Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nat Immunol ; 14(10): 986-95, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24048120

ABSTRACT

Tissue-resident macrophages are a heterogeneous population of immune cells that fulfill tissue-specific and niche-specific functions. These range from dedicated homeostatic functions, such as clearance of cellular debris and iron processing, to central roles in tissue immune surveillance, response to infection and the resolution of inflammation. Recent studies highlight marked heterogeneity in the origins of tissue macrophages that arise from hematopoietic versus self-renewing embryo-derived populations. We discuss the tissue niche-specific factors that dictate cell phenotype, the definition of which will allow new strategies to promote the restoration of tissue homeostasis. Understanding the mechanisms that dictate tissue macrophage heterogeneity should explain why simplified models of macrophage activation do not explain the extent of heterogeneity seen in vivo.


Subject(s)
Macrophages/immunology , Animals , Humans , Immunity , Immunologic Surveillance , Inflammation/immunology , Macrophage Activation/immunology , Macrophages/cytology , Macrophages/metabolism , Organ Specificity/immunology , Wound Healing/immunology
2.
EMBO J ; 39(14): e103454, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32484988

ABSTRACT

The alarm cytokine interleukin-1ß (IL-1ß) is a potent activator of the inflammatory cascade following pathogen recognition. IL-1ß production typically requires two signals: first, priming by recognition of pathogen-associated molecular patterns leads to the production of immature pro-IL-1ß; subsequently, inflammasome activation by a secondary signal allows cleavage and maturation of IL-1ß from its pro-form. However, despite the important role of IL-1ß in controlling local and systemic inflammation, its overall regulation is still not fully understood. Here we demonstrate that peritoneal tissue-resident macrophages use an active inhibitory pathway, to suppress IL-1ß processing, which can otherwise occur in the absence of a second signal. Programming by the transcription factor Gata6 controls the expression of prostacyclin synthase, which is required for prostacyclin production after lipopolysaccharide stimulation and optimal induction of IL-10. In the absence of secondary signal, IL-10 potently inhibits IL-1ß processing, providing a previously unrecognized control of IL-1ß in tissue-resident macrophages.


Subject(s)
Epoprostenol/immunology , Interleukin-10/immunology , Interleukin-1beta/immunology , Macrophages, Peritoneal/immunology , Animals , Epoprostenol/genetics , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Interleukin-1beta/genetics , Macrophages, Peritoneal/pathology , Mice , Mice, Transgenic
3.
PLoS Pathog ; 15(6): e1007850, 2019 06.
Article in English | MEDLINE | ID: mdl-31242262

ABSTRACT

Invasive candidiasis, mainly caused by Candida albicans, is a serious healthcare problem with high mortality rates, particularly in immunocompromised patients. Innate immune cells express pathogen recognition receptors (PRRs) including C-type lectin-like receptors (CLRs) that bind C. albicans to initiate an immune response. Multiple CLRs including Dectin-1, Dectin-2 and Mincle have been proposed individually to contribute to the immune response to C. albicans. However how these receptors collaborate to clear a fungal infection is unknown. Herein, we used novel multi-CLR knockout (KO) mice to decipher the individual, collaborative and collective roles of Dectin-1, Dectin-2 and Mincle during systemic C. albicans infection. These studies revealed an unappreciated and profound role for CLR co-operation in anti-fungal immunity. The protective effect of multiple CLRs was markedly greater than any single receptor, and was mediated through inflammatory monocytes via recognition and phagocytosis of C. albicans, and production of C. albicans-induced cytokines and chemokines. These CLRs were dispensable for mediating similar responses from neutrophils, likely due to lower expression of these CLRs on neutrophils compared to inflammatory monocytes. Concurrent deletion of Dectin-1 and Dectin-2, or all three CLRs, resulted in dramatically increased susceptibility to systemic C. albicans infection compared to mice lacking a single CLR. Multi-CLR KO mice were unable to control fungal growth due to an inadequate early inflammatory monocyte-mediated response. In response to excessive fungal growth, the multi-CLR KO mice mounted a hyper-inflammatory response, likely leading to multiple organ failure. Thus, these data reveal a critical role for CLR co-operation in the effective control of C. albicans and maintenance of organ function during infection.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Lectins, C-Type/immunology , Membrane Proteins/immunology , Monocytes/immunology , Animals , Candidiasis/genetics , Chemokines/genetics , Chemokines/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lectins, C-Type/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Monocytes/pathology , Neutrophils/pathology
4.
Eur J Immunol ; 46(9): 2222-32, 2016 09.
Article in English | MEDLINE | ID: mdl-27378515

ABSTRACT

The inflammatory activation and recruitment of defined myeloid populations is essential for controlling the bridge between innate and adaptive immunity and shaping the immune response to microbial challenge. However, these cells exhibit significant functional heterogeneity and the inflammatory signals that differentially influence their effector characteristics are poorly characterized. In this study, we defined the phenotype of discrete subsets of effective antigen-presenting cells (APCs) in the peritoneal cavity during peritonitis. When the functional properties of these cells were compared to inflammatory monocyte-derived macrophages we noted differential responses to the immune-modulatory cytokine IL-10. In contrast to the suppressive actions of IL-10 on inflammatory macrophages, the recruitment of APCs was relatively refractory and we found no evidence for selective inhibition of APC differentiation. This differential response of myeloid cell subsets to IL-10 may thus have limited impact on development of potentially tissue-damaging adaptive immune responses, while restricting the magnitude of the inflammatory response. These findings may have clinical relevance in the context of peritoneal dialysis patients, where recurrent infections are associated with immune-mediated membrane dysfunction, treatment failure, and increased morbidity.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/metabolism , Macrophages/immunology , Macrophages/metabolism , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/pathology , Biomarkers , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Immunomodulation , Immunophenotyping , Inflammation/pathology , Interleukin-10/genetics , Macrophages/pathology , Mice , Mice, Knockout , Peritonitis/immunology , Peritonitis/metabolism , Peritonitis/pathology , Phenotype , Receptors, CCR2/metabolism
5.
Immunology ; 144(4): 541-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25684236

ABSTRACT

Macrophages have been at the heart of immune research for over a century and are an integral component of innate immunity. Macrophages are often viewed as terminally differentiated monocytic phagocytes. They infiltrate tissues during inflammation, and form polarized populations that perform pro-inflammatory or anti-inflammatory functions. Tissue-resident macrophages were regarded as differentiated monocytes, which seed the tissues to perform immune sentinel and homeostatic functions. However, tissue-resident macrophages are not a homogeneous population, but are in fact a grouping of cells with similar functions and phenotypes. In the last decade, it has been revealed that many of these cells are not terminally differentiated and, in most cases, are not derived from haematopoiesis in the adult. Recent research has highlighted that tissue-resident macrophages cannot be grouped into simple polarized categories, especially in vivo, when they are exposed to complex signalling events. It has now been demonstrated that the tissue environment itself is a major controller of macrophage phenotype, and can influence the expression of many genes regardless of origin. This is consistent with the concept that cells within different tissues have diverse responses in inflammation. There is still a mountain to climb in the field, as it evolves to encompass not only tissue-resident macrophage diversity, but also categorization of specific tissue environments and the plasticity of macrophages themselves. This knowledge provides a new perspective on therapeutic strategies, as macrophage subsets can potentially be manipulated to control the inflammatory environment in a tissue-specific manner.


Subject(s)
Immunity, Innate , Macrophages/immunology , Animals , Gene Expression Regulation , Homeostasis , Humans , Inflammation/immunology , Inflammation Mediators/immunology , Macrophage Activation , Macrophages/classification , Macrophages/metabolism , Phenotype , Signal Transduction , Transcription Factors/immunology
6.
Am J Pathol ; 184(4): 996-1009, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24508230

ABSTRACT

Aristolochic acid nephropathy is characterized by rapidly progressive tubulointerstitial nephritis culminating in end-stage renal failure and urothelial malignancy. Profibrotic effects of aristolochic acid are linked to growth arrest of proximal tubular epithelial cells; however, the underlying mechanisms are largely undetermined. miRNAs are small, endogenous, post-transcriptional regulators of gene expression implicated in numerous physiological and pathological processes. In the present study, we characterized the mechanism of aristolochic acid-induced cell cycle arrest and its regulation by miRNAs. Incubation with aristolochic acid led to profound G2/M arrest in proximal tubular epithelial cells via p53-mediated inactivation of the maturation-promoting complex, CDK1/cyclin-B1. Analysis of miRNA expression identified up-regulation of miRNAs, including miR-192, miR-194, miR-450a, and miR-542-3p. The stable overexpression of miR-192 recapitulated G2/M arrest via repression of the E3 ubiquitin ligase, murine double-minute 2, a negative regulator of p53. p53-induced transcription of p21(cip1) and growth arrest and DNA damage 45 and resulted in the inactivation and dissociation of the maturation-promoting complex. These data demonstrate a core role for miR-192 in mediating proximal tubular epithelial cell G2/M arrest after toxic injury by aristolochic acid. Because numerous studies have linked such growth arrest to fibrosis after proximal tubular epithelial cell injury, this mechanism may have widespread relevance to recovery/nonrecovery after acute kidney injury.


Subject(s)
Aristolochic Acids/poisoning , G2 Phase Cell Cycle Checkpoints/genetics , Kidney Diseases/pathology , M Phase Cell Cycle Checkpoints/genetics , MicroRNAs/genetics , Blotting, Western , Cells, Cultured , Epithelial Cells/pathology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Kidney Diseases/chemically induced , Kidney Tubules, Proximal/pathology , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
7.
J Vis Exp ; (204)2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38436380

ABSTRACT

Peritoneal tissue-resident macrophages have broad functions in the maintenance of homeostasis and are involved in pathologies within local and neighboring tissues. Their functions are dictated by microenvironmental cues; thus, it is essential to investigate their behavior in an in vivo physiological niche. Currently, specific peritoneal macrophage-targeting methodologies employ whole-mouse transgenic models. Here, a protocol for effective in vivo modulation of mRNA and small RNA species (e.g., microRNA) expression in peritoneal macrophages using lentivirus particles is described. Lentivirus preparations were made in HEK293T cells and purified on a single sucrose layer. In vivo validation of lentivirus effectivity following intraperitoneal injection revealed predominant infection of macrophages restricted to local tissue. Targeting of peritoneal macrophages was successful during homeostasis and thioglycolate-induced peritonitis. The limitations of the protocol, including low-level inflammation induced by intraperitoneal delivery of lentivirus and time restrictions for potential experiments, are discussed. Overall, this study presents a quick and accessible protocol for the rapid assessment of gene function in peritoneal macrophages in vivo.


Subject(s)
MicroRNAs , Humans , Animals , Mice , MicroRNAs/genetics , Peritoneal Cavity , Lentivirus/genetics , HEK293 Cells , Macrophages , Disease Models, Animal
8.
iScience ; 27(5): 109779, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38736550

ABSTRACT

Metabolic heterogeneity is a determinant of immune cell function. The normal physiological metabolic reprogramming of pregnancy that ensures the fuel requirements of mother and baby are met, might also underpin changes in immunity that occur with pregnancy and manifest as altered responses to pathogens and changes to autoimmune disease symptoms. Using peripheral blood from pregnant women at term, we reveal that monocytes lose M2-like and gain M1-like properties accompanied by reductions in mitochondrial mass, maximal respiration, and cardiolipin content in pregnancy; glycolysis is unperturbed. We establish that muramyl dipeptide (MDP)-stimulated cytokine production relies on oxidative metabolism, then show in pregnancy reduced cytokine production in response to MDP but not LPS. Overall, mitochondrially centered metabolic capabilities of late gestation monocytes are down-regulated revealing natural plasticity in monocyte phenotype and function that could reveal targets for improving pregnancy outcomes but also yield alternative therapeutic approaches to diverse metabolic and/or immune-mediated diseases beyond pregnancy.

9.
BMJ Open ; 13(3): e067002, 2023 03 27.
Article in English | MEDLINE | ID: mdl-36972964

ABSTRACT

INTRODUCTION: Early recognition and appropriate management of paediatric sepsis are known to improve outcomes. A previous system's biology investigation of the systemic immune response in neonates to sepsis identified immune and metabolic markers that showed high accuracy for detecting bacterial infection. Further gene expression markers have also been reported previously in the paediatric age group for discriminating sepsis from control cases. More recently, specific gene signatures were identified to discriminate between COVID-19 and its associated inflammatory sequelae. Through the current prospective cohort study, we aim to evaluate immune and metabolic blood markers which discriminate between sepses (including COVID-19) from other acute illnesses in critically unwell children and young persons, up to 18 years of age. METHODS AND ANALYSIS: We describe a prospective cohort study for comparing the immune and metabolic whole-blood markers in patients with sepsis, COVID-19 and other illnesses. Clinical phenotyping and blood culture test results will provide a reference standard to evaluate the performance of blood markers from the research sample analysis. Serial sampling of whole blood (50 µL each) will be collected from children admitted to intensive care and with an acute illness to follow time dependent changes in biomarkers. An integrated lipidomics and RNASeq transcriptomics analyses will be conducted to evaluate immune-metabolic networks that discriminate sepsis and COVID-19 from other acute illnesses. This study received approval for deferred consent. ETHICS AND DISSEMINATION: The study has received research ethics committee approval from the Yorkshire and Humber Leeds West Research Ethics Committee 2 (reference 20/YH/0214; IRAS reference 250612). Submission of study results for publication will involve making available all anonymised primary and processed data on public repository sites. TRIAL REGISTRATION NUMBER: NCT04904523.


Subject(s)
COVID-19 , Sepsis , Adolescent , Child , Humans , Infant, Newborn , Acute Disease , COVID-19/diagnosis , Prospective Studies , SARS-CoV-2 , Sepsis/diagnosis
10.
Eur J Immunol ; 41(9): 2503-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21952806

ABSTRACT

In this Viewpoint, we concentrate on the aspects of macrophage biology that we believe are fundamental for an appropriate contextual understanding of macrophage function during acute inflammation. These are the different origins of macrophage populations (and the implications of this for the renewal of these populations in the adult); and the impact of specific homeostatic or disease-associated microenvironments upon cellular heterogeneity, activation and effector functions.


Subject(s)
Macrophages/immunology , Acute Disease , Adult , Animals , Cell Differentiation/immunology , Homeostasis/immunology , Humans , Inflammation , Macrophage Activation , Regeneration/immunology
11.
Eur J Immunol ; 41(8): 2155-64, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21710478

ABSTRACT

Macrophage (MØ) biology is routinely modelled in the peritoneal cavity, a vascular tissue readily infiltrated by leukocytes during inflammation. After several decades of study, no consensus has emerged regarding the importance of in situ proliferation versus peripheral monocyte recruitment for the maintenance of tissue resident MØs. By applying specific measures of mitosis, we have monitored tissue MØ proliferation during newborn development, adulthood and acute resolving inflammation in young adult mice. Despite the vascular nature of the tissue and ease of peripheral leukocyte entry, tissue MØs in the newborn increase in number by local proliferation. On the contrary, in the adult, tissue MØ proliferation is considerably reduced and most likely provides homeostatic control of cell numbers. Importantly, during an acute inflammatory response, when substantial numbers of inflammatory MØs are recruited from the circulation, tissue-resident MØs survive and then undergo a transient and intense proliferative burst in situ to repopulate the tissue. Our data indicate that local proliferation is a general mechanism for the self-sufficient renewal of tissue MØs during development and acute inflammation and not one restricted to non-vascular tissues, which has implications for the therapeutic modulation of MØ activity during the resolution of inflammation.


Subject(s)
Cell Proliferation , Homeostasis/immunology , Macrophages/immunology , Peritonitis/immunology , Acute Disease , Animals , Cell Cycle/immunology , Cell Survival/immunology , Cells, Cultured , DNA/immunology , DNA/metabolism , Female , Flow Cytometry , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Peritoneal Cavity/pathology , Peritonitis/metabolism , Peritonitis/pathology , Time Factors
12.
Eur J Immunol ; 41(2): 356-65, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268006

ABSTRACT

We have examined the potential to generate bona fide macrophages (MØ) from conditionally immortalised murine bone marrow precursors. MØ can be derived from Hoxb8 conditionally immortalised macrophage precursor cell lines (MØP) using either M-CSF or GM-CSF. When differentiated in GM-CSF (GM-MØP) the resultant cells resemble GM-CSF bone marrow-derived dendritic cells (BMDC) in morphological phenotype, antigen phenotype and functional responses to microbial stimuli. In spite of this high similarity between the two cell types and the ability of GM-MØP to effectively present antigen to a T-cell hybridoma, these cells are comparatively poor at priming the expansion of IFN-γ responses from naïve CD4(+) T cells. The generation of MØP from transgenic or genetically aberrant mice provides an excellent opportunity to study the inflammatory role of GM-MØP, and reduces the need for mouse colonies in many studies. Hence differentiation of conditionally immortalised MØPs in GM-CSF represents a unique in vitro model of inflammatory monocyte-like cells, with important differences from bone marrow-derived dendritic cells, which will facilitate functional studies relating to the many 'sub-phenotypes' of inflammatory monocytes.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Homeodomain Proteins/genetics , Macrophages/cytology , Monocyte-Macrophage Precursor Cells/cytology , Animals , Antigen Presentation/immunology , Antigens, Surface/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Line, Transformed , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Lectins, C-Type , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Monocyte-Macrophage Precursor Cells/drug effects , Monocyte-Macrophage Precursor Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nitric Oxide/metabolism , Ovalbumin/immunology , Transduction, Genetic , Zymosan/pharmacology , beta-Glucans/pharmacology
13.
EBioMedicine ; 86: 104337, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36470829

ABSTRACT

Physiological shifts during pregnancy predispose women to a higher risk of developing sepsis resulting from a maladapted host-response to infection. Insightful studies have delineated subtle point-changes to the immune system during pregnancy. Here, we present an overlay of these point-changes, asking what changes and when, at a physiological, cellular, and molecular systems-level in the context of sepsis. We identify distinct immune phases in pregnancy delineated by placental hormone-driven changes in homeostasis setpoints of the immune and metabolic systems that subtly mirrors changes observed in sepsis. We propose that pregnancy immune-metabolic setpoint changes impact feedback thresholds that increase risk for a maladapted host-response to infection and thus act as a stepping-stone to sepsis. Defining maternal immune-metabolic setpoint changes is not only vital for tailoring the right diagnostic tools for early management of maternal sepsis but will facilitate an unravelling of the pathophysiological pathways that predispose an individual to sepsis.


Subject(s)
Pregnancy Complications, Infectious , Sepsis , Humans , Pregnancy , Female , Placenta/metabolism , Sepsis/diagnosis , Sepsis/etiology , Sepsis/metabolism , Adaptation, Physiological , Homeostasis
14.
Nat Commun ; 13(1): 139, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013270

ABSTRACT

Oxylipins are potent biological mediators requiring strict control, but how they are removed en masse during infection and inflammation is unknown. Here we show that lipopolysaccharide (LPS) dynamically enhances oxylipin removal via mitochondrial ß-oxidation. Specifically, genetic or pharmacological targeting of carnitine palmitoyl transferase 1 (CPT1), a mitochondrial importer of fatty acids, reveal that many oxylipins are removed by this protein during inflammation in vitro and in vivo. Using stable isotope-tracing lipidomics, we find secretion-reuptake recycling for 12-HETE and its intermediate metabolites. Meanwhile, oxylipin ß-oxidation is uncoupled from oxidative phosphorylation, thus not contributing to energy generation. Testing for genetic control checkpoints, transcriptional interrogation of human neonatal sepsis finds upregulation of many genes involved in mitochondrial removal of long-chain fatty acyls, such as ACSL1,3,4, ACADVL, CPT1B, CPT2 and HADHB. Also, ACSL1/Acsl1 upregulation is consistently observed following the treatment of human/murine macrophages with LPS and IFN-γ. Last, dampening oxylipin levels by ß-oxidation is suggested to impact on their regulation of leukocyte functions. In summary, we propose mitochondrial ß-oxidation as a regulatory metabolic checkpoint for oxylipins during inflammation.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Lipid Metabolism/genetics , Mitochondria/drug effects , Oxylipins/metabolism , Peritonitis/genetics , Sepsis/genetics , Acyl-CoA Dehydrogenase, Long-Chain/blood , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Animals , Carnitine O-Palmitoyltransferase/blood , Carnitine O-Palmitoyltransferase/genetics , Coenzyme A Ligases/blood , Coenzyme A Ligases/genetics , Female , Gene Expression Regulation , Humans , Infant, Newborn , Interferon-gamma/pharmacology , Lipidomics/methods , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/blood , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Oxidation-Reduction , Peritonitis/blood , Peritonitis/chemically induced , Peritonitis/pathology , RAW 264.7 Cells , Sepsis/blood , Sepsis/pathology
15.
BMJ Open ; 12(9): e066382, 2022 09 17.
Article in English | MEDLINE | ID: mdl-36115679

ABSTRACT

INTRODUCTION: Maternal sepsis remains a leading cause of death in pregnancy. Physiological adaptations to pregnancy obscure early signs of sepsis and can result in delays in recognition and treatment. Identifying biomarkers that can reliably diagnose sepsis will reduce morbidity and mortality and antibiotic overuse. We have previously identified an immune-metabolic biomarker network comprising three pathways with a >99% accuracy for detecting bacterial neonatal sepsis. In this prospective study, we will describe physiological parameters and novel biomarkers in two cohorts-healthy pregnant women and pregnant women with suspected sepsis-with the aim of mapping pathophysiological drivers and evaluating predictive biomarkers for diagnosing maternal sepsis. METHODS AND ANALYSIS: Women aged over 18 with an ultrasound-confirmed pregnancy will be recruited to a pilot and two main study cohorts. The pilot will involve blood sample collection from 30 pregnant women undergoing an elective caesarean section. Cohort A will follow 100 healthy pregnant women throughout their pregnancy journey, with collection of blood samples from participants at routine time points in their pregnancy: week 12 'booking', week 28 and during labour. Cohort B will follow 100 pregnant women who present with suspected sepsis in pregnancy or labour and will have at least two blood samples taken during their care pathway. Study blood samples will be collected during routine clinical blood sampling. Detailed medical history and physiological parameters at the time of blood sampling will be recorded, along with the results of routine biochemical tests, including C reactive protein, lactate and white blood cell count. In addition, study blood samples will be processed and analysed for transcriptomic, lipidomic and metabolomic analyses and both qualitative and functional immunophenotyping. ETHICS AND DISSEMINATION: Ethical approval has been obtained from the Wales Research Ethics Committee 2 (SPON1752-19, 30 October 2019). TRIAL REGISTRATION NUMBER: NCT05023954.


Subject(s)
Pre-Eclampsia , Pregnancy Complications, Infectious , Sepsis , Adolescent , Adult , Anti-Bacterial Agents , Biomarkers , C-Reactive Protein , Cesarean Section , Cohort Studies , Female , Humans , Infant, Newborn , Lactates , Observational Studies as Topic , Pregnancy , Pregnancy Complications, Infectious/diagnosis , Pregnant Women , Prospective Studies
16.
BMJ Open ; 11(12): e050100, 2021 12 30.
Article in English | MEDLINE | ID: mdl-37010923

ABSTRACT

INTRODUCTION: Diagnosing neonatal sepsis is heavily dependent on clinical phenotyping as culture-positive body fluid has poor sensitivity, and existing blood biomarkers have poor specificity.A combination of machine learning, statistical and deep pathway biology analyses led to the identification of a tripartite panel of biologically connected immune and metabolic markers that showed greater than 99% accuracy for detecting bacterial infection with 100% sensitivity. The cohort study described here is designed as a large-scale clinical validation of this previous work. METHODS AND ANALYSIS: This multicentre observational study will prospectively recruit a total of 1445 newborn infants (all gestations)-1084 with suspected early-or late-onset sepsis, and 361 controls-over 4 years. A small volume of whole blood will be collected from infants with suspected sepsis at the time of presentation. This sample will be used for integrated transcriptomic, lipidomic and targeted proteomics profiling. In addition, a subset of samples will be subjected to cellular phenotype and proteomic analyses. A second sample from the same patient will be collected at 24 hours, with an opportunistic sampling for stool culture. For control infants, only one set of blood and stool sample will be collected to coincide with clinical blood sampling. Along with detailed clinical information, blood and stool samples will be analysed and the information will be used to identify and validate the efficacy of immune-metabolic networks in the diagnosis of bacterial neonatal sepsis and to identify new host biomarkers for viral sepsis. ETHICS AND DISSEMINATION: The study has received research ethics committee approval from the Wales Research Ethics Committee 2 (reference 19/WA/0008) and operational approval from Health and Care Research Wales. Submission of study results for publication will involve making available all anonymised primary and processed data on public repository sites. TRIAL REGISTRATION NUMBER: NCT03777670.


Subject(s)
Neonatal Sepsis , Sepsis , Humans , Biomarkers , Cohort Studies , Multicenter Studies as Topic , Neonatal Sepsis/diagnosis , Neonatal Sepsis/microbiology , Observational Studies as Topic , Prospective Studies , Proteomics
18.
Mol Ther Methods Clin Dev ; 16: 21-31, 2020 Mar 13.
Article in English | MEDLINE | ID: mdl-31720306

ABSTRACT

Tissue-resident macrophages exhibit specialized phenotypes dependent on their in vivo physiological niche. Investigation of their function often relies upon complex whole mouse transgenic studies. While some appropriate lineage-associated promoters exist, there are no options for tissue-specific targeting of macrophages. We have developed full protocols for in vivo productive infection (defined by stable transgene expression) of tissue-resident macrophages with lentiviral vectors, enabling RNA and protein overexpression, including expression of small RNA species such as shRNA, to knock down and modulate gene expression. These approaches allow robust infection of peritoneal tissue-resident macrophages without significant infection of other cell populations. They permit rapid functional study of macrophages in homeostatic and inflammatory settings, such as thioglycolate-induced peritonitis, while maintaining the cells in their physiological context. Here we provide detailed protocols for the whole workflow: viral production, purification, and quality control; safety considerations for administration of the virus to mice; and assessment of in vivo transduction efