención & control , Ratones , Células Th17 , Vacunación
12.
J Clin Lab Anal ; 36(1): e24069, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34783398

RESUMEN

BACKGROUND: This study aimed to investigate the implementation and quality control of the quantitative detection of serum Helicobacter pylori (H. pylori) antibody in clinical laboratories in China. METHODS: Online external quality assessment (EQA) questionnaires were distributed to the clinical laboratories by National Center for Clinical Laboratories (NCCL) of China. We collected information on the quantitative detection procedures of serum H. pylori antibody in clinical laboratories, including detection reagents, methods, instruments, calibrators, and internal quality control (IQC). We distributed quality control products to some select laboratories that conducted quantitative detection and analyzed the obtained test data. We evaluated the quantitative detection procedure based on the standard evaluation criteria set at a target value of ±30%. RESULTS: 70.9% (146/206) of the laboratories conducted quantitative detection of H. pylori antibody; 29.1% (60/206) of the laboratories performed qualitative detection. Domestic reagents and matching calibrators accounted for more than 97.1% (200/206) of all reagents. Latex-enhanced immunoturbidimetry was used in 89.7% (131/146) of the laboratories for quantitative determination, while the colloidal gold method was used in 66.7% (40/60) of the laboratories for qualitative determination. A total of 130 laboratories participated in the EQA; 123 completed the assessment, and the pass rate was 75.6% (93/123). CONCLUSION: Clinical quantitative detection of serum H. pylori antibody is performed at a high rate in China. Thus, further studies on the specificity of commercial detection reagents are needed. EQAs are useful to monitor and improve the detection quality of H. pylori antibodies.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Helicobacter pylori/inmunología , Laboratorios Clínicos , China , Infecciones por Helicobacter/diagnóstico , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Humanos , Inmunoturbidimetría/normas , Laboratorios Clínicos/normas , Laboratorios Clínicos/estadística & datos numéricos , Control de Calidad , Encuestas y Cuestionarios
13.
Int J Mol Sci ; 22(23)2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34884957

RESUMEN

The identification of novel strategies to control Helicobacter pylori (Hp)-associated chronic inflammation is, at present, a considerable challenge. Here, we attempt to combat this issue by modulating the innate immune response, targeting formyl peptide receptors (FPRs), G-protein coupled receptors that play key roles in both the regulation and the resolution of the innate inflammatory response. Specifically, we investigated, in vitro, whether Caulerpin-a bis-indole alkaloid isolated from algae of the genus Caulerpa-could act as a molecular antagonist scaffold of FPRs. We showed that Caulerpin significantly reduces the immune response against Hp culture filtrate, by reverting the FPR2-related signaling cascade and thus counteracting the inflammatory reaction triggered by Hp peptide Hp(2-20). Our study suggests Caulerpin to be a promising therapeutic or adjuvant agent for the attenuation of inflammation triggered by Hp infection, as well as its related adverse clinical outcomes.


Asunto(s)
Proteínas Bacterianas/farmacología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/metabolismo , Indoles/farmacología , Fragmentos de Péptidos/farmacología , Receptores de Formil Péptido/metabolismo , Receptores de Lipoxina/metabolismo , Proteínas Bacterianas/inmunología , Línea Celular , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Indoles/química , Modelos Moleculares , Fragmentos de Péptidos/inmunología , Unión Proteica , Receptores de Formil Péptido/química , Receptores de Lipoxina/química , Transducción de Señal/efectos de los fármacos , Células THP-1
14.
Immunity ; 54(12): 2812-2824.e4, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34861182

RESUMEN

The composition of the intestinal microbiota is associated with both the development of tumors and the efficacy of anti-tumor immunity. Here, we examined the impact of microbiota-specific T cells in anti-colorectal cancer (CRC) immunity. Introduction of Helicobacter hepaticus (Hhep) in a mouse model of CRC did not alter the microbial landscape but increased tumor infiltration by cytotoxic lymphocytes and inhibited tumor growth. Anti-tumor immunity was independent of CD8+ T cells but dependent upon CD4+ T cells, B cells, and natural killer (NK) cells. Hhep colonization induced Hhep-specific T follicular helper (Tfh) cells, increased the number of colon Tfh cells, and supported the maturation of Hhep+ tumor-adjacent tertiary lymphoid structures. Tfh cells were necessary for Hhep-mediated tumor control and immune infiltration, and adoptive transfer of Hhep-specific CD4+ T cells to Tfh cell-deficient Bcl6fl/flCd4Cre mice restored anti-tumor immunity. Thus, introduction of immunogenic intestinal bacteria can promote Tfh-associated anti-tumor immunity in the colon, suggesting therapeutic approaches for the treatment of CRC.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Colon/patología , Neoplasias Colorrectales/inmunología , Microbioma Gastrointestinal/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter hepaticus/fisiología , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Células T Auxiliares Foliculares/inmunología , Estructuras Linfoides Terciarias/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo
15.
Future Microbiol ; 16: 1229-1238, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34615380

