Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4326, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773113

ABSTRACT

Resolving inflammation is thought to return the affected tissue back to homoeostasis but recent evidence supports a non-linear model of resolution involving a phase of prolonged immune activity. Here we show that within days following resolution of Streptococcus pneumoniae-triggered lung inflammation, there is an influx of antigen specific lymphocytes with a memory and tissue-resident phenotype as well as macrophages bearing alveolar or interstitial phenotype. The transcriptome of these macrophages shows enrichment of genes associated with prostaglandin biosynthesis and genes that drive T cell chemotaxis and differentiation. Therapeutic depletion of post-resolution macrophages, inhibition of prostaglandin E2 (PGE2) synthesis or treatment with an EP4 antagonist, MF498, reduce numbers of lung CD4+/CD44+/CD62L+ and CD4+/CD44+/CD62L-/CD27+ T cells as well as their expression of the α-integrin, CD103. The T cells fail to reappear and reactivate upon secondary challenge for up to six weeks following primary infection. Concomitantly, EP4 antagonism through MF498 causes accumulation of lung macrophages and marked tissue fibrosis. Our study thus shows that PGE2 signalling, predominantly via EP4, plays an important role during the second wave of immune activity following resolution of inflammation. This secondary immune activation drives local tissue-resident T cell development while limiting tissue injury.


Subject(s)
Dinoprostone , Disease Models, Animal , Lung , Macrophages , Mice, Inbred C57BL , Pneumonia, Pneumococcal , Receptors, Prostaglandin E, EP4 Subtype , Streptococcus pneumoniae , Animals , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/pathology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/metabolism , Mice , Dinoprostone/metabolism , Streptococcus pneumoniae/immunology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Macrophages/immunology , Macrophages/metabolism , Lung/immunology , Lung/pathology , Lung/microbiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Integrin alpha Chains/metabolism , Integrin alpha Chains/genetics , Female , Antigens, CD/metabolism , Antigens, CD/genetics , T-Lymphocytes/immunology
2.
Front Oncol ; 13: 1156743, 2023.
Article in English | MEDLINE | ID: mdl-37342197

ABSTRACT

Background: Patient-derived xenograft (PDX) models involve the engraftment of tumour tissue in immunocompromised mice and represent an important pre-clinical oncology research method. A limitation of non-small cell lung cancer (NSCLC) PDX model derivation in NOD-scid IL2Rgammanull (NSG) mice is that a subset of initial engraftments are of lymphocytic, rather than tumour origin. Methods: The immunophenotype of lymphoproliferations arising in the lung TRACERx PDX pipeline were characterised. To present the histology data herein, we developed a Python-based tool for generating patient-level pathology overview figures from whole-slide image files; PATHOverview is available on GitHub (https://github.com/EpiCENTR-Lab/PATHOverview). Results: Lymphoproliferations occurred in 17.8% of lung adenocarcinoma and 10% of lung squamous cell carcinoma transplantations, despite none of these patients having a prior or subsequent clinical history of lymphoproliferative disease. Lymphoproliferations were predominantly human CD20+ B cells and had the immunophenotype expected for post-transplantation diffuse large B cell lymphoma with plasma cell features. All lymphoproliferations expressed Epstein-Barr-encoded RNAs (EBER). Analysis of immunoglobulin light chain gene rearrangements in three tumours where multiple tumour regions had resulted in lymphoproliferations suggested that each had independent clonal origins. Discussion: Overall, these data suggest that B cell clones with lymphoproliferative potential are present within primary NSCLC tumours, and that these are under continuous immune surveillance. Since these cells can be expanded following transplantation into NSG mice, our data highlight the value of quality control measures to identify lymphoproliferations within xenograft pipelines and support the incorporation of strategies to minimise lymphoproliferations during the early stages of xenograft establishment pipelines.

3.
Nature ; 614(7947): 334-342, 2023 02.
Article in English | MEDLINE | ID: mdl-36697826

ABSTRACT

The liver is bathed in bacterial products, including lipopolysaccharide transported from the intestinal portal vasculature, but maintains a state of tolerance that is exploited by persistent pathogens and tumours1-4. The cellular basis mediating this tolerance, yet allowing a switch to immunity or immunopathology, needs to be better understood for successful immunotherapy of liver diseases. Here we show that a variable proportion of CD8+ T cells compartmentalized in the human liver co-stain for CD14 and other prototypic myeloid membrane proteins and are enriched in close proximity to CD14high myeloid cells in hepatic zone 2. CD14+CD8+ T cells preferentially accumulate within the donor pool in liver allografts, among hepatic virus-specific and tumour-infiltrating responses, and in cirrhotic ascites. CD14+CD8+ T cells exhibit increased turnover, activation and constitutive immunomodulatory features with high homeostatic IL-10 and IL-2 production ex vivo, and enhanced antiviral/anti-tumour effector function after TCR engagement. This CD14+CD8+ T cell profile can be recapitulated by the acquisition of membrane proteins-including the lipopolysaccharide receptor complex-from mononuclear phagocytes, resulting in augmented tumour killing by TCR-redirected T cells in vitro. CD14+CD8+ T cells express integrins and chemokine receptors that favour interactions with the local stroma, which can promote their induction through CXCL12. Lipopolysaccharide can also increase the frequency of CD14+CD8+ T cells in vitro and in vivo, and skew their function towards the production of chemotactic and regenerative cytokines. Thus, bacterial products in the gut-liver axis and tissue stromal factors can tune liver immunity by driving myeloid instruction of CD8+ T cells with immunomodulatory ability.


Subject(s)
CD8-Positive T-Lymphocytes , Immune Tolerance , Lipopolysaccharide Receptors , Lipopolysaccharides , Liver , Myeloid Cells , Humans , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasms/immunology , Neoplasms/pathology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Immune Tolerance/drug effects , Immune Tolerance/immunology , Liver/drug effects , Liver/immunology , Liver/pathology , Liver/virology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Chemotaxis, Leukocyte , Bacteria/immunology , Intestines/immunology , Intestines/microbiology
4.
Nat Metab ; 4(4): 414-415, 2022 04.
Article in English | MEDLINE | ID: mdl-35361956

Subject(s)
Apoptosis , Inflammation , Humans
5.
Nat Aging ; 2(4): 280-281, 2022 04.
Article in English | MEDLINE | ID: mdl-37117749
6.
JHEP Rep ; 3(6): 100332, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34825153

ABSTRACT

BACKGROUND & AIMS: Infection is a major problem in advanced liver disease secondary to monocyte dysfunction. Elevated prostaglandin (PG)E2 is a mediator of monocyte dysfunction in cirrhosis; thus, we examined PGE2 signalling in outpatients with ascites and in patients hospitalised with acute decompensation to identify potential therapeutic targets aimed at improving monocyte dysfunction. METHODS: Using samples from 11 outpatients with ascites and 28 patients hospitalised with decompensated cirrhosis, we assayed plasma levels of PGE2 and lipopolysaccharide (LPS); performed quantitative real-time PCR on monocytes; and examined peripheral blood monocyte function. We performed western blotting and immunohistochemistry for PG biosynthetic machinery expression in liver tissue. Finally, we investigated the effect of PGE2 antagonists in whole blood using polychromatic flow cytometry and cytokine production. RESULTS: We show that hepatic production of PGE2 via the cyclo-oxygenase 1-microsomal PGE synthase 1 pathway, and circulating monocytes contributes to increased plasma PGE2 in decompensated cirrhosis. Transjugular intrahepatic sampling did not reveal whether hepatic or monocytic production was larger. Blood monocyte numbers increased, whereas individual monocyte function decreased as patients progressed from outpatients with ascites to patients hospitalised with acute decompensation, as assessed by Human Leukocyte Antigen (HLA)-DR isotype expression and tumour necrosis factor alpha and IL6 production. PGE2 mediated this dysfunction via its EP4 receptor. CONCLUSIONS: PGE2 mediates monocyte dysfunction in decompensated cirrhosis via its EP4 receptor and dysfunction was worse in hospitalised patients compared with outpatients with ascites. Our study identifies a potential drug target and therapeutic opportunity in these outpatients with ascites to reverse this process to prevent infection and hospital admission. LAY SUMMARY: Patients with decompensated cirrhosis (jaundice, fluid build-up, confusion, and vomiting blood) have high infection rates that lead to high mortality rates. A white blood cell subset, monocytes, function poorly in these patients, which is a key factor underlying their sensitivity to infection. We show that monocyte dysfunction in decompensated cirrhosis is mediated by a lipid hormone in the blood, prostaglandin E2, which is present at elevated levels, via its EP4 pathway. This dysfunction worsens when patients are hospitalised with complications of cirrhosis compared with those in the outpatients setting, which supports the EP4 pathway as a potential therapeutic target for patients to prevent infection and hospitalisation.

7.
Immunol Lett ; 230: 1-10, 2021 02.
Article in English | MEDLINE | ID: mdl-33309673

ABSTRACT

Ageing is a global burden. Increasing age is associated with increased incidence of infections and cancer and decreased vaccine efficacy. This increased morbidity observed with age, is believed to be due in part to a decline in adaptive immunity, termed immunosenescence. However not all aspects of immunity decrease with age as ageing presents with systemic low grade chronic inflammation, characterised by elevated concentrations of mediators such as IL-6, TNFα and C Reactive protein (CRP). Inflammation is a strong predictor of morbidity and mortality, and chronic inflammation is known to be detrimental to a functioning immune system. Although the source of the inflammation is much discussed, the key cells which are believed to facilitate the inflammageing phenomenon are the monocytes and macrophages. In this review we detail how macrophage and monocyte phenotype and function change with age. The impact of ageing on macrophages includes decreased phagocytosis and immune resolution, increased senescent-associated markers, increased inflammatory cytokine production, reduced autophagy, and a decrease in TLR expression. With monocytes there is an increase in circulating CD16+ monocytes, decreased type I IFN production, and decreased efferocytosis. In conclusion, we believe that monocytes and macrophages contribute to immunosenescence and inflammageing and as a result have an important role in defective immunity with age.


Subject(s)
Aging/physiology , Inflammation/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Humans , Immunosenescence
8.
Pharmacol Ther ; 221: 107745, 2021 05.
Article in English | MEDLINE | ID: mdl-33188794

ABSTRACT

While COVID-19, the disease driven by SARS-CoV-2 has ignited interest in the host immune response to this infection, it has also highlighted the lack of treatment options for the damaging inflammatory responses driven by pathogens that precipitate the acute respiratory distress syndrome (ARDS). With the global prevalence of SARS-CoV-2 and the likelihood of a second winter spike alongside seasonal flu, the need for effective and targeted anti-inflammatory agents is even more pressing. Here we discuss the aetiology of COVID-19 and the common signalling pathways driven by SARS-CoV-2, namely p38 MAP kinase. We highlight that p38 MAP kinase becomes elevated with increasing age, thereby driving many of the inflammatory pathways that precipitate death in old people with the added drawback of impairing vaccine efficacy in this susceptible age group. Finally, we review drugs available to inhibit p38 MAP kinase, their risks-versus-benefits as well as suggested dosing regimen to combat over-exuberant innate immune responses and potentially reverse vaccine inefficacy in older patients.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , COVID-19 Drug Treatment , MAP Kinase Signaling System/drug effects , Pneumonia/drug therapy , Protein Kinase Inhibitors/therapeutic use , Respiratory Distress Syndrome/drug therapy , Anti-Inflammatory Agents/pharmacology , COVID-19/epidemiology , COVID-19/immunology , Clinical Trials as Topic/methods , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , MAP Kinase Signaling System/physiology , Pneumonia/epidemiology , Pneumonia/immunology , Protein Kinase Inhibitors/pharmacology , Respiratory Distress Syndrome/epidemiology , Respiratory Distress Syndrome/immunology
9.
Aging Cell ; 19(12): e13272, 2020 12.
Article in English | MEDLINE | ID: mdl-33166035

ABSTRACT

The development of senescence in tissues of different organs and in the immune system are usually investigated independently of each other although during ageing, senescence in both cellular systems develop concurrently. Senescent T cells are highly inflammatory and secrete cytotoxic mediators and express natural killer cells receptors (NKR) that bypass their antigen specificity. Instead they recognize stress ligands that are induced by inflammation or infection of different cell types in tissues. In this article we discuss data on T cell senescence, how it is regulated and evidence for novel functional attributes of senescent T cells. We discuss an interactive loop between senescent T cells and senescent non-lymphoid cells and conclude that in situations of intense inflammation, senescent cells may damage healthy tissue. While the example for immunopathology induced by senescent cells that we highlight is cutaneous leishmaniasis, this situation of organ damage may apply to other infections, including COVID-19 and also rheumatoid arthritis, where ageing, inflammation and senescent cells are all part of the same equation.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cellular Senescence/physiology , Killer Cells, Natural/immunology , Leishmaniasis, Cutaneous/immunology , Receptors, Natural Killer Cell/immunology , Aging/immunology , Arthritis, Rheumatoid/immunology , COVID-19/immunology , Humans , Leishmania braziliensis/immunology , SARS-CoV-2/immunology
10.
Nat Immunol ; 21(6): 684-694, 2020 06.
Article in English | MEDLINE | ID: mdl-32231301

ABSTRACT

Aging is associated with remodeling of the immune system to enable the maintenance of life-long immunity. In the CD8+ T cell compartment, aging results in the expansion of highly differentiated cells that exhibit characteristics of cellular senescence. Here we found that CD27-CD28-CD8+ T cells lost the signaling activity of the T cell antigen receptor (TCR) and expressed a protein complex containing the agonistic natural killer (NK) receptor NKG2D and the NK adaptor molecule DAP12, which promoted cytotoxicity against cells that expressed NKG2D ligands. Immunoprecipitation and imaging cytometry indicated that the NKG2D-DAP12 complex was associated with sestrin 2. The genetic inhibition of sestrin 2 resulted in decreased expression of NKG2D and DAP12 and restored TCR signaling in senescent-like CD27-CD28-CD8+ T cells. Therefore, during aging, sestrins induce the reprogramming of non-proliferative senescent-like CD27-CD28-CD8+ T cells to acquire a broad-spectrum, innate-like killing activity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cellular Senescence/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cytotoxicity, Immunologic , Gene Expression Profiling , Humans , Membrane Proteins/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nuclear Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Natural Killer Cell/metabolism , Signal Transduction , Yellow Fever/genetics , Yellow Fever/immunology , Yellow Fever/metabolism , Yellow Fever/virology , Yellow fever virus/immunology
11.
Nat Immunol ; 21(6): 696, 2020 06.
Article in English | MEDLINE | ID: mdl-32303726

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Nat Immunol ; 21(6): 615-625, 2020 06.
Article in English | MEDLINE | ID: mdl-32251403

ABSTRACT

Increasing age alters innate immune-mediated responses; however, the mechanisms underpinning these changes in humans are not fully understood. Using a human dermal model of acute inflammation, we found that, although inflammatory onset is similar between young and elderly individuals, the resolution phase was substantially impaired in elderly individuals. This arose from a reduction in T cell immunoglobulin mucin receptor-4 (TIM-4), a phosphatidylserine receptor expressed on macrophages that enables the engulfment of apoptotic bodies, so-called efferocytosis. Reduced TIM-4 in elderly individuals was caused by an elevation in macrophage p38 mitogen-activated protein kinase (MAPK) activity. Administering an orally active p38 inhibitor to elderly individuals rescued TIM-4 expression, cleared apoptotic bodies and restored a macrophage resolution phenotype. Thus, inhibiting p38 in elderly individuals rejuvenated their resolution response to be more similar to that of younger people. This is the first resolution defect identified in humans that has been successfully reversed, thereby highlighting the tractability of targeting pro-resolution biology to treat diseases driven by chronic inflammation.


