Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Eur Respir J ; 61(5)2023 05.
Article in English | MEDLINE | ID: mdl-36922030

ABSTRACT

BACKGROUND: COVID-19 is associated with a dysregulated immune response but it is unclear how immune dysfunction contributes to the chronic morbidity persisting in many COVID-19 patients during convalescence (long COVID). METHODS: We assessed phenotypical and functional changes of monocytes in COVID-19 patients during hospitalisation and up to 9 months of convalescence following COVID-19, respiratory syncytial virus or influenza A. Patients with progressive fibrosing interstitial lung disease were included as a positive control for severe, ongoing lung injury. RESULTS: Monocyte alterations in acute COVID-19 patients included aberrant expression of leukocyte migration molecules, continuing into convalescence (n=142) and corresponding with specific symptoms of long COVID. Long COVID patients with unresolved lung injury, indicated by sustained shortness of breath and abnormal chest radiology, were defined by high monocyte expression of C-X-C motif chemokine receptor 6 (CXCR6) (p<0.0001) and adhesion molecule P-selectin glycoprotein ligand 1 (p<0.01), alongside preferential migration of monocytes towards the CXCR6 ligand C-X-C motif chemokine ligand 16 (CXCL16) (p<0.05), which is abundantly expressed in the lung. Monocyte CXCR6 and lung CXCL16 were heightened in patients with progressive fibrosing interstitial lung disease (p<0.001), confirming a role for the CXCR6-CXCL16 axis in ongoing lung injury. Conversely, monocytes from long COVID patients with ongoing fatigue exhibited a sustained reduction of the prostaglandin-generating enzyme cyclooxygenase 2 (p<0.01) and CXCR2 expression (p<0.05). These monocyte changes were not present in respiratory syncytial virus or influenza A convalescence. CONCLUSIONS: Our data define unique monocyte signatures that define subgroups of long COVID patients, indicating a key role for monocyte migration in COVID-19 pathophysiology. Targeting these pathways may provide novel therapeutic opportunities in COVID-19 patients with persistent morbidity.


Subject(s)
COVID-19 , Influenza, Human , Lung Injury , Humans , Monocytes/metabolism , Chemokines, CXC/metabolism , Receptors, Virus/metabolism , Receptors, CXCR6 , Receptors, Chemokine/metabolism , Post-Acute COVID-19 Syndrome , Ligands , Convalescence , Receptors, Scavenger/metabolism , Chemokine CXCL16 , Patient Acuity
2.
Immunology ; 166(1): 138-152, 2022 05.
Article in English | MEDLINE | ID: mdl-35199335

ABSTRACT

Intestinal macrophages play a vital role in the maintenance of gut homeostasis through signals derived from the microbiota. We previously demonstrated that microbial-derived metabolites can shape the metabolic functions of macrophages. Here, we show that antibiotic-induced disruption of the intestinal microbiota dramatically alters both the local metabolite environment and the metabolic functions of macrophages in the colon. Broad-spectrum antibiotic administration in mice increased the expression of the large neutral amino acid transporter LAT1 and accordingly, amino acid uptake. Subsequently, antibiotic administration enhanced the metabolic functions of colonic macrophages, increasing phosphorylation of components of mammalian/mechanistic target of rapamycin signalling pathways, with increased expression of genes involved in glycolysis and oxidative phosphorylation (OXPHOS), increased mitochondrial function, increased rate of extracellular acidification (ECAR; measure of glycolysis) and increased rate of oxygen consumption (OCR; measure of OXPHOS). Small bowel macrophages were less metabolically active than their colonic counterparts, with macrophage metabolism in the small intestine being independent of the microbiota. Finally, we reveal tissue-resident Tim4+  CD4+ macrophages exhibit enhanced fatty acid uptake alongside reduced fatty acid synthesis compared to recruited macrophages. Thus, the microbiota shapes gut macrophage metabolism in a compartment-specific manner, with important implications for monocyte recruitment and macrophage differentiation.


Subject(s)
Gastrointestinal Microbiome , Macrophages , Animals , Anti-Bacterial Agents/pharmacology , Colon , Fatty Acids/metabolism , Macrophages/metabolism , Mammals , Mice
3.
Immunology ; 159(1): 26-38, 2020 01.
Article in English | MEDLINE | ID: mdl-31777068

ABSTRACT

Mucosal tissues contain distinct microbial communities that differ drastically depending on the barrier site, and as such, mucosal immune responses have evolved to be tailored specifically for their location. Whether protective or regulatory immune responses against invading pathogens or the commensal microbiota occur is controlled by local mononuclear phagocytes (MNPs). Comprising macrophages and dendritic cells (DCs), the functions of these cells are highly dependent on the local environment. For example, the intestine contains the greatest bacterial load of any site in the body, and hence, intestinal MNPs are hyporesponsive to bacterial stimulation. This is thought to be one of the major mechanisms by which harmful immune responses directed against the trillions of harmless bacteria that line the gut lumen are avoided. Regulation of MNP function by the microbiota has been characterized in the most depth in the intestine but there are several mucosal sites that also contain their own microbiota. In this review, we present an overview of how MNP function is regulated by the microbiota at mucosal sites, highlighting recent novel pathways by which this occurs in the intestine, and new studies elucidating these interactions at mucosal sites that have been characterized in less depth, including the urogenital tract.


Subject(s)
Immunity, Mucosal , Microbiota/immunology , Mononuclear Phagocyte System/immunology , Mucous Membrane/microbiology , Symbiosis/immunology , Animals , Host Microbial Interactions/immunology , Humans , Immune Tolerance , Intestines/immunology , Intestines/microbiology , Lung/microbiology , Models, Animal , Mouth/immunology , Mouth/microbiology , Mucous Membrane/immunology , Urogenital System/immunology , Urogenital System/microbiology
4.
PLoS One ; 14(12): e0225021, 2019.
Article in English | MEDLINE | ID: mdl-31821343

ABSTRACT

Type 1, or autoimmune, diabetes is caused by the T-cell mediated destruction of the insulin-producing pancreatic beta cells. Non-obese diabetic (NOD) mice spontaneously develop autoimmune diabetes akin to human type 1 diabetes. For this reason, the NOD mouse has been the preeminent murine model for human type 1 diabetes research for several decades. However, humanized mouse models are highly sought after because they offer both the experimental tractability of a mouse model and the clinical relevance of human-based research. Autoimmune T-cell responses against insulin, and its precursor proinsulin, play central roles in the autoimmune responses against pancreatic beta cells in both humans and NOD mice. As a first step towards developing a murine model of the human autoimmune response against pancreatic beta cells we set out to replace the murine insulin 1 gene (Ins1) with the human insulin gene (Ins) using CRISPR/Cas9. Here we describe a NOD mouse strain that expresses human insulin in place of murine insulin 1, referred to as HuPI. HuPI mice express human insulin, and C-peptide, in their serum and pancreata and have normal glucose tolerance. Compared with wild type NOD mice, the incidence of diabetes is much lower in HuPI mice. Only 15-20% of HuPI mice developed diabetes after 300 days, compared to more than 60% of unmodified NOD mice. Immune-cell infiltration into the pancreatic islets of HuPI mice was not detectable at 100 days but was clearly evident by 300 days. This work highlights the feasibility of using CRISPR/Cas9 to create mouse models of human diseases that express proteins pivotal to the human disease. Furthermore, it reveals that even subtle changes in proinsulin protect NOD mice from diabetes.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Insulin-Secreting Cells/metabolism , Insulin/genetics , Animals , CRISPR-Cas Systems , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Inbred NOD , Pancreas/metabolism
5.
Front Immunol ; 9: 2800, 2018.
Article in English | MEDLINE | ID: mdl-30555479

ABSTRACT

Diabetogenic T cells infiltrate the pancreatic islets by transmigrating across the microcapillaries residing close to, or within, the pancreatic islets. Deficiency in IFNγ signaling prevents efficient migration of T cells into the pancreatic islets, but the IFNγ-regulated molecules that mediate this are uncertain. Homing of autoreactive T cells into target tissues may require antigen specificity through presentation of cognate antigen by MHC expressed on the vascular endothelium. We investigated the hypothesis that IFNγ promotes the migration of islet antigen-specific CD4+ T cells by upregulating MHC class II on islet endothelial cells (IEC), thereby providing an antigen-specific signal for islet infiltration. Upon IFNγ stimulation, MHC class II, which is not constitutively expressed on IEC, was induced. IFNγ-dependent upregulation of MHC class II was detected in IEC isolated from prediabetic NOD mice at the earliest stages of insulitis, before other markers of inflammation were present. Using a CD4+ T cell-mediated adoptive transfer model of autoimmune diabetes we observed that even though diabetes does not develop in recipient mice lacking IFNγ receptors, mice with MHC class II-deficient IEC were not protected from disease. Thus, IFNγ-regulated molecules, but not MHC class II or antigen presentation by IECs is required for the early migration of antigen-specific CD4+ T cells into the pancreatic islets.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endothelial Cells/immunology , Histocompatibility Antigens Class II/immunology , Interferon-gamma/immunology , Islets of Langerhans/immunology , Prediabetic State/immunology , Animals , Biomarkers , CD4-Positive T-Lymphocytes/pathology , Endothelial Cells/pathology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class II/genetics , Interferon-gamma/genetics , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Prediabetic State/genetics , Prediabetic State/pathology
6.
Sci Transl Med ; 10(464)2018 10 24.
Article in English | MEDLINE | ID: mdl-30355800

ABSTRACT

Macrophages in the healthy intestine are highly specialized and usually respond to the gut microbiota without provoking an inflammatory response. A breakdown in this tolerance leads to inflammatory bowel disease (IBD), but the mechanisms by which intestinal macrophages normally become conditioned to promote microbial tolerance are unclear. Strong epidemiological evidence linking disruption of the gut microbiota by antibiotic use early in life to IBD indicates an important role for the gut microbiota in modulating intestinal immunity. Here, we show that antibiotic use causes intestinal macrophages to become hyperresponsive to bacterial stimulation, producing excess inflammatory cytokines. Re-exposure of antibiotic-treated mice to conventional microbiota induced a long-term, macrophage-dependent increase in inflammatory T helper 1 (TH1) responses in the colon and sustained dysbiosis. The consequences of this dysregulated macrophage activity for T cell function were demonstrated by increased susceptibility to infections requiring TH17 and TH2 responses for clearance (bacterial Citrobacter rodentium and helminth Trichuris muris infections), corresponding with increased inflammation. Short-chain fatty acids (SCFAs) were depleted during antibiotic administration; supplementation of antibiotics with the SCFA butyrate restored the characteristic hyporesponsiveness of intestinal macrophages and prevented T cell dysfunction. Butyrate altered the metabolic behavior of macrophages to increase oxidative phosphorylation and also promoted alternative macrophage activation. In summary, the gut microbiota is essential to maintain macrophage-dependent intestinal immune homeostasis, mediated by SCFA-dependent pathways. Oral antibiotics disrupt this process to promote sustained T cell-mediated dysfunction and increased susceptibility to infections, highlighting important implications of repeated broad-spectrum antibiotic use.


Subject(s)
Anti-Bacterial Agents/pharmacology , Homeostasis/drug effects , Immunity, Innate/drug effects , Intestines/cytology , Macrophages/metabolism , T-Lymphocytes/immunology , Animals , Butyrates/pharmacology , Cytokines/metabolism , Fatty Acids/metabolism , Gastrointestinal Microbiome/drug effects , Inflammation/pathology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice, Inbred C57BL , Receptors, CCR2/metabolism , T-Lymphocytes/drug effects , Th1 Cells/drug effects
7.
Diabetes ; 66(6): 1650-1660, 2017 06.
Article in English | MEDLINE | ID: mdl-28292965

ABSTRACT

Recent advances in immunotherapeutics have not yet changed the routine management of autoimmune type 1 diabetes. There is an opportunity to repurpose therapeutics used to treat other diseases to treat type 1 diabetes, especially when there is evidence for overlapping mechanisms. Janus kinase (JAK) 1/JAK2 inhibitors are in development or clinical use for indications including rheumatoid arthritis. There is good evidence for activation of the JAK1/JAK2 and signal transducer and activator of transcription (STAT) 1 pathway in human type 1 diabetes and in mouse models, especially in ß-cells. We tested the hypothesis that using these drugs to block the JAK-STAT pathway would prevent autoimmune diabetes. The JAK1/JAK2 inhibitor AZD1480 blocked the effect of cytokines on mouse and human ß-cells by inhibiting MHC class I upregulation. This prevented the direct interaction between CD8+ T cells and ß-cells, and reduced immune cell infiltration into islets. NOD mice treated with AZD1480 were protected from autoimmune diabetes, and diabetes was reversed in newly diagnosed NOD mice. This provides mechanistic groundwork for repurposing clinically approved JAK1/JAK2 inhibitors for type 1 diabetes.


Subject(s)
Blood Glucose/metabolism , CD8-Positive T-Lymphocytes/drug effects , Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens Class II/drug effects , Insulin-Secreting Cells/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10/immunology , Cytokines/immunology , Diabetes Mellitus, Type 1/metabolism , Flow Cytometry , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Immunohistochemistry , In Vitro Techniques , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/immunology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Mice , Mice, Inbred NOD , Up-Regulation
8.
Eur J Immunol ; 45(9): 2494-503, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25959978

ABSTRACT

Type 1 diabetes results from destruction of pancreatic beta cells by autoreactive T cells. Both CD4(+) and CD8(+) T cells have been shown to mediate beta-cell killing. While CD8(+) T cells can directly recognize MHC class I on beta cells, the interaction between CD4(+) T cells and beta cells remains unclear. Genetic association studies have strongly implicated HLA-DQ alleles in human type 1 diabetes. Here we studied MHC class II expression on beta cells in nonobese diabetic mice that were induced to develop diabetes by diabetogenic CD4(+) T cells with T-cell receptors that recognize beta-cell antigens. Acute infiltration of CD4(+) T cells in islets occurred with rapid onset of diabetes. Beta cells from islets with immune infiltration expressed MHC class II mRNA and protein. Exposure of beta cells to IFN-γ increased MHC class II gene expression, and blocking IFN-γ signaling in beta cells inhibited MHC class II upregulation. IFN-γ also increased HLA-DR expression in human islets. MHC class II(+) beta cells stimulated the proliferation of beta-cell-specific CD4(+) T cells. Our study indicates that MHC class II molecules may play an important role in beta-cell interaction with CD4(+) T cells in the development of type 1 diabetes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , HLA-DQ Antigens/immunology , HLA-DR Antigens/immunology , Insulin-Secreting Cells/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Communication/immunology , Cell Proliferation/drug effects , Cells, Cultured , Cytotoxicity, Immunologic , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Gene Expression Regulation , HLA-DQ Antigens/genetics , HLA-DR Antigens/genetics , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Interferon-gamma/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred NOD , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Tissue Culture Techniques
9.
Diabetologia ; 58(1): 140-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25301392

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetes results from T cell-mediated destruction of pancreatic beta cells. The mechanisms of beta cell destruction in vivo, however, remain unclear. We aimed to test the relative roles of the main cell death pathways: apoptosis, necrosis and necroptosis, in beta cell death in the development of CD4(+) T cell-mediated autoimmune diabetes. METHODS: We altered expression levels of critical cell death proteins in mouse islets and tested their ability to survive CD4(+) T cell-mediated attack using an in vivo graft model. RESULTS: Loss of the B cell leukaemia/lymphoma 2 (BCL-2) homology domain 3-only proteins BIM, PUMA or BID did not protect beta cells from this death. Overexpression of the anti-apoptotic protein BCL-2 or combined deficiency of the pro-apoptotic multi-BCL2 homology domain proteins BAX and BAK also failed to prevent beta cell destruction. Furthermore, loss of function of the death receptor Fas or its essential downstream signalling molecule Fas-associated death domain (FADD) in islets was also not protective. Using electron microscopy we observed that dying beta cells showed features of necrosis. However, islets deficient in receptor-interacting serine/threonine protein kinase 3 (RIPK3), a critical initiator of necroptosis, were still normally susceptible to CD4(+) T cell-mediated destruction. Remarkably, simultaneous inhibition of apoptosis and necroptosis by combining loss of RIPK3 and overexpression of BCL-2 in islets did not protect them against immune attack either. CONCLUSIONS/INTERPRETATION: Collectively, our data indicate that beta cells die by necrosis in autoimmune diabetes and that the programmed cell death pathways apoptosis and necroptosis are both dispensable for this process.


Subject(s)
Autoimmunity/physiology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans Transplantation/immunology , Islets of Langerhans/pathology , T-Lymphocytes/immunology , Animals , Apoptosis/genetics , Apoptosis/physiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Necrosis/genetics , Necrosis/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Receptors, Death Domain/genetics , Receptors, Death Domain/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...