RESUMEN

Helicobacter pylori (H. pylori) infection is highly prevalent, affecting 4.4 billion people globally. This pathogen is a risk factor in the pathogenesis of more than 75% of worldwide cases of gastric cancer. Pattern recognition receptors are essential in the innate immune response to H. pylori infection. They recognize conserved pathogen structures and myriad alarmins released by host cells in response to microbial components, cytokines or cellular stress, thus triggering a robust proinflammatory response, which is crucial in H. pylori-induced gastric carcinogenesis. In this review, we intend to highlight the main pattern recognition receptors involved in the recognition and host response to H. pylori, as well as the main structures recognized and the subsequent inflammatory response.


Asunto(s)
Infecciones por Helicobacter , Receptores de Reconocimiento de Patrones , Infecciones por Helicobacter/inmunología , Helicobacter pylori , Humanos , Receptores de Reconocimiento de Patrones/inmunología
16.
FASEB J ; 35(11): e21942, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34670018

RESUMEN

Atherosclerosis is a chronic inflammatory disease. Pathophysiological similarities between chronic infections and atherosclerosis triggered interests between these conditions. The seroepidemiological study showed that Helicobacter pylori strains that express cytotoxin-associated gene A (CagA), an oncoprotein and a major virulence factor, was positively correlated with atherosclerosis and related clinical events. Nevertheless, the underlying mechanism is poorly understood. In this study, the seroprevalence of infection by H. pylori and by strains express CagA assessed by enzyme-linked immunosorbent assay (ELISA) showed that the prevalence of CagA strains rather than H. pylori in patients was positively correlated with atherogenesis. Correspondingly, we found that CagA augmented the growth of plaque of ApoE-/- mice in the early stage of atherosclerosis and promoted the expression of adhesion molecules and inflammatory cytokines in mouse aortic endothelial cells (MAECs). Mechanistically, both si-NLRP3 and si-IL-1ß mitigated the promoting effect of CagA on the inflammatory activation of HAECs. In vivo, the inhibition of NLRP3 by MCC950 significantly attenuated the promoting effect of CagA on plaque growth of ApoE-/- mice. We also propose NLRP3 as a potential therapeutic target for CagA-positive H. pylori infection-related atherosclerosis and emphasize the importance of inflammation in atherosclerosis pathology.


Asunto(s)
Antígenos Bacterianos/metabolismo , Aorta/patología , Aterosclerosis/sangre , Proteínas Bacterianas/metabolismo , Caspasa 1/metabolismo , Células Endoteliales/metabolismo , Infecciones por Helicobacter/sangre , Helicobacter pylori/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Placa Aterosclerótica/sangre , Anciano , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Aorta/metabolismo , Aterosclerosis/microbiología , Proteínas Bacterianas/inmunología , Modelos Animales de Enfermedad , Femenino , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Persona de Mediana Edad , Placa Aterosclerótica/microbiología , Estudios Seroepidemiológicos , Células THP-1
17.
Front Immunol ; 12: 702156, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34707599

RESUMEN

Podoplanin (Pdpn) is a mucin-type transmembrane protein that has been implicated in multiple physiological settings including lymphangiogenesis, platelet aggregation, and cancer metastasis. Here, we reported an absence of Pdpn transcript expression in the resting mouse monocytic macrophages, RAW264.7 cells; intriguingly, a substantial upregulation of Pdpn was observed in activated macrophages following Helicobacter pylori or lipopolysaccharide stimulation. Pdpn-knockout macrophages demonstrated intact phagocytic and intracellular bactericidal activities comparable to wild type but exhibited impaired migration due to attenuated filopodia formation. In contrast, an ectopic expression of Pdpn augmented filopodia protrusion in activated macrophages. NanoString analysis uncovered a close dependency of Filamin C gene on the presence of Pdpn, highlighting an involvement of Filamin C in modulation of actin polymerization activity, which controls cell filopodia formation and migration. In addition, interleukin-1ß production was significantly declined in the absence of Pdpn, suggesting a role of Pdpn in orchestrating inflammation during H. pylori infection besides cellular migration. Together, our findings unravel the Pdpn network that modulates movement of active macrophages.


Asunto(s)
Movimiento Celular/inmunología , Filaminas/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Animales , Infecciones por Helicobacter/inmunología , Helicobacter pylori , Humanos , Ratones , Células RAW 264.7 , Células THP-1
18.
Front Immunol ; 12: 736269, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34589088

RESUMEN

Gastric CD4+T cells contribute to Helicobacter pylori (H. pylori)-induced gastritis by amplifying mucosal inflammation and exacerbating mucosal injuries. However, the pathogenic CD4+ T cell subset involved in gastritis and the potential regulators are still unclear. Here we identified an IL-21-producing gastric CD4+T cell subset, which exhibited tissue-resident CXCR5-BTLA-PD-1hi TFH-like phenotype in H. pylori-positive gastritis patients. Meanwhile, we identified glucocorticoid-induced tumor necrosis factor receptor (GITR) as an important regulator to facilitate IL-21 production by CD4+T cells and accelerate mucosal inflammation in gastritis patients with H. pylori infection. Moreover, GITR expression was increased in gastric CD4+T cells of gastritis patients compared to healthy controls, along with the upregulated expression of its ligand GITRL in mucosal macrophages (Mϕ) of gastritis patients. Further observations showed that the activation of GITR/GITRL signal promoted the IL-21 production of CD4+T cells via the STAT3 pathway. Besides this, IL-21 from CD4+T cells induced the proliferation of B cell and promoted the production of inflammatory cytokines IL-1ß and IL-6 and chemokines MIP-3α and CCL-25 as well as matrix metalloproteinase (MMP)-3 and MMP-9 by human gastric epithelial cells, suggesting the facilitating effect of IL-21-producing CD4+T cells on mucosal inflammation and injuries. Taking these data together, we revealed that GITR/GITRL signal promoted the polarization of mucosal IL-21-producing CD4+T cells in H. pylori-positive gastritis, which may provide therapeutic strategies for the clinical treatment of H. pylori-induced gastritis.


Asunto(s)
Mucosa Gástrica/metabolismo , Gastritis/metabolismo , Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/patogenicidad , Inmunidad Mucosa , Interleucinas/metabolismo , Células T Auxiliares Foliculares/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Técnicas de Cocultivo , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Mucosa Gástrica/inmunología , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Gastritis/diagnóstico , Gastritis/inmunología , Gastritis/microbiología , Infecciones por Helicobacter/diagnóstico , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Ligandos , Fenotipo , Transducción de Señal , Células T Auxiliares Foliculares/inmunología , Factores de Necrosis Tumoral/metabolismo
19.
J Microbiol Methods ; 188: 106298, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34364886

RESUMEN

We assessed the impact of Helicobacter pylori seropositivity on recombinant antigen-based Lyme serology. We compared the IgG ELISA+LIA (line immunoassay) reactivity of anti-Helicobacter IgG positive and negative samples. The ELISA S/Co values and LIA band numbers were identical. Our results suggest that Helicobacter seropositivity lacks an apparent effect on Lyme disease test reactivity.


Asunto(s)
Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Infecciones por Helicobacter/inmunología , Helicobacter pylori/genética , Enfermedad de Lyme/inmunología , Adulto , Anciano , Anticuerpos Antibacterianos/sangre , Infecciones por Helicobacter/diagnóstico , Helicobacter pylori/aislamiento & purificación , Humanos , Inmunoensayo/métodos , Inmunoglobulina G , Inmunoglobulina M , Enfermedad de Lyme/diagnóstico , Masculino , Persona de Mediana Edad , Proteínas Recombinantes
20.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34360770

RESUMEN

Eosinophils are granulocytes primarily associated with TH2 responses to parasites or immune hyper-reactive states, such as asthma, allergies, or eosinophilic esophagitis. However, it does not make sense from an evolutionary standpoint to maintain a cell type that is only specific for parasitic infections and that otherwise is somehow harmful to the host. In recent years, there has been a shift in the perception of these cells. Eosinophils have recently been recognized as regulators of immune homeostasis and suppressors of over-reactive pro-inflammatory responses by secreting specific molecules that dampen the immune response. Their role during parasitic infections has been well investigated, and their versatility during immune responses to helminths includes antigen presentation as well as modulation of T cell responses. Although it is known that eosinophils can present antigens during viral infections, there are still many mechanistic aspects of the involvement of eosinophils during viral infections that remain to be elucidated. However, are eosinophils able to respond to bacterial infections? Recent literature indicates that Helicobacter pylori triggers TH2 responses mediated by eosinophils; this promotes anti-inflammatory responses that might be involved in the long-term persistent infection caused by this pathogen. Apparently and on the contrary, in the respiratory tract, eosinophils promote TH17 pro-inflammatory responses during Bordetella bronchiseptica infection, and they are, in fact, critical for early clearance of bacteria from the respiratory tract. However, eosinophils are also intertwined with microbiota, and up to now, it is not clear if microbiota regulates eosinophils or vice versa, or how this connection influences immune responses. In this review, we highlight the current knowledge of eosinophils as regulators of pro and anti-inflammatory responses in the context of both infection and naïve conditions. We propose questions and future directions that might open novel research avenues in the future.


Asunto(s)
Infecciones por Bordetella/inmunología , Bordetella bronchiseptica/inmunología , Eosinófilos/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Microbiota/inmunología , Animales , Humanos , Células Th17/inmunología , Células Th2/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...