Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Br J Dermatol ; 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38913409

ABSTRACT

Hidradenitis suppurativa (HS) is a chronic inflammatory skin disease characterised by recurrent inflammatory lesions, which affect skin and hair follicles in intertriginous areas. HS has a multifactorial aetiology resulting in barrier dysfunction associated with aberrant immune activation. There is increased evidence for the role of inflammasomes in the pathophysiology of inflammatory skin diseases, including HS. Inflammasomes are multiprotein complexes activated following exposure to danger signals including microbial ligands and components of damaged host cells. Inflammasome activation induces many signalling cascades and subsequent cleavage of pro-inflammatory cytokines, most notably interleukin (IL)-1ß, which have a role in HS pathogenesis. Limited immunotherapies are approved for treating moderate-to-severe HS, with variable response rates influenced by disease heterogeneity. Inflammasomes represent attractive targets to suppress multiple inflammatory pathways in HS including IL-1ß and IL-17. This review aims to summarise the role of inflammasomes in HS and to evaluate evidence for inflammasomes as therapeutic targets for HS treatment.

3.
Exp Dermatol ; 30(10): 1554-1568, 2021 10.
Article in English | MEDLINE | ID: mdl-34418166

ABSTRACT

The innate immune system of human skin consists of a multi-layered barrier consisting of cells and soluble effector molecules charged with maintaining homeostasis and responding to insults and infections. It has become increasingly clear that these barrier layers become compromised in skin diseases, especially in disorders of an (auto)inflammatory nature. In the case of hidradenitis suppurativa, great strides have been made in recent years in characterizing the underlying breakdown in homeostatic innate immunity, including an increasing understanding of the central role of the hair follicle in this process. This breakdown appears to occur at multiple levels: the pilosebaceous unit, associated epithelium, the cutaneous microbiome, alteration of immune cell function and local molecular events such as complement activation. This review seeks to summarize, contextualize and analyse critically our current understanding of how these innate immune barriers become dysregulated in the early stage(s) of hidradenitis suppurativa, and to speculate on where potential hidradenitis suppurativa research could be most fruitful.


Subject(s)
Hidradenitis Suppurativa/immunology , Immunity, Innate/immunology , Microbiota/immunology , Antimicrobial Peptides/immunology , Humans
4.
Cell Rep ; 30(1): 124-136.e4, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914380

ABSTRACT

Increased glycolytic metabolism recently emerged as an essential process driving host defense against Mycobacterium tuberculosis (Mtb), but little is known about how this process is regulated during infection. Here, we observe repression of host glycolysis in Mtb-infected macrophages, which is dependent on sustained upregulation of anti-inflammatory microRNA-21 (miR-21) by proliferating mycobacteria. The dampening of glycolysis by miR-21 is mediated through targeting of phosphofructokinase muscle (PFK-M) isoform at the committed step of glycolysis, which facilitates bacterial growth by limiting pro-inflammatory mediators, chiefly interleukin-1ß (IL-1ß). Unlike other glycolytic genes, PFK-M expression and activity is repressed during Mtb infection through miR-21-mediated regulation, while other less-active isoenzymes dominate. Notably, interferon-γ (IFN-γ), which drives Mtb host defense, inhibits miR-21 expression, forcing an isoenzyme switch in the PFK complex, augmenting PFK-M expression and macrophage glycolysis. These findings place the targeting of PFK-M by miR-21 as a key node controlling macrophage immunometabolic function.


Subject(s)
Glycolysis , Host-Pathogen Interactions , Interleukin-1beta/metabolism , MicroRNAs/metabolism , Mycobacterium tuberculosis/physiology , Phosphofructokinase-1/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Base Sequence , Cell Proliferation , HEK293 Cells , Humans , Interferon-gamma/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , MicroRNAs/genetics , Phosphofructokinase-1/genetics , RAW 264.7 Cells , Tuberculosis/microbiology
5.
Immunity ; 50(2): 432-445.e7, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30683619

ABSTRACT

Host microbial cross-talk is essential to maintain intestinal homeostasis. However, maladaptation of this response through microbial dysbiosis or defective host defense toward invasive intestinal bacteria can result in chronic inflammation. We have shown that macrophages differentiated in the presence of the bacterial metabolite butyrate display enhanced antimicrobial activity. Butyrate-induced antimicrobial activity was associated with a shift in macrophage metabolism, a reduction in mTOR kinase activity, increased LC3-associated host defense and anti-microbial peptide production in the absence of an increased inflammatory cytokine response. Butyrate drove this monocyte to macrophage differentiation program through histone deacetylase 3 (HDAC3) inhibition. Administration of butyrate induced antimicrobial activity in intestinal macrophages in vivo and increased resistance to enteropathogens. Our data suggest that (1) increased intestinal butyrate might represent a strategy to bolster host defense without tissue damaging inflammation and (2) that pharmacological HDAC3 inhibition might drive selective macrophage functions toward antimicrobial host defense.


