Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancers (Basel) ; 13(9)2021 Apr 25.
Article in English | MEDLINE | ID: mdl-33922955

ABSTRACT

Chronic inflammation plays an important role in the pathogenesis of oesophageal adenocarcinoma (EAC) and its only known precursor, Barrett's oesophagus (BE). Recent studies have shown that oesophageal TLR2 levels increase from normal epithelium towards EAC. TLR2 signalling is therefore likely to be important during EAC development and progression, which requires an inflammatory microenvironment. Here, we show that, in response to TLR2 stimulation, BE organoids and early-stage EAC cells secrete pro-inflammatory cytokines and chemokines which recruit macrophages to the tumour site. Factors secreted from TLR2-stimulated EAC cells are shown to subsequently activate TLR2 on naïve macrophages, priming them for inflammasome activation and inducing their differentiation to an M2/TAM-like phenotype. We identify the endogenous TLR2 ligand, HMGB1, as the factor secreted from EAC cells responsible for the observed TLR2-mediated effects on macrophages. Our results indicate that HMGB1 signalling between EAC cells and macrophages creates an inflammatory tumour microenvironment to facilitate EAC progression. In addition to identifying HMGB1 as a potential target for early-stage EAC treatment, our data suggest that blocking TLR2 signalling represents a mechanism to limit HMGB1 release, inflammatory cell infiltration and inflammation during EAC progression.

2.
Methods Mol Biol ; 2283: 175-190, 2021.
Article in English | MEDLINE | ID: mdl-33765318

ABSTRACT

Western blot and enzyme-linked immunosorbent assay (ELISA) are antibody-mediated techniques which are widely used for the detection and characterization of alterations in host protein expression following H. pylori infection . Both techniques are highly specific and sensitive for protein detection, with Western blot detection sensitivity as low as picogram amounts of the protein of interest, while the typical ELISA detection range is 0.01-0.1 ng. Here we provide an experimental example to demonstrate the application of these techniques for the determination of macrophage inflammatory responses following H. pylori infection .


Subject(s)
Cytokines/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/immunology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Helicobacter pylori/pathogenicity , Humans , Immunity, Innate
3.
Immunohorizons ; 4(10): 627-633, 2020 10 12.
Article in English | MEDLINE | ID: mdl-33046484

ABSTRACT

Peptic ulcers are caused by the interaction between bacterial and host factors. This study demonstrates enhanced expression of caspase-4 in peptic ulcer patient biopsies, indicating that pyroptosis and noncanonical inflammasome activity may be processes involved in peptic ulcer disease. We show that primary murine macrophages infected with Helicobacter pylori upregulate caspase-11 (the ortholog of human caspase-4), activate caspase-1, and secrete IL-1ß. We demonstrate that misoprostol (a stable PGE1 analogue) decreased IL-1ß secretion and delayed lethality in vivo in a murine peritonitis model. PGE2 was shown to inhibit caspase-11-driven pyroptosis and IL-1ß secretion in macrophages. Overall, we provide evidence for a pathological role of caspase-4/11 in peptic ulcer disease and propose that targeting caspase-4 or inhibiting pyroptosis may have therapeutic potential in the management of peptic ulcers.


Subject(s)
Caspases, Initiator/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/pathogenicity , Interleukin-1beta/metabolism , Peptic Ulcer/metabolism , Animals , Caspases, Initiator/drug effects , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Humans , Inflammasomes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Misoprostol/pharmacology , Peptic Ulcer/pathology , Pyroptosis/drug effects
4.
Cancer Immunol Immunother ; 69(12): 2635-2649, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32613271

ABSTRACT

Barrett's esophagus (BE) is an inflammatory condition and a neoplastic precursor to esophageal adenocarcinoma (EAC). Inflammasome signaling, which contributes to acute and chronic inflammation, results in caspase-1 activation leading to the secretion of IL-1ß and IL-18, and inflammatory cell death (pyroptosis). This study aimed to characterize caspase-1 expression, and its functional importance, during disease progression to BE and EAC. Three models of disease progression (Normal-BE-EAC) were employed to profile caspase-1 expression: (1) a human esophageal cell line model; (2) a murine model of BE; and (3) resected tissue from BE-associated EAC patients. BE patient biopsies and murine BE organoids were cultured ex vivo in the presence of a caspase-1 inhibitor, to determine the importance of caspase-1 for inflammatory cytokine and chemokine secretion.Epithelial caspase-1 expression levels were significantly enhanced in BE (p < 0.01). In contrast, stromal caspase-1 levels correlated with histological inflammation scores during disease progression (p < 0.05). Elevated secretion of IL-1ß from BE explanted tissue, compared to adjacent normal tissue (p < 0.01), confirmed enhanced activity of caspase-1 in BE tissue. Caspase-1 inhibition in LPS-stimulated murine BE organoids caused a significant reduction in IL-1ß (p < 0.01) and CXCL1 (p < 0.05) secretion, confirming the importance of caspase-1 in the production of cytokines and chemokines associated with disease progression from BE to EAC. Targeting caspase-1 activity in BE patients should therefore be tested as a novel strategy to prevent inflammatory complications associated with disease progression.