Subject(s)
Inflammation/etiology , Inflammation/metabolism , Phagocytosis/immunology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Age Factors , Aged , Animals , Apoptosis , Blister/immunology , Blister/metabolism , Blister/pathology , Cantharidin , Gene Expression , Humans , Immunity, Innate , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Receptors, Cell Surface/metabolism , Signal Transduction
13.
JCI Insight ; 3(6)2018 03 22.
Article in English | MEDLINE | ID: mdl-29563331

ABSTRACT

While the treatment of inflammatory disorders is generally based on inhibiting factors that drive onset of inflammation, these therapies can compromise healing (NSAIDs) or dampen immunity against infections (biologics). In search of new antiinflammatories, efforts have focused on harnessing endogenous pathways that drive resolution of inflammation for therapeutic gain. Identification of specialized pro-resolving mediators (SPMs) (lipoxins, resolvins, protectins, maresins) as effector molecules of resolution has shown promise in this regard. However, their action on inflammatory resolution in humans is unknown. Here, we demonstrate using a model of UV-killed Escherichia coli-triggered skin inflammation that SPMs are biosynthesized at the local site at the start of resolution, coinciding with the expression of receptors that transduce their actions. These include receptors for lipoxin A4 (ALX/FPR2), resolvin E1 (ChemR23), resolvin D2 (GPR18), and resolvin D1 (GPR32) that were differentially expressed on the endothelium and infiltrating leukocytes. Administering SPMs into the inflamed site 4 hours after bacterial injection caused a reduction in PMN numbers over the ensuing 6 hours, the phase of active resolution in this model. These results indicate that in humans, the appearance of SPMs and their receptors is associated with the beginning of inflammatory resolution and that their therapeutic supplementation enhanced the resolution response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Escherichia coli/immunology , Inflammation/immunology , Inflammation/metabolism , Skin/immunology , Skin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Blister/immunology , Blister/metabolism , Chemokines/metabolism , Cytokines/metabolism , Docosahexaenoic Acids/pharmacology , Eicosanoids/immunology , Eicosanoids/pharmacology , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Escherichia coli/radiation effects , Humans , Inflammation/drug therapy , Leukocytes/immunology , Leukocytes/metabolism , Lipoxins/pharmacology , Male , Middle Aged , Neutrophils/drug effects , Receptors, Chemokine/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, G-Protein-Coupled , Receptors, Lipoxin/metabolism , Skin/drug effects , Skin/pathology , Volunteers , Young Adult
14.
Cell Rep ; 20(13): 3162-3175, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28954232

ABSTRACT

Acute inflammation is characterized by granulocyte infiltration followed by efferocytosing mononuclear phagocytes, which pave the way for inflammatory resolution. Until now, it was believed that resolution then leads back to homeostasis, the physiological state tissues experience before inflammation occurred. However, we discovered that resolution triggered a prolonged phase of immune suppression mediated by prostanoids. Specifically, once inflammation was switched off, natural killer cells, secreting interferon γ (IFNγ), infiltrated the post-inflamed site. IFNγ upregulated microsomal prostaglandin E synthase-1 (mPGES-1) alongside cyclo-oxygenase (COX-1) within macrophage populations, resulting in sustained prostaglandin (PG)E2 biosynthesis. Whereas PGE2 suppressed local innate immunity to bacterial infection, it also inhibited lymphocyte function and generated myeloid-derived suppressor cells, the net effect of which was impaired uptake/presentation of exogenous antigens. Therefore, we have defined a sequence of post-resolution events that dampens the propensity to develop autoimmune responses to endogenous antigens at the cost of local tissue infection.


Subject(s)
Cyclooxygenase 1/immunology , Dinoprostone/immunology , Inflammation/immunology , Membrane Proteins/immunology , Prostaglandin-E Synthases/immunology , Animals , Inflammation/enzymology , Interferon-gamma/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL
15.
J Pathol Clin Res ; 2(3): 154-65, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27499924

ABSTRACT

Whilst numerous studies investigating the aetiology of inflammatory diseases have been performed in rodents, the applicability of these data to human pathophysiology is frequently debated. Regardless of the strengths and weaknesses of rodent models in biomedical research, there is a need to develop models of experimental inflammation in humans. Here, we describe a self-resolving acute inflammatory response triggered by the intradermal injection of UV-killed Escherichia coli into the forearm of healthy volunteers. Cells and exudates were harvested from onset to resolution by applying negative pressure over the inflamed site. Onset was characterized by high blood flow, neutrophilia and peak levels of pro-inflammatory cytokines, whilst resolution showed a decline in blood blow, reduction in neutrophils, increase in monocytes/macrophages and waning of classic pro-inflammatory cytokine levels. An anti-inflammatory effect, defined as suppression of onset phase events, was demonstrated by administering naproxen, a conventional non-steroidal anti-inflammatory drug. In summary, this model of resolving acute inflammation is minimally invasive, highly tractable and allows simultaneous investigation of the vascular response, cellular trafficking and chemical mediator profile of onset and resolution phases of acute inflammation in humans. It can serve as a translational platform to provide mechanistic insights and to test the clinical efficacy of novel anti-inflammatory and pro-resolving drugs, and also as a tool in patients to explore inherent defects in resolution pathways.

16.
J Vis Exp ; (111)2016 05 16.
Article in English | MEDLINE | ID: mdl-27213711

ABSTRACT

Activation of inflammatory pathways represents a central mechanism in multiple disease states both acute and chronic. Triggered via either pathogen or tissue damage-associated molecular motifs, common biochemical pathways lead to conserved yet variable physiological and immunological alterations. Dissection and delineation of the determinants and mechanisms underlying phenotypic variance in response is expected to yield novel therapeutic advances. Intravenous (IV) administration of endotoxin (gram-negative bacterial lipopolysaccharide), a specific Toll-like receptor 4 agonist, represents an in vivo model of systemic inflammation in man. National Institutes for Health Clinical Center Reference Endotoxin (CCRE, Escherichia coli O:113:H10:K negative) is employed to reliably and reproducibly generate vascular, hematological, endocrine, immunological and organ-specific functional effects that parallel, to varying degrees, those seen in the early stages of pathological states. Alteration of dose (0.06 - 4 ng/kg) and time-scale of exposure (bolus vs. infusion) allows replication of either acute or chronic inflammation and a range of severity to be elicited, with higher doses (2 - 4 ng/kg) frequently being used to create a 'sepsis-like' state. Established and novel medicinal compounds may additionally be administered prior to or post endotoxin exposure to appreciate their effect on the inflammatory cascade. Despite limitations in scope and generalizability, human IV endotoxin challenge offers a unique platform to gain mechanistic insights into inducible physiological responses and inflammatory pathways. Rationally employed it may aid translation of this knowledge into therapeutic innovations.


Subject(s)
Endotoxins/administration & dosage , Escherichia coli , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Inflammation/immunology , Adult , Blood Pressure/drug effects , Body Temperature/drug effects , Cytokines/blood , Heart Rate/drug effects , Humans , Inflammation/pathology , Infusions, Intravenous , Leukocyte Count , Lipopolysaccharides/administration & dosage , Male , Respiratory Rate/drug effects , Young Adult
17.
Proc Natl Acad Sci U S A ; 113(23): E3240-9, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27226306

ABSTRACT

Resolution of inflammation has emerged as an active process in immunobiology, with cells of the mononuclear phagocyte system being critical in mediating efferocytosis and wound debridement and bridging the gap between innate and adaptive immunity. Here we investigated the roles of cytochrome P450 (CYP)-derived epoxy-oxylipins in a well-characterized model of sterile resolving peritonitis in the mouse. Epoxy-oxylipins were produced in a biphasic manner during the peaks of acute (4 h) and resolution phases (24-48 h) of the response. The epoxygenase inhibitor SKF525A (epoxI) given at 24 h selectively inhibited arachidonic acid- and linoleic acid-derived CYP450-epoxy-oxlipins and resulted in a dramatic influx in monocytes. The epoxI-recruited monocytes were strongly GR1(+), Ly6c(hi), CCR2(hi), CCL2(hi), and CX3CR1(lo) In addition, expression of F4/80 and the recruitment of T cells, B cells, and dendritic cells were suppressed. sEH (Ephx2)(-/-) mice, which have elevated epoxy-oxylipins, demonstrated opposing effects to epoxI-treated mice: reduced Ly6c(hi) monocytes and elevated F4/80(hi) macrophages and B, T, and dendritic cells. Ly6c(hi) and Ly6c(lo) monocytes, resident macrophages, and recruited dendritic cells all showed a dramatic change in their resolution signature following in vivo epoxI treatment. Markers of macrophage differentiation CD11b, MerTK, and CD103 were reduced, and monocyte-derived macrophages and resident macrophages ex vivo showed greatly impaired phagocytosis of zymosan and efferocytosis of apoptotic thymocytes following epoxI treatment. These findings demonstrate that epoxy-oxylipins have a critical role in monocyte lineage recruitment and activity to promote inflammatory resolution and represent a previously unidentified internal regulatory system governing the establishment of adaptive immunity.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Monocytes/metabolism , Oxylipins/metabolism , Peritonitis/metabolism , Animals , Epoxide Hydrolases/genetics , Epoxide Hydrolases/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis
18.
BMJ Open ; 6(1): e010132, 2016 Jan 25.
Article in English | MEDLINE | ID: mdl-26810999

ABSTRACT

INTRODUCTION: Circulating prostaglandin E2 levels are elevated in acutely decompensated cirrhosis and have been shown to contribute to immune suppression. Albumin binds and inactivates this hormone. Human albumin solution could thus be repurposed as an immune restorative drug in these patients.This feasibility study aims to determine whether it is possible and safe to restore serum albumin to >30 g/L and maintain it at this level in patients admitted with acute decompensated cirrhosis using repeated 20% human albumin infusions according to daily serum albumin levels. METHODS AND ANALYSIS: Albumin To prevenT Infection in chronic liveR failurE (ATTIRE) stage 1 is a multicentre, open label dose feasibility trial. Patients with acutely decompensated cirrhosis admitted to hospital with a serum albumin of <30 g/L are eligible, subject to exclusion criteria. Daily intravenous human albumin solution will be infused, according to serum albumin levels, for up to 14 days or discharge in all patients. The primary end point is daily serum albumin levels for the duration of the treatment period and the secondary end point is plasma-induced macrophage dysfunction. The trial will recruit 80 patients. Outcomes will be used to assist with study design for an 866 patient randomised controlled trial at more than 30 sites across the UK. ETHICS AND DISSEMINATION: Research ethics approval was given by the London-Brent research ethics committee (ref: 15/LO/0104). The clinical trials authorisation was issued by the medicines and healthcare products regulatory agency (ref: 20363/0350/001-0001). RESULTS: Will be disseminated through peer reviewed journals and international conferences. Recruitment of the first participant occurred on 26/05/2015. TRIAL REGISTRATION NUMBER: The trial is registered with the European Medicines Agency (EudraCT 2014-002300-24) and has been adopted by the NIHR (ISRCTN 14174793). This manuscript refers to V.4.0 of the protocol; Pre-results.


Subject(s)
Albumins/administration & dosage , Cross Infection/prevention & control , End Stage Liver Disease/complications , Liver Cirrhosis/complications , Adult , Aged , Clinical Protocols , Cytokines/metabolism , Feasibility Studies , Humans , Infusions, Intravenous , Macrophages/immunology , Middle Aged , Serum Albumin/metabolism , Treatment Outcome , Young Adult
19.
Semin Immunol ; 27(3): 161-8, 2015 May.
Article in English | MEDLINE | ID: mdl-26037968

ABSTRACT

The goal of treating chronic inflammatory diseases must be to inhibit persistent inflammation and restore tissue function. To achieve this we need to improve our understanding of the pathways that drive inflammation as well as those that bring about its resolution. In particular, resolution of inflammation is driven by a complex set of mediators that regulate cellular events required to clear inflammatory cells from sites of injury or infection and restore homeostasis. Indeed, it may be argued that dysfunctional resolution may underpin the aetiology of some chronic inflammatory disease and that a novel goal in treating such diseases is to develop drugs based on the mode of endogenous pro-resolution factors in order to drive on-going inflammation down a pro-resolution pathway. And while we are improving our understanding of the resolution of acute and chronic inflammation, much remains to be discovered. Here we will discuss the key endogenous checkpoints necessary for mounting an effective yet limited inflammatory response and the crucial biochemical pathways necessary to prevent its persistence and trigger its resolution. In doing so, we will provide an update on what is known about resolution of acute inflammation, in particular its link with adaptive immunity.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Inflammation Mediators/immunology , Inflammation/immunology , Macrophages/immunology , Animals , Anti-Inflammatory Agents/therapeutic use , Chronic Disease , Cytokines/immunology , Homeostasis/immunology , Humans , Inflammation/drug therapy , Mice , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...