Subject(s)
Anti-Infective Agents/pharmacology , Butyrates/pharmacology , Cell Differentiation/drug effects , Macrophages/drug effects , Monocytes/drug effects , Animals , Cell Differentiation/genetics , Cells, Cultured , Colon/drug effects , Colon/metabolism , Colon/microbiology , Cytokines/genetics , Cytokines/metabolism , Dysbiosis/microbiology , Gene Expression Regulation/drug effects , Humans , Intestines/drug effects , Intestines/microbiology , Macrophages/metabolism , Macrophages/microbiology , Mice, Inbred C57BL , Microbiota/drug effects , Microbiota/physiology , Monocytes/metabolism , Monocytes/microbiology
6.
J Crohns Colitis ; 12(7): 835-848, 2018 Jun 28.
Article in English | MEDLINE | ID: mdl-29608690

ABSTRACT

BACKGROUND AND AIMS: microRNAs regulate gene expression and influence the pathogenesis of human diseases. The present study investigated the role of microRNA-21 [miR-21] in the pathogenesis of intestinal inflammation, because miR-21 is highly expressed in inflammatory bowel disease. Inflammatory bowel disease is associated with intestinal barrier dysfunction and an altered gut microbiota. Recent studies have demonstrated that host microRNAs can shape the microbiota. Thus, we determined the influence of miR-21 on the gut microbiota and observed the subsequent impact in a dextran sodium sulphate [DSS]-induced colitis model. METHODS: The influence of miR-21 on the gut microbiota and inflammation was assessed in wild-type [WT] and miR-21-/- mice, in co-housed mice, following antibiotic depletion of the microbiota, or by colonization of germ-free [GF] mice with fecal homogenate, prior to DSS administration. We carried out 16S rRNA sequencing on WT and miR-21-/- mice to dissect potential differences in the gut microbiota. RESULTS: miR-21-/- mice have reduced susceptibility to DSS-induced colitis compared with WT mice. Co-housing conferred some protection to WT mice, while GF mice colonized with fecal homogenate from miR-21-/- were protected from DSS colitis compared with those colonized with WT homogenate. Further supporting a role for the microbiota in the observed phenotype, the protection afforded by miR-21 depletion was lost when mice were pre-treated with antibiotics. The 16S rRNA sequencing revealed significant differences in the composition of WT and miR-21-/- intestinal microbiota. CONCLUSIONS: These findings suggest that miR-21 influences the pathogenesis of intestinal inflammation by causing propagation of a disrupted gut microbiota.


Subject(s)
Colitis/genetics , Colitis/microbiology , Gastrointestinal Microbiome/genetics , Genetic Predisposition to Disease , MicroRNAs/genetics , Animals , Anti-Bacterial Agents/pharmacology , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate , Disease Models, Animal , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Gene Deletion , Male , Mice , Protective Factors , RNA, Ribosomal, 16S/analysis
7.
Article in English | MEDLINE | ID: mdl-28589100

ABSTRACT

MiRNAs are important post-transcriptional regulators of gene expression. MiRNA expression is a crucial part of host responses to bacterial infection, however there is limited knowledge of their impact on the outcome of infections. We investigated the influence of miR-21 on macrophage responses during infection with Listeria monocytogenes, which establishes an intracellular niche within macrophages. MiR-21 is induced following infection of bone marrow-derived macrophages (BMDMs) with Listeria. MiR-21-/- macrophages display an increased bacterial burden with Listeria at 30 min and 2 h post-infection. This phenotype was reversed by the addition of synthetic miR-21 mimics to the system. To assess the immune response of wildtype (WT) and miR-21-/- macrophages, BMDMs were treated with bacterial LPS or infected with Listeria. There was no difference in IL-10 and IL-6 between WT and miR-21-/- BMDMs in response to LPS or Listeria. TNF-α was increased in miR-21-/- BMDMs stimulated with LPS or Listeria compared to WT macrophages. We next assessed the production of nitric oxide (NO), a key bactericidal factor in Listeria infection. There was no significant difference in NO production between WT and miR-21-/- cells, indicating that the increased bacterial burden may not be due to impaired killing. As the increased bacterial load was observed early following infection (30 min), we questioned whether this is due to differences in uptake of Listeria by WT and miR-21-/- macrophages. We show that miR-21-deficiency enhances uptake of FITC-dextran and FITC-Escherichia coli bioparticles by macrophages. The previously observed Listeria burden phenotype was ablated by pre-treatment of cells with the actin polymerization inhibitor cytochalasin-D. From analysis of miR-21 targets, we selected the pro-phagocytic regulators myristoylated alanine-rich C-kinase substrate (MARCKS) and Ras homolog gene family, member B (RhoB) for further investigation. MARCKS and RhoB are increased in miR-21-/- BMDMs, correlating with increased uptake of Listeria. Finally, intra-peritoneal infection of mice with Listeria led to increased bacterial burden in livers of miR-21-/- mice compared to WT mice. These findings suggest a possible role for miR-21 in regulation of phagocytosis during infection, potentially by repression of MARCKS and RhoB, thus serving to limit the availability of the intracellular niche of pathogens like L. monocytogenes.


Subject(s)
Listeria monocytogenes/immunology , Listeriosis/immunology , Macrophages/immunology , Macrophages/microbiology , MicroRNAs/metabolism , Animals , Cytochalasin D/metabolism , Cytokines/metabolism , Cytoplasm/microbiology , Gene Expression , Interleukin-10/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/immunology , Myristoylated Alanine-Rich C Kinase Substrate , Nitric Oxide/metabolism , Phagocytosis , Tumor Necrosis Factor-alpha/metabolism , rho GTP-Binding Proteins/metabolism
8.
Methods Mol Biol ; 1390: 287-300, 2016.
Article in English | MEDLINE | ID: mdl-26803636

ABSTRACT

Epithelial barrier function and innate immunity are fundamental to the pathogenesis of inflammatory and infectious disease. Along with plasma membranes, epithelial cells are the primary cellular determinant of epithelial barrier function. The mechanism by which polarized epithelia form a permeability barrier is of fundamental importance to the prevention of many infectious and inflammatory diseases. Moreover, epithelial cells express Toll-like receptors (TLRs) which upon recognition of conserved microbial factors such as lipopolysaccharide (LPS) induce epithelial responses including epithelial cell proliferation, secretion of secretory IgA into the lumen and production mucins and antimicrobial peptides, thereby promoting intestinal barrier function. Understanding gut barrier integrity and regulation of permeability is crucial to increase our understanding of the pathogenesis of intestinal disease. A variety of tests have been developed to assess this barrier, including assessing intestinal epithelial cell proliferation or death, intestinal tight junction status and the consequence of intestinal barrier integrity loss such as increased intestinal permeability and susceptibility to bacterial infection. Using a mouse model, this chapter describes some of the methods to assess the functional integrity of this epithelial barrier and the part played by a TLR signalling pathway.


Subject(s)
Intestinal Mucosa/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , Animals , Apoptosis , Bone Marrow Transplantation , Disease Models, Animal , Epithelial Cells/metabolism , Mice , Mice, Knockout , Permeability , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella typhimurium , Tight Junctions/metabolism , Transplantation Chimera
9.
Cell Metab ; 21(1): 65-80, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25565206

ABSTRACT

Macrophages activated by the TLR4 agonist LPS undergo dramatic changes in their metabolic activity. We here show that LPS induces expression of the key metabolic regulator Pyruvate Kinase M2 (PKM2). Activation of PKM2 using two well-characterized small molecules, DASA-58 and TEPP-46, inhibited LPS-induced Hif-1α and IL-1ß, as well as the expression of a range of other Hif-1α-dependent genes. Activation of PKM2 attenuated an LPS-induced proinflammatory M1 macrophage phenotype while promoting traits typical of an M2 macrophage. We show that LPS-induced PKM2 enters into a complex with Hif-1α, which can directly bind to the IL-1ß promoter, an event that is inhibited by activation of PKM2. Both compounds inhibited LPS-induced glycolytic reprogramming and succinate production. Finally, activation of PKM2 by TEPP-46 in vivo inhibited LPS and Salmonella typhimurium-induced IL-1ß production, while boosting production of IL-10. PKM2 is therefore a critical determinant of macrophage activation by LPS, promoting the inflammatory response.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Pyruvate Kinase/metabolism , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Enzyme Activators/pharmacology , Gene Expression/drug effects , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Interleukin-1beta/genetics , Lipopolysaccharides/toxicity , Macrophage Activation/drug effects , Macrophages/cytology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Protein Binding , Pyruvate Kinase/chemistry , Pyruvate Kinase/genetics , RNA, Messenger/metabolism , Salmonella typhimurium/physiology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/metabolism
11.
Am J Physiol Gastrointest Liver Physiol ; 306(9): G769-78, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24603458

ABSTRACT

Toll-like receptors (TLRs) play a central role in the recognition and response to microbial pathogens and in the maintenance and function of the epithelial barrier integrity in the gut. The protein MyD88 adaptor-like (Mal/TIRAP) serves as a bridge between TLR2/TLR4- and MyD88-mediated signaling to orchestrate downstream inflammatory responses. Whereas MyD88 has an essential function in the maintenance of intestinal homeostasis, a role for Mal in this context is less well described. Colitis was induced in wild-type (WT) and Mal-deficient (Mal(-/-)) mice by administration of dextran sodium sulfate (DSS). Colitis-associated cancer was induced by DSS and azoxymethane (AOM) treatment. Chimeric mice were generated by total body gamma irradiation followed by transplantation of bone marrow cells. In the DSS model of colon epithelial injury, Mal(-/-) mice developed increased inflammation and severity of colitis relative to WT mice. Mal(-/-) mice demonstrated the presence of inflammatory cell infiltrates, increased crypt proliferation, and presence of neoformations. Furthermore, in the AOM/DSS model, Mal(-/-) mice had greater incidence of tumors. Mal(-/-) and WT bone marrow chimeras demonstrated that nonhematopoietic cell expression of Mal had an important protective role in the control of intestinal inflammation and inflammation-associated cancer. Mal is essential for the maintenance of intestinal homeostasis and expression of Mal in nonhematopoietic cells prevents chronic intestinal inflammation that may predispose to colon neoplasia.


Subject(s)
Colitis/etiology , Colon/metabolism , Colorectal Neoplasms/etiology , Membrane Glycoproteins/metabolism , Receptors, Interleukin-1/metabolism , Animals , Azoxymethane , Bone Marrow Transplantation , Caco-2 Cells , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Colitis/prevention & control , Colon/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Dextran Sulfate , Disease Models, Animal , Female , Homeostasis , Humans , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Severity of Illness Index , Time Factors , Transplantation Chimera
12.
Mol Cell Neurosci ; 56: 148-58, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23659921

ABSTRACT

Accumulating evidence continues to underpin the role of the innate immune system in pathologies associated with neuroinflammation. Innate immunity is regulated by pattern recognition receptors that detect pathogens, and in the case of Gram-positive bacteria, binding of bacterial lipopeptides to toll-like receptor (TLR)2 is emerging as an important mechanism controlling glial cell activation. In the present study, we employed the use of the synthetic bacterial lipoprotein and a selective TLR2 agonist, Pam3CSK4, to induce inflammatory signaling in microglia and astrocytes. The adaptor proteins, downstream of kinase (Dok)1 and Dok2, are known to have a role in negatively regulating the Ras-ERK signaling cascade, with downstream consequences on pro-inflammatory cytokine expression. Data presented herein demonstrate that TLR2 enhanced the tyrosine phosphorylation of Dok1 and Dok2 in astrocytes and microglia, and that knockdown of these adaptors using small interfering RNA robustly elevated TLR2-induced ERK activation. Importantly, TLR2-induced NF-κB activation, and IL-6 production was exacerbated in astrocytes transfected with Dok1 and Dok2 siRNA, indicating that both Dok proteins negatively regulate TLR2-induced inflammatory signaling in astrocytes. In contrast, Dok1 knockdown attenuated TLR2-induced NF-κB activation and IL-6 production in microglia, while Dok2 siRNA failed to affect TLR2-induced NF-κB activity and subsequent cytokine expression in this cell type. Overall, this indicates that Dok1 and Dok2 are novel adaptors for TLR2 in glial cells and importantly indicates that Dok1 and Dok2 differentially regulate TLR2-induced pro-inflammatory signaling in astrocytes and microglia.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Astrocytes/metabolism , DNA-Binding Proteins/metabolism , MAP Kinase Signaling System , Microglia/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Toll-Like Receptor 2/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopeptides/pharmacology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...