Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Clin Immunol ; 252: 109648, 2023 07.
Article in English | MEDLINE | ID: mdl-37209806

ABSTRACT

Helicobacter pylori infection is involved in development of diverse gastro-pathologies. Our aim is to investigate potential signature of cytokines-chemokine levels (IL-17A, IL-1ß, and CXCL-8) in H. pylori-infected patients and their impact on immune response in both corpus and antrum. Multivariate level analysis with machine learning model were carried out using cytokines/chemokine levels of infected Moroccan patients. In addition, Geo dataset was used to run enrichment analysis following CXCL-8 upregulation. Our analysis showed that combination of cytokines-chemokine levels allowed prediction of positive H. pylori density score with <5% of miss-classification error, with fundus CXCL-8 being the most important variable for this discrimination. Furthermore, CXCL-8 dependent expression profile was mainly associated to IL6/JAK/STAT3 signaling in the antrum, interferons alpha and gamma responses in the corpus and commonly induced transcriptional /proliferative activities. To conclude, CXCL-8 level might be a signature of Moroccan H. pylori-infected patients and an inducer of regional-dependent immune response at the gastric level. Larger trials must be carried out to validate the relevance of these results for diverse populations.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Humans , Cytokines/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter pylori/metabolism , Immunity , Stomach/pathology
2.
Cytokine ; 165: 156172, 2023 05.
Article in English | MEDLINE | ID: mdl-36924609

ABSTRACT

The COVID-19 pandemic has caused millions of deaths and has resulted in disastrous societal and economic impacts worldwide. During SARS-CoV-2 infection, abnormal levels of pro-inflammatory cytokines have been observed and were associated to the severity of the disease. Type I (-α/ß) and Type III (IFN-λ) interferons are family members of cytokines that play an important role in fighting viral replication during the early phases of infection. The location and timing of the IFNs production have been shown to be decisive for the COVID-19 outcome. Despite the effectiveness of COVID-19 vaccines and with the emergence of new SARS-CoV-2 variants, a better understanding of the involvement of IFNs as players in antiviral immunity in the COVID-19 pathophysiology is necessary to implement additional potent prophylactic and/or therapeutic approaches. In this study, we investigated the role of type I and III IFN in COVID-19 pathophysiology. We first analyzed the IFN-α, IFN-ß and IFN- λ mRNA expression in nasopharyngeal swabs and blood samples from Moroccan patients infected with SARS-CoV-2 and secondly correlated these IFNs expressions with COVID-19 clinical and biological parameters. Our results showed that in the upper airways of patients with mild, non-severe, or severe COVID-19 manifestations, the IFN- α, - ß and - λ are expressed in the same manner as in controls. However, in blood samples their expression was downregulated in all groups. Univariate linear models with interferons as predictors to evaluate clinical-biological parameters highlighted that the main clinical-biological relations were found when testing: FiO2, Lymphocyte values and virus load. Furthermore, the multivariate models confirmed that quantifications of interferons during COVID-19 are good biological markers for tracking COVID-19 pathophysiology.


Subject(s)
COVID-19 , Interferon Type I , Humans , Interferons , COVID-19 Vaccines , Pandemics , SARS-CoV-2 , Antiviral Agents , Cytokines , Interferon-alpha , Interferon Lambda
3.
Microorganisms ; 11(2)2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36838318

ABSTRACT

Genetic polymorphisms at the IL-1 cluster are associated with increased Helicobacter pylori (H. pylori)-associated disease risk in an ethnically dependent manner. Due to the corroborated role of IL-1ß in H. pylori infection progression, our aim is to depict the impact of IL1B rs1143627 and rs16944 as well as the IL1RN variable number of identical tandem repeats (VNTR) on the clinical and biological features of Moroccan H. pylori-infected patients. A total of 58 patients with epigastralgic pain were referred to the gastroenterology department for histopathological and clinical analysis. DNA extraction from antrum and fundus biopsies and PCR-RFLP were performed to detect polymorphisms. As a result, VNTR was significantly associated with IL-1ß antrum levels (p-value = 0.029), where the *1/*4 genotype showed a positive association with upregulated cytokine levels in the antrum and was clustered with H. pylori-infected patients' features and higher levels of IL-1ß in the antrum and fundus. Likewise, *1/*1 genotype carriers clustered with severe gastritis activity and H. pylori density scores along with low levels of IL-1ß in the antrum and fundus, while the *1/*2 genotype was clustered with non-infected-patient features and normal IL-1ß levels. In conclusion, VNTR might be an interesting predictor to identify patients at risk of developing H. pylori-associated pathologies.

4.
J Immunol ; 210(4): 459-474, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36602965

ABSTRACT

Leptospira interrogans are bacteria that can infect all vertebrates and are responsible for leptospirosis, a neglected zoonosis. Some hosts, such as humans, are susceptible to the disease, whereas mice are resistant and get chronically colonized. Although leptospires escape recognition by some immune receptors, they activate the NOD-like receptor pyrin 3-inflammasome and trigger IL-1ß secretion. Classically, IL-1ß secretion is associated with lytic inflammatory cell death called pyroptosis, resulting from cytosolic LPS binding to inflammatory caspases, such as caspase 11. Interestingly, we showed that L. interrogans and Leptospira biflexa do not trigger cell death in either murine, human, hamster, or bovine macrophages, escaping both pyroptosis and apoptosis. We showed, in murine cells, that the mild IL-1ß secretion induced by leptospires occurred through nonlytic caspase 8-dependent gasdermin D pore formation and not through activation of caspase 11/noncanonical inflammasome. Strikingly, we demonstrated a potent antagonistic effect of pathogenic L. interrogans and their atypical LPS on spontaneous and Escherichia coli LPS-induced cell death. Indeed, LPS of L. interrogans efficiently prevents caspase 11 dimerization and subsequent massive gasdermin D cleavage. Finally, we showed that pyroptosis escape by leptospires prevents massive IL-1ß release, and we consistently found no major role of IL-1R in controlling experimental leptospirosis in vivo. Overall, to our knowledge, our findings described a novel mechanism by which leptospires dampen inflammation, thus potentially contributing to their stealthiness.


Subject(s)
Leptospira interrogans , Leptospirosis , Animals , Cattle , Cricetinae , Humans , Mice , Caspases/metabolism , Gasdermins , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Leptospira interrogans/metabolism , Leptospirosis/metabolism , Leptospirosis/microbiology , Lipopolysaccharides , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , Cell Death
5.
Future Microbiol ; 17: 633-641, 2022 05.
Article in English | MEDLINE | ID: mdl-35322705

ABSTRACT

Helicobacter pylori (H. pylori) infection is a causal factor of gastric cancer. Among the cytokines secreted during this infection, IL-1ß is highly associated with promotion and progression of gastric cancer. On the therapeutic front, eradication of H. pylori was thought to be efficient to restore gastric homeostasis. However, successful H. pylori eradication in patients with advanced stages (intestinal metaplasia) failed to diminish inflammation that is due to heightened Th17 response and elevated IL-1ß levels. In fact, association between these two components was established, suggesting that IL-1ß is a critical target in these cases. In this review, we will discuss the functional relevance of IL-1ß in advanced H. pylori infection and how its targeting may bring clinical benefit.


Helicobacter pylori (H. pylori) infection is a causal factor of gastric cancer. This disease is strongly associated to an inflammatory factor (interleukin 1ß). On the therapeutic front, eradication of H. pylori was thought to be efficient to restore gastric comfort. However, successful H. pylori eradication in patients with advanced stages of this infection failed to diminish inflammation, due to the inflammatory factor cited preciously, thereby exacerbating gastric tissue damage. In this review, we will discuss the functional relevance of IL-1ß in advanced H. pylori infection and how its targeting may bring clinical benefit.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Cytokines , Gastric Mucosa , Helicobacter Infections/complications , Helicobacter Infections/drug therapy , Humans , Interleukin-1beta , Stomach Neoplasms/drug therapy , Stomach Neoplasms/microbiology
6.
Parasite Epidemiol Control ; 9: e00129, 2020 May.
Article in English | MEDLINE | ID: mdl-32322694

ABSTRACT

Leishmaniases are a group of infectious diseases caused by protozoan Leishmania parasites and are transmitted by the bites of infected phlebotomine sandflies. The heterogeneity of these diseases is influenced by both parasitic properties and host factors. Cutaneous leishmaniasis (CL) is a major public health problem in Morocco, where the geographical expansion of CL (particularly CL caused by Leishmania tropica), the heterogeneous appearance of lesions and the difficulty in diagnosing CL contribute to late diagnosis of CL and delayed treatment of patients. Therefore, the main objective of this study was to describe the epidemiological and clinical profiles of patients with CL diagnosed in Casablanca (Morocco), which is a non-endemic area for CL. A cross-sectional study was conducted between 2010 and 2016, during which epidemiological and clinical data were collected from patients that met the inclusion criteria through an information sheet. Then, samples were obtained from each patient for parasitological and molecular diagnosis, and only patients with positive polymerase chain reaction and genotyping results were included in the study. Overall, 106 cases of CL were genotyped, of which 61 (57.5%) were caused by L. tropica, 38 (35.9%) by L. major and 7 (6.6%) by L. infantum. While all age groups were affected, CL cases wherein L. tropica was the causative agent were most frequently diagnosed in children aged 0-9 years (p = 0.005), whereas those caused by L. major were more frequently diagnosed in elderly patients (p = 0.004). Multivariate logistic regression analysis showed that two clinical variables were significantly associated with CL caused by L. tropica: lesion size (p = 0.002) and occurrence of lesion on the face (p = 0.005). Furthermore, the results of our survey highlighted the association of Leishmania infection when travelling to endemic areas. The high number of endemic foci where patients with CL were infected with L. tropica illustrated the tendency of this form to spread and generate epidemics, exposing young people to a greater degree to the disease. The epidemic status of CL caused by L. tropica in Morocco and the increased movement of the population from rural to urban areas indicate a possible introduction of this species to urban areas.

7.
Cytokine ; 126: 154893, 2020 02.
Article in English | MEDLINE | ID: mdl-31877554

ABSTRACT

INTRODUCTION: Helicobacter pylori induces acute gastritis that can progress to serious diseases such as gastric cancer. H. pylori interacts with host cells within the gastric mucosa, resulting in activation of multiple innate immune signalling pathways, leading to pro-inflammatory cytokines production and immune cells recruitment. Various studies have shown that there are ethnic- and population-related differences in the expression of these cytokines. Although the H. pylori infection is a major public health problem in Morocco, to our knowledge, no study has been carried out in gastric cytokine expression from H. pylori-infected Moroccan patients. Thus we aimed to (i) determine the IL-1ß, IL-8 and IL-17A gene expression in gastric biopsies from Moroccan patients infected with H. pylori, and (ii) to determine the cytokine signature of each pathological stages associated with this infection. MATERIAL AND METHODS: 71 patients with epigastralgic pain were included in this study. The H. pylori detection on gastric biopsies was performed by histopathological and PCR analysis. The IL-1ß, IL-8 and IL-17A mRNA expression in the antrun and fundus biopsies was performed by RT-qPCR. RESULTS: The histopathological and PCR analyses revealed that 87.32% of the patients were infected with H. pylori. IL-1ß mRNA expression was significantly lower in the antral mucosa of H. pylori-infected patients (p = 0.0038) than in the uninfected while there was no significant difference in the expression of IL-8 and IL-17A mRNA. The expression of the three cytokines was higher in the fundic mucosa of H. pylori-infected patients than in the uninfected patients, but only IL-8 and IL-17A expression reached statistical significance (p = 0.042 and p = 0.0179 respectively). Furthermore, the multivariate predictive analysis highlighted a cytokine signature that may predict metaplasia during the infection progression that involves a specific down-regulation of IL17A and an up-regulation of IL1ß in antral and fundic metaplasia respectively.


Subject(s)
Gastritis/pathology , Helicobacter Infections/pathology , Helicobacter pylori/immunology , Interleukin-17/analysis , Interleukin-1beta/analysis , Interleukin-8/analysis , Adult , Female , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis/diagnosis , Gastritis/microbiology , Helicobacter Infections/diagnosis , Humans , Male , Middle Aged , Morocco , Signal Transduction/immunology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
8.
Science ; 365(6448)2019 07 05.
Article in English | MEDLINE | ID: mdl-31273097

ABSTRACT

Multiple cytosolic innate sensors form large signalosomes after activation, but this assembly needs to be tightly regulated to avoid accumulation of misfolded aggregates. We found that the eIF2α kinase heme-regulated inhibitor (HRI) controls NOD1 signalosome folding and activation through a process requiring eukaryotic initiation factor 2α (eIF2α), the transcription factor ATF4, and the heat shock protein HSPB8. The HRI/eIF2α signaling axis was also essential for signaling downstream of the innate immune mediators NOD2, MAVS, and TRIF but dispensable for pathways dependent on MyD88 or STING. Moreover, filament-forming α-synuclein activated HRI-dependent responses, which suggests that the HRI pathway may restrict toxic oligomer formation. We propose that HRI, eIF2α, and HSPB8 define a novel cytosolic unfolded protein response (cUPR) essential for optimal innate immune signaling by large molecular platforms, functionally homologous to the PERK/eIF2α/HSPA5 axis of the endoplasmic reticulum UPR.


Subject(s)
Cytosol/enzymology , Cytosol/immunology , Immunity, Innate , Protein Serine-Threonine Kinases/physiology , Unfolded Protein Response/immunology , Activating Transcription Factor 4/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , Fibroblasts , Heat-Shock Proteins/metabolism , Humans , Listeria/immunology , Membrane Proteins/metabolism , Mice , Mice, Mutant Strains , Molecular Chaperones/metabolism , Myeloid Differentiation Factor 88/metabolism , Nod1 Signaling Adaptor Protein/chemistry , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Protein Serine-Threonine Kinases/genetics , Salmonella/immunology , Salmonella Infections , Shigella/immunology , Signal Transduction
9.
Cell Death Dis ; 10(5): 346, 2019 04 25.
Article in English | MEDLINE | ID: mdl-31024004

ABSTRACT

Early detection of viruses by the innate immune system is crucial for host defense. The NLRP3 inflammasome, through activation of caspase-1, promotes the maturation of IL-1ß and IL-18, which are critical for antiviral immunity and inflammatory response. However, the mechanism by which viruses activate this inflammasome is still debated. Here, we report that the replication of cytopathogenic RNA viruses such as vesicular stomatitis virus (VSV) or encephalomyocarditis virus (EMCV) induced a lytic cell death leading to potassium efflux, the common trigger of NLRP3 inflammasome activation. This lytic cell death was not prevented by a chemical or genetic inhibition of apoptosis, pyroptosis, or necroptosis but required the viral replication. Hence, the viruses that stimulated type I IFNs production after their sensing did not activate NLRP3 inflammasome due to an inhibition of their replication. In contrast, NLRP3 inflammasome activation induced by RNA virus infection was stimulated in IFNAR-deficient or MAVS-deficient cells consequently to an increased viral replication and ensuing lytic cell death. Therefore, in a context of inefficient IFN response, viral replication-induced lytic cell death activates of the NLRP3 inflammasome to fight against infection.


Subject(s)
Encephalomyocarditis virus/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Potassium/metabolism , Vesiculovirus/physiology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Cells/cytology , Dynamins/antagonists & inhibitors , Dynamins/genetics , Dynamins/metabolism , Humans , Inflammasomes/metabolism , Interleukin-1beta/analysis , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/metabolism , Macrophages/virology , Mice , Necroptosis , RNA Interference , RNA, Small Interfering/metabolism , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Virus Replication
10.
Front Immunol ; 9: 853, 2018.
Article in English | MEDLINE | ID: mdl-29755463

ABSTRACT

After a viral infection and the stimulation of some pattern-recognition receptors as the toll-like receptor 3 in the endosomes or the RIG-I-like receptors in the cytosol, activation of the IKK-related kinase TBK1 leads to the production of type I interferons (IFNs) after phosphorylation of the transcription factors IRF3 and IRF7. Recent findings indicate an involvement of K63-linked polyubiquitination and of the Golgi-localized protein optineurin (OPTN) in the activation of this crucial kinase involved in innate antiviral immunity. This review summarizes the sensing of viruses and the signaling leading to type I IFN production following TBK1 activation through its ubiquitination and the sensing of ubiquitin chains by OPTN at the Golgi apparatus.


Subject(s)
Immunity, Innate , Interferon Type I/metabolism , Signal Transduction , Transcription Factor TFIIIA/immunology , Ubiquitination , Virus Diseases/immunology , Animals , Cell Cycle Proteins , Golgi Apparatus/immunology , Humans , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factor-7/metabolism , Interferon Type I/immunology , Membrane Transport Proteins , Mice , Phosphorylation , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Transcription Factor TFIIIA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...