Subject(s)
Adenocarcinoma/immunology , Barrett Esophagus/immunology , Caspase 1/metabolism , Esophageal Mucosa/pathology , Esophageal Neoplasms/immunology , Inflammasomes/immunology , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Aged , Animals , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Biopsy , Caspase 1/immunology , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Cells, Cultured , Chemokine CXCL1/metabolism , Disease Models, Animal , Disease Progression , Esophageal Mucosa/cytology , Esophageal Mucosa/immunology , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Esophagectomy , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Primary Cell Culture , Signal Transduction/drug effects , Signal Transduction/immunology
5.
Nat Commun ; 11(1): 1055, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32103022

ABSTRACT

Activated caspase-1 and caspase-11 induce inflammatory cell death in a process termed pyroptosis. Here we show that Prostaglandin E2 (PGE2) inhibits caspase-11-dependent pyroptosis in murine and human macrophages. PGE2 suppreses caspase-11 expression in murine and human macrophages and in the airways of mice with allergic inflammation. Remarkably, caspase-11-deficient mice are strongly resistant to developing experimental allergic airway inflammation, where PGE2 is known to be protective. Expression of caspase-11 is elevated in the lung of wild type mice with allergic airway inflammation. Blocking PGE2 production with indomethacin enhances, whereas the prostaglandin E1 analog misoprostol inhibits lung caspase-11 expression. Finally, alveolar macrophages from asthma patients exhibit increased expression of caspase-4, a human homologue of caspase-11. Our findings identify PGE2 as a negative regulator of caspase-11-driven pyroptosis and implicate caspase-4/11 as a critical contributor to allergic airway inflammation, with implications for pathophysiology of asthma.


Subject(s)
Asthma/pathology , Caspases, Initiator/metabolism , Dinoprostone/metabolism , Macrophages/immunology , Pyroptosis/physiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Asthma/immunology , Caspases, Initiator/genetics , Caspases, Initiator/immunology , Cells, Cultured , Drug Synergism , Female , Humans , Indomethacin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Misoprostol/pharmacology
6.
Oncogene ; 38(14): 2658-2674, 2019 04.
Article in English | MEDLINE | ID: mdl-30538296

ABSTRACT

Murine inflammatory caspase-11 has an important role in intestinal epithelial inflammation and barrier function. Activation of the non-canonical inflammasome, mediated by caspase-11, serves as a regulatory pathway for the production of the pro-inflammatory cytokines IL-1ß and IL-18, and has a key role in pyroptotic cell death. We have previously demonstrated a protective role for caspase-11 during dextran sulphate sodium (DSS)-induced colitis, however the importance of caspase-11 during colorectal tumour development remains unclear. Here, we show that Casp11-/- mice are highly susceptible to the azoxymethane (AOM)-DSS model of colitis-associated cancer (CAC), compared to their wild type (WT) littermates. We show that deficient IL-18 production occurs at initial inflammation stages of disease, and that IL-1ß production is more significantly impaired in Casp11-/- colons during established CAC. We identify defective STAT1 activation in Casp11-/- colons during disease progression, and show that IL-1ß signalling induces caspase-11 expression and STAT1 activation in primary murine macrophages and intestinal epithelial cells. These findings uncover an anti-tumour role for the caspase-11 and the non-canonical inflammasome during CAC, and suggest a critical role for caspase-11, linking IL-1ß and STAT1 signalling pathways.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Caspases/metabolism , Colitis/metabolism , Colitis/pathology , STAT1 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Caspases, Initiator , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Female , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
7.
J Immunol ; 198(9): 3558-3564, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28298525

ABSTRACT

PGE2 has been shown to increase the transcription of pro-IL-1ß. However, recently it has been demonstrated that PGE2 can block the maturation of IL-1ß by inhibiting the NLRP3 inflammasome in macrophages. These apparently conflicting results have led us to reexamine the effect of PGE2 on IL-1ß production. We have found that in murine bone marrow-derived macrophages, PGE2 via the cAMP/protein kinase A pathway is potently inducing IL-1ß transcription, as well as boosting the ability of LPS to induce IL-1ß mRNA and pro-IL-1ß while inhibiting the production of TNF-α. This results in an increase in mature IL-1ß production in macrophages treated with ATP. We also examined the effect of endogenously produced PGE2 on IL-1ß production. By blocking PGE2 production with indomethacin, we made a striking finding that endogenous PGE2 is essential for LPS-induced pro-IL-1ß production, suggesting a positive feedback loop. The effect of endogenous PGE2 was mediated by EP2 receptor. In primary human monocytes, where LPS alone is sufficient to induce mature IL-1ß, PGE2 boosted LPS-induced IL-1ß production. PGE2 did not inhibit ATP-induced mature IL-1ß production in monocytes. Because PGE2 mediates the pyrogenic effect of IL-1ß, these effects might be especially relevant for the role of monocytes in the induction of fever. A positive feedback loop from IL-1ß and back to PGE2, which itself is induced by IL-1ß, is likely to be operating. Furthermore, fever might therefore occur in the absence of a septic shock response because of the inhibiting effect of PGE2 on TNF-α production.


Subject(s)
Dinoprostone/metabolism , Fever/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Monocytes/immunology , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Dinoprostone/antagonists & inhibitors , Feedback, Physiological , Humans , Indomethacin/pharmacology , Inflammasomes/metabolism , Interleukin-1beta/genetics , Lipopolysaccharides/immunology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Monocytes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...