Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Lancet ; 401(10379): 843-855, 2023 03 11.
Article in English | MEDLINE | ID: mdl-36906345

ABSTRACT

BACKGROUND: Bordetella pertussis epidemics persist as transmission remains unabated despite high acellular pertussis vaccination rates. BPZE1, a live attenuated intranasal pertussis vaccine, was designed to prevent B pertussis infection and disease. We aimed to assess the immunogenicity and safety of BPZE1 compared with the tetanus-diphtheria-acellular pertussis vaccine (Tdap). METHODS: In this double-blind, phase 2b trial at three research centres in the USA, healthy adults aged 18-50 years were randomly assigned (2:2:1:1) via a permuted block randomisation schedule to receive BPZE1 vaccination followed by BPZE1 attenuated challenge, BPZE1 vaccination followed by placebo challenge, Tdap followed by BPZE1 attenuated challenge, or Tdap followed by placebo challenge. On day 1, lyophilised BPZE1 was reconstituted with sterile water and given intranasally (0·4 mL delivered to each nostril), whereas Tdap was given intramuscularly. To maintain masking, participants in the BPZE1 groups received an intramuscular saline injection, and those in the Tdap groups received intranasal lyophilised placebo buffer. The attenuated challenge took place on day 85. The primary immunogenicity endpoint was the proportion of participants achieving nasal secretory IgA seroconversion against at least one B pertussis antigen on day 29 or day 113. Reactogenicity was assessed up to 7 days after vaccination and challenge, and adverse events were recorded for 28 days after vaccination and challenge. Serious adverse events were monitored throughout the study. This trial is registered with ClinicalTrials.gov, NCT03942406. FINDINGS: Between June 17 and Oct 3, 2019, 458 participants were screened and 280 were randomly assigned to the main cohort: 92 to the BPZE1-BPZE1 group, 92 to the BPZE1-placebo group, 46 to the Tdap-BPZE1 group, and 50 to the Tdap-placebo group. Seroconversion of at least one B pertussis-specific nasal secretory IgA was recorded in 79 (94% [95% CI 87-98]) of 84 participants in the BPZE1-BPZE1 group, 89 (95% [88-98]) of 94 in the BPZE1-placebo group, 38 (90% [77-97]) of 42 in the Tdap-BPZE1 group, and 42 (93% [82-99]) of 45 in the Tdap-placebo group. BPZE1 induced broad and consistent B pertussis-specific mucosal secretory IgA responses, whereas Tdap did not induce consistent mucosal secretory IgA responses. Both vaccines were well tolerated, with mild reactogenicity and no serious adverse events related to study vaccination. INTERPRETATION: BPZE1 induced nasal mucosal immunity and produced functional serum responses. BPZE1 has the potential to avert B pertussis infections, which ultimately could lead to reduced transmission and diminished epidemic cycles. These results should be confirmed in large phase 3 trials. FUNDING: ILiAD Biotechnologies.


Subject(s)
Diphtheria-Tetanus-acellular Pertussis Vaccines , Diphtheria , Tetanus , Whooping Cough , Adult , Humans , Diphtheria/prevention & control , Diphtheria-Tetanus-acellular Pertussis Vaccines/immunology , Double-Blind Method , Immunoglobulin A, Secretory , Tetanus/prevention & control , Vaccines, Attenuated/immunology , Whooping Cough/prevention & control , Young Adult , Middle Aged , Adolescent
2.
Nucleic Acids Res ; 49(11): 6082-6099, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34057477

ABSTRACT

Oligonucleotide-based therapeutics have the capacity to engage with nucleic acid immune sensors to activate or block their response, but a detailed understanding of these immunomodulatory effects is currently lacking. We recently showed that 2'-O-methyl (2'OMe) gapmer antisense oligonucleotides (ASOs) exhibited sequence-dependent inhibition of sensing by the RNA sensor Toll-Like Receptor (TLR) 7. Here we discovered that 2'OMe ASOs can also display sequence-dependent inhibitory effects on two major sensors of DNA, namely cyclic GMP-AMP synthase (cGAS) and TLR9. Through a screen of 80 2'OMe ASOs and sequence mutants, we characterized key features within the 20-mer ASOs regulating cGAS and TLR9 inhibition, and identified a highly potent cGAS inhibitor. Importantly, we show that the features of ASOs inhibiting TLR9 differ from those inhibiting cGAS, with only a few sequences inhibiting both pathways. Together with our previous studies, our work reveals a complex pattern of immunomodulation where 95% of the ASOs tested inhibited at least one of TLR7, TLR9 or cGAS by ≥30%, which may confound interpretation of their in vivo functions. Our studies constitute the broadest analysis of the immunomodulatory effect of 2'OMe ASOs on nucleic acid sensing to date and will support refinement of their therapeutic development.


Subject(s)
Nucleotidyltransferases/antagonists & inhibitors , Oligonucleotides, Antisense/chemistry , Toll-Like Receptor 9/antagonists & inhibitors , Adult , Animals , Base Sequence , Cells, Cultured , DNA , Humans , Mice , Signal Transduction , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 7/antagonists & inhibitors
3.
Gastroenterology ; 159(1): 169-182.e8, 2020 07.
Article in English | MEDLINE | ID: mdl-32169428

ABSTRACT

BACKGROUND & AIMS: Helicobacter pylori induces strong inflammatory responses that are directed at clearing the infection, but if not controlled, these responses can be harmful to the host. We investigated the immune-regulatory effects of the innate immune molecule, nucleotide-binding oligomerization domain-like receptors (NLR) family CARD domain-containing 5 (NLRC5), in patients and mice with Helicobacter infection. METHODS: We obtained gastric biopsies from 30 patients in Australia. We performed studies with mice that lack NLRC5 in the myeloid linage (Nlrc5møKO) and mice without Nlrc5 gene disruption (controls). Some mice were gavaged with H pylori SS1 or Helicobacter felis; 3 months later, stomachs, spleens, and sera were collected, along with macrophages derived from bone marrow. Human and mouse gastric tissues and mouse macrophages were analyzed by histology, immunohistochemistry, immunoblots, and quantitative polymerase chain reaction. THP-1 cells (human macrophages, controls) and NLRC5-/- THP-1 cells (generated by CRISPR-Cas9 gene editing) were incubated with Helicobacter and gene expression and production of cytokines were analyzed. RESULTS: Levels of NLRC5 messenger RNA were significantly increased in gastric tissues from patients with H pylori infection, compared with patients without infection (P < .01), and correlated with gastritis severity (P < .05). H pylori bacteria induced significantly higher levels of chemokine and cytokine production by NLRC5-/- THP-1 macrophages than by control THP-1 cells (P < .05). After 3 months of infection with H felis, Nlrc5mø-KO mice developed gastric hyperplasia (P < .0001), splenomegaly (P < .0001), and increased serum antibody titers (P < .01), whereas control mice did not. Nlrc5mø-KO mice with chronic H felis infection had increased numbers of gastric B-cell follicles expressing CD19 (P < .0001); these follicles had features of mucosa-associated lymphoid tissue lymphoma. We identified B-cell-activating factor as a protein that promoted B-cell hyperproliferation in Nlrc5mø-KO mice. CONCLUSIONS: NLRC5 is a negative regulator of gastric inflammation and mucosal lymphoid formation in response to Helicobacter infection. Aberrant NLRC5 signaling in macrophages can promote B-cell lymphomagenesis during chronic Helicobacter infection.


Subject(s)
Helicobacter Infections/complications , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoma, B-Cell, Marginal Zone/immunology , Stomach Neoplasms/immunology , Animals , B-Lymphocytes/immunology , Biopsy , Cell Proliferation , Disease Models, Animal , Female , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gene Expression Regulation, Neoplastic/immunology , Gene Knockout Techniques , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter felis/immunology , Helicobacter pylori/immunology , Humans , Hyperplasia/immunology , Hyperplasia/microbiology , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/microbiology , Lymphoid Tissue/pathology , Lymphoma, B-Cell, Marginal Zone/microbiology , Lymphoma, B-Cell, Marginal Zone/pathology , Male , Mice , Mice, Knockout , Signal Transduction/immunology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , THP-1 Cells
4.
Nucleic Acids Res ; 45(1): 198-205, 2017 Jan 09.
Article in English | MEDLINE | ID: mdl-27694309

ABSTRACT

Acridine dyes, including proflavine and acriflavine, were commonly used as antiseptics before the advent of penicillins in the mid-1940s. While their mode of action on pathogens was originally attributed to their DNA intercalating activity, work in the early 1970s suggested involvement of the host immune responses, characterized by induction of interferon (IFN)-like activities through an unknown mechanism. We demonstrate here that sub-toxic concentrations of a mixture of acriflavine and proflavine instigate a cyclic-GMP-AMP (cGAMP) synthase (cGAS)-dependent type-I IFN antiviral response. This pertains to the capacity of these compounds to induce low level DNA damage and cytoplasmic DNA leakage, resulting in cGAS-dependent cGAMP-like activity. Critically, acriflavine:proflavine pre-treatment of human primary bronchial epithelial cells significantly reduced rhinovirus infection. Collectively, our findings constitute the first evidence that non-toxic DNA binding agents have the capacity to act as indirect agonists of cGAS, to exert potent antiviral effects in mammalian cells.


Subject(s)
Acriflavine/pharmacology , Antiviral Agents/pharmacology , Immunologic Factors/pharmacology , Intercalating Agents/pharmacology , Membrane Proteins/genetics , Nucleotidyltransferases/genetics , Proflavine/pharmacology , Animals , Bronchi/drug effects , Bronchi/immunology , Bronchi/virology , Cell Line, Transformed , Chlorocebus aethiops , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/virology , Gene Expression Regulation , HEK293 Cells , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Membrane Proteins/agonists , Membrane Proteins/immunology , Mice , Nucleotides, Cyclic/immunology , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/immunology , Primary Cell Culture , Rhinovirus/drug effects , Rhinovirus/growth & development , Signal Transduction , Vero Cells , Viral Load/drug effects
5.
Nucleic Acids Res ; 44(11): 5356-64, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27166376

ABSTRACT

Gene-recombinase technologies, such as Cre/loxP-mediated DNA recombination, are important tools in the study of gene function, but have potential side effects due to damaging activity on DNA. Here we show that DNA recombination by Cre instigates a robust antiviral response in mammalian cells, independent of legitimate loxP recombination. This is due to the recruitment of the cytosolic DNA sensor STING, concurrent with Cre-dependent DNA damage and the accumulation of cytoplasmic DNA. Importantly, we establish a direct interplay between this antiviral response and cell-cell interactions, indicating that low cell densities in vitro could be useful to help mitigate these effects of Cre. Taking into account the wide range of interferon stimulated genes that may be induced by the STING pathway, these results have broad implications in fields such as immunology, cancer biology, metabolism and stem cell research. Further, this study sets a precedent in the field of gene-engineering, possibly applicable to other enzymatic-based genome editing technologies.


Subject(s)
Homologous Recombination , Immunity, Innate , Integrases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Animals , Cell Line , Epithelial Cells/metabolism , Fibroblasts/metabolism , Humans , Integrases/genetics , Macrophages/metabolism , Mice
6.
Nucleic Acids Res ; 43(2): 1177-88, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25539920

ABSTRACT

Anti-microRNA (miRNA) oligonucleotides (AMOs) with 2'-O-Methyl (2'OMe) residues are commonly used to study miRNA function and can achieve high potency, with low cytotoxicity. Not withstanding this, we demonstrate the sequence-dependent capacity of 2'OMe AMOs to inhibit Toll-like receptor (TLR) 7 and 8 sensing of immunostimulatory RNA, independent of their miRNA-targeting function. Through a screen of 29 AMOs targeting common miRNAs, we found a subset of sequences highly inhibitory to TLR7 sensing in mouse macrophages. Interspecies conservation of this inhibitory activity was confirmed on TLR7/8 activity in human peripheral blood mononuclear cells. Significantly, we identified a core motif governing the inhibitory activity of these AMOs, which is present in more than 50 AMOs targeted to human miRNAs in miRBaseV20. DNA/locked nucleic acids (LNA) AMOs synthesized with a phosphorothioate backbone also inhibited TLR7 sensing in a sequence-dependent manner, demonstrating that the off-target effects of AMOs are not restricted to 2'OMe modification. Taken together, our work establishes the potential for off-target effects of AMOs on TLR7/8 function, which should be taken into account in their therapeutic development and in vivo application.


Subject(s)
MicroRNAs/antagonists & inhibitors , Oligonucleotides/chemistry , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 8/antagonists & inhibitors , Adjuvants, Immunologic/pharmacology , Animals , Base Sequence , HEK293 Cells , Humans , Mice , Nucleotide Motifs , RNA/pharmacology
7.
J Immunol ; 190(7): 3706-15, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23460743

ABSTRACT

Virulent Helicobacter pylori strains that specifically activate signaling in epithelial cells via the innate immune molecule, nucleotide oligomerization domain 1 (NOD1), are more frequently associated with IFN-γ-dependent inflammation and with severe clinical outcomes (i.e., gastric cancer and peptic ulceration). In cell culture models, we showed that H. pylori activation of the NOD1 pathway caused enhanced proinflammatory signaling in epithelial cells in response to IFN-γ stimulation through the direct effects of H. pylori on two components of the IFN-γ signaling pathway, STAT1 and IFN regulatory factor 1 (IRF1). Specifically, H. pylori activation of the NOD1 pathway was shown to increase the levels of STAT1-Tyr(701)/Ser(727) phosphorylation and IRF1 expression/synthesis in cells, resulting in enhanced production of the NOD1- and IFN-γ-regulated chemokines, IL-8- and IFN-γ-induced protein 10, respectively. Consistent with the notion that heightened proinflammatory signaling in epithelial cells may have an impact on disease severity, we observed significantly increased expression levels of NOD1, CXCL8, IRF1, and CXCL10 in human gastric biopsies displaying severe gastritis, when compared with those without gastritis (p < 0.05, p < 0.001, p < 0.01, and p < 0.05, respectively). Interestingly, NOD1, CXCL8, and IRF1 expression levels were also significantly upregulated in gastric tumor tissues, when compared with paired nontumor samples (p < 0.0001, p < 0.05, and p < 0.05, respectively). Thus, we propose that cross-talk between NOD1 and IFN-γ signaling pathways contribute to H. pylori-induced inflammatory responses, potentially revealing a novel mechanism whereby virulent H. pylori strains promote more severe disease.


Subject(s)
Epithelial Cells/metabolism , Gastric Mucosa/metabolism , Helicobacter Infections/metabolism , Interferon-gamma/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Signal Transduction , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cell Line , Chemokines/biosynthesis , Disease Progression , Epithelial Cells/microbiology , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Interferon Regulatory Factor-1/genetics , Phosphorylation , STAT1 Transcription Factor/metabolism , Transcription, Genetic
8.
Gastroenterology ; 142(2): 281-91, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22062361

ABSTRACT

BACKGROUND & AIMS: Studies in animal models have shown that bone marrow-derived cells (BMDC) could be involved in the formation of carcinomas of the upper gastrointestinal tract, including gastric carcinoma. Most gastric carcinomas in humans have been associated with chronic infection with Helicobacter pylori; we investigated the bacteria's potential to induce premalignant lesions in mice and studied the kinetics of BMDC settlement in the gastric epithelium. METHODS: C57BL/6J female chimeric mice with BMDCs from male donors that express green fluorescent protein were infected with human-derived and mouse-adapted strains of H pylori and followed. We assessed development of pathologic features and recruitment of BMDC to the gastric mucosa using immunohistochemistry and fluorescent in situ hybridization analyses of gastric tissue sections. RESULTS: Infection of mice with different strains of H pylori led to the development of chronic inflammation, hyperplasia, and mucinous metaplasia, and, later in life, of pseudointestinal metaplasia and dysplasia. After 1 year, gastric glands that contained green fluorescent protein-positive male cells were detected in 50%-90% of female chimeric mice infected with H pylori strains; the presence of these glands correlated with the development of pseudointestinal metaplasia. Twenty-two percent of H pylori-induced dysplastic lesions were composed of glands that contained epithelial BMDCs. CONCLUSIONS: H pylori infection leads to development of chronic inflammation, hyperplasia, metaplasia, and dysplasia, as well as the recruitment and accumulation of BMDC in the gastric epithelial mucosa. Nearly 25% of dysplastic lesions include cells that originate from the BM.


Subject(s)
Bone Marrow Cells/pathology , Gastric Mucosa/pathology , Helicobacter Infections/pathology , Helicobacter pylori , Neoplastic Stem Cells/pathology , Precancerous Conditions/microbiology , Stomach Neoplasms/microbiology , Animals , Disease Models, Animal , Female , Gastric Mucosa/microbiology , Helicobacter Infections/complications , Hyperplasia/microbiology , In Situ Hybridization, Fluorescence , Inflammation/microbiology , Inflammation/pathology , Male , Metaplasia/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precancerous Conditions/pathology , Stomach Neoplasms/pathology
9.
Helicobacter ; 13(3): 167-73, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18466391

ABSTRACT

BACKGROUND: Helicobacter pylori infection leads to different chronic diseases, suggesting that this bacterium can evade the host immune defense system. The ability to control lymphocyte proliferation may be a mechanism leading to the development of gastric pathologies. Our aim was to characterize the effects of mucosa-associated lymphoid tissue (MALT) associated H. pylori strains on lymphocyte proliferation. MATERIALS AND METHODS: We measured the in vitro proliferation of human lymphocytes originally from blood or tonsil samples in the presence or absence of viable bacteria or lysates. RESULTS: We showed that MALT lymphoma-associated strains are not likely to be directly responsible for anarchical B-cell proliferation in vitro. On the other hand, proliferation of prestimulated T lymphocytes was abolished in vitro by the presence of all H. pylori strains, whether associated with MALT lymphoma or not. CONCLUSION: Inhibition of T-cell proliferation may be of major importance in the gastric colonization and in the persistence of the infection. Furthermore, this inhibition may favor anarchical B-cell proliferation in vivo and predispose the host to gastric MALT lymphoma, whereas MALT-associated H. pylori strains do not appear to possess a specific capability to directly stimulate B-lymphocyte proliferation.


Subject(s)
Gastric Mucosa/microbiology , Helicobacter Infections/immunology , Helicobacter pylori/physiology , Lymphocyte Activation , Lymphoma, B-Cell, Marginal Zone/microbiology , Cell Proliferation , Helicobacter Infections/microbiology , Helicobacter pylori/classification , Humans , Lymphocytes/cytology , Lymphocytes/immunology , Lymphoma, B-Cell, Marginal Zone/immunology , Lymphoma, B-Cell, Marginal Zone/pathology
10.
Article in English | MEDLINE | ID: mdl-29616197

ABSTRACT

Murine models of Salmonella enterica serovar Typhimurium infection are one of the commonest tools to study host-pathogen interactions during bacterial infections. Critically, the outcome of S. Typhimurium infection is impacted by the genetic background of the mouse strain used, with macrophages from C57BL/6 and BALB/c mice lacking the capacity to control intracellular bacterial replication. For this reason, the use of congenic strains, which mix the genetic backgrounds of naturally protected mouse strains with those of susceptible strains, has the capacity to significantly alter results and interpretation of S. Typhimurium infection studies. Here, we describe how macrophage knockout cell lines generated by CRISPR/Cas9 gene editing can help determine the contribution of background contaminations in the phenotypes of primary macrophages from congenic mice, on the outcome of S. Typhimurium infection studies. Our own experience illustrates how the CRISPR/Cas9 technology can be used to complement pre-existing knockout models, and shows that there is great merit in performing concurrent studies with both genetic models, to exclude unanticipated side-effects on host-pathogen interactions.


Subject(s)
Gene Editing , Salmonella Infections/genetics , Salmonella typhimurium/physiology , Animals , Animals, Congenic , CRISPR-Cas Systems , Female , Host-Pathogen Interactions , Humans , Macrophages/immunology , Macrophages/microbiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology
11.
Methods Mol Biol ; 1517: 127-135, 2017.
Article in English | MEDLINE | ID: mdl-27924479

ABSTRACT

MicroRNAs (miRNAs) are involved in most cellular processes and are deregulated in several diseases. Antisense miRNA oligonucleotides (AMOs) therefore present novel therapeutic opportunities. Currently, in vivo delivery of AMOs often relies on high doses of nucleic acids, with nonspecific uptake by most tissues. Critically, AMOs accumulate in phagocytic cells where they can interfere with immune functions, such as the activation of Toll-Like Receptors (TLRs). In this chapter, we describe a method to assess the possible off-target effects of AMOs on TLR7, 8, and 9 sensing.


Subject(s)
Immunity, Innate/genetics , MicroRNAs/genetics , Molecular Biology/methods , Oligonucleotides, Antisense/genetics , Humans , MicroRNAs/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Phagocytes/immunology , Phagocytes/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 8/genetics , Toll-Like Receptor 9/genetics
12.
mBio ; 8(5)2017 10 03.
Article in English | MEDLINE | ID: mdl-28974621

ABSTRACT

Inflammatory responses, while essential for pathogen clearance, can also be deleterious to the host. Chemical inhibition of topoisomerase 1 (Top1) by low-dose camptothecin (CPT) can suppress transcriptional induction of antiviral and inflammatory genes and protect animals from excessive and damaging inflammatory responses. We describe the unexpected finding that minor DNA damage from topoisomerase 1 inhibition with low-dose CPT can trigger a strong antiviral immune response through cyclic GMP-AMP synthase (cGAS) detection of cytoplasmic DNA. This argues against CPT having only anti-inflammatory activity. Furthermore, expression of the simian virus 40 (SV40) large T antigen was paramount to the proinflammatory antiviral activity of CPT, as it potentiated cytoplasmic DNA leakage and subsequent cGAS recruitment in human and mouse cell lines. This work suggests that the capacity of Top1 inhibitors to blunt inflammatory responses can be counteracted by viral oncogenes and that this should be taken into account for their therapeutic development.IMPORTANCE Recent studies suggest that low-dose DNA-damaging compounds traditionally used in cancer therapy can have opposite effects on antiviral responses, either suppressing (with the example of CPT) or potentiating (with the example of doxorubicin) them. Our work demonstrates that the minor DNA damage promoted by low-dose CPT can also trigger strong antiviral responses, dependent on the presence of viral oncogenes. Taken together, these results call for caution in the therapeutic use of low-dose chemotherapy agents to modulate antiviral responses in humans.


Subject(s)
DNA Topoisomerases, Type I/drug effects , Immunity, Innate/drug effects , Nucleotides, Cyclic/metabolism , Simian virus 40/drug effects , Topoisomerase I Inhibitors/pharmacology , Animals , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/immunology , Antiviral Agents/pharmacology , Camptothecin/pharmacology , Cell Line , Coculture Techniques , DNA Damage , DNA Topoisomerases, Type I/metabolism , Fibroblasts/drug effects , Fibroblasts/virology , Humans , Inflammation , Mice , Simian virus 40/immunology , Simian virus 40/physiology , Virus Diseases/drug therapy , Virus Diseases/immunology , Virus Diseases/virology
13.
Methods Mol Biol ; 1390: 79-90, 2016.
Article in English | MEDLINE | ID: mdl-26803623

ABSTRACT

Aberrant sensing of self-nucleic acids by Toll-like receptor (TLR) 7, 8, or 9 is associated with several autoimmune disorders, including systemic lupus erythematosus (SLE), rheumatoid arthritis, psoriasis, or systemic sclerosis. In recent years, several classes of synthetic oligonucleotides have been shown to antagonize sensing of immunostimulatory nucleic acids by TLR7/8/9, indicating that these molecules could have therapeutic applications in such autoimmune diseases. Conversely, synthetic oligonucleotides used in therapeutic technologies such as antisense and microRNA inhibitors also have the potential to inhibit TLR7/8/9 sensing, rendering patients more susceptible to viral/bacterial infections. This chapter describes a protocol to define the inhibitory activity of synthetic oligonucleotides on TLR7.


Subject(s)
Oligonucleotides/metabolism , Oligonucleotides/pharmacology , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 7/metabolism , Cell Culture Techniques , Cell Line , Enzyme-Linked Immunosorbent Assay/methods , Humans , Immunomodulation/drug effects , Ligands , Oligonucleotides/genetics , Protein Binding , RNA/genetics , RNA/immunology , Transfection , Tumor Necrosis Factor-alpha/metabolism
14.
Front Immunol ; 4: 344, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24155747

ABSTRACT

Innate immune recognition of bacteria is the first requirement for mounting an effective immune response able to control infection. Over the previous decade, the general paradigm was that extracellular bacteria were only sensed by cell surface-expressed Toll-like receptors (TLRs), whereas cytoplasmic sensors, including members of the Nod-like receptor (NLR) family, were specific to pathogens capable of breaching the host cell membrane. It has become apparent, however, that intracellular innate immune molecules, such as the NLRs, play key roles in the sensing of not only intracellular, but also extracellular bacterial pathogens or their components. In this review, we will discuss the various mechanisms used by bacteria to activate NLR signaling in host cells. These mechanisms include bacterial secretion systems, pore-forming toxins, and outer membrane vesicles. We will then focus on the influence of NLR activation on the development of adaptive immune responses in different cell types.

15.
PLoS One ; 6(12): e29007, 2011.
Article in English | MEDLINE | ID: mdl-22216156

ABSTRACT

The role of bone marrow-derived mesenchymal stem cells (MSC) in the physiology of the gastrointestinal tract epithelium is currently not well established. These cells can be recruited in response to inflammation due to epithelial damage, home, and participate in tissue repair. In addition, in the case of tissue repair failure, these cells could transform and be at the origin of carcinomas. However, the chemoattractant molecules responsible for MSC recruitment and migration in response to epithelial damage, and particularly to Helicobacter pylori infection, remain unknown although the role of some chemokines has been suggested. This work aimed to get insight into the mechanisms of mouse MSC migration during in vitro infection of mouse gastrointestinal epithelial cells by H. pylori. Using a cell culture insert system, we showed that infection of gastrointestinal epithelial cells by different H. pylori strains is able to stimulate the migration of MSC. This mechanism involves the secretion by infected epithelial cells of multiple cytokines, with a major role of TNFα, mainly via a Nuclear Factor-kappa B-dependent pathway. This study provides the first evidence of the role of H. pylori infection in MSC migration and paves the way to a better understanding of the role of bone marrow-derived stem cells in gastric pathophysiology and carcinogenesis.


Subject(s)
Cell Movement , Epithelial Cells/microbiology , Gastrointestinal Tract/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Mesenchymal Stem Cells/pathology , NF-kappa B/metabolism , Animals , Helicobacter Infections/microbiology , Mice , Signal Transduction
16.
PLoS One ; 6(5): e19569, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21573181

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSC) have the ability to differentiate into a variety of cell types and are a potential source for epithelial tissue repair. Several studies have demonstrated their ability to repopulate the gastrointestinal tract (GIT) in bone marrow transplanted patients or in animal models of gastrointestinal carcinogenesis where they were the source of epithelial cancers. However, mechanism of MSC epithelial differentiation still remains unclear and controversial with trans-differentiation or fusion events being evoked. This study aimed to investigate the ability of MSC to acquire epithelial characteristics in the particular context of the gastrointestinal epithelium and to evaluate the role of cell fusion in this process. In vitro coculture experiments were performed with three gastrointestinal epithelial cell lines and MSC originating from two patients. After an 8 day coculture, MSC expressed epithelial markers. Use of a semi-permeable insert did not reproduce this effect, suggesting importance of cell contacts. Tagged cells coculture or FISH on gender-mismatched cells revealed clearly that epithelial differentiation resulted from cellular fusion events, while expression of mesenchymal markers on fused cells decreased over time. In vivo cell xenograft in immunodeficient mice confirmed fusion of MSC with gastrointestinal epithelial cells and self-renewal abilities of these fused cells. In conclusion, our results indicate that fusion could be the predominant mechanism by which human MSC may acquire epithelial characteristics when in close contact with epithelial cells from gastrointestinal origin . These results could contribute to a better understanding of the cellular and molecular mechanisms allowing MSC engraftment into the GIT epithelium.


Subject(s)
Bone Marrow Cells/cytology , Cell Fusion/methods , Epithelial Cells/cytology , Gastrointestinal Tract/cytology , Mesenchymal Stem Cells/cytology , Animals , Biomarkers/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Chromosomes, Human/metabolism , Coculture Techniques , Epithelial Cells/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , In Situ Hybridization, Fluorescence , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Neoplasms/pathology , Phenotype , Transplantation, Heterologous
17.
J Infect Dis ; 198(9): 1379-87, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18811585

ABSTRACT

BACKGROUND: homB codes for a putative Helicobacter pylori outer membrane protein and has previously been associated with peptic ulcer disease (PUD) in children. METHODS: A total of 190 H. pylori strains isolated from children and adults were studied to evaluate the clinical importance of the homB gene. In vitro experiments were performed to identify HomB mechanisms of bacterial pathogenicity. RESULTS: Characterization of the isolates demonstrated that homB was significantly associated with PUD in 86 children (odds ratio [OR], 7.64 [95% confidence interval {CI}, 2.65-22.05]) and in 32 adults < or =40 years of age (OR, 11.25 [95% CI, 1.86-68.13]). homB was correlated with the presence of cagA, babA2, vacAs1, hopQI, and oipA "on" genotype (P< .001) The HomB protein was found to be expressed in the H. pylori outer membrane and was noted to be antigenic in humans. H. pylori homB knockout mutant strains presented reduced ability to induce interleukin-8 secretion from human gastric epithelial cells, as well as reduced capacity to bind to these cells. Both of these functions correlated with the number of homB copies present in a strain. CONCLUSION: homB can be considered a comarker of H. pylori strains associated with PUD. Moreover, results strongly suggest that HomB is involved in the inflammatory response and in H. pylori adherence, constituting a novel putative virulence factor.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Genetic Markers , Helicobacter pylori/genetics , Peptic Ulcer/microbiology , Adult , Antigens, Bacterial , Bacterial Adhesion , Bacterial Outer Membrane Proteins/metabolism , Cell Line, Tumor , Child , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genetic Variation , Helicobacter pylori/pathogenicity , Humans , Interleukin-8/metabolism , Logistic Models , Male , Odds Ratio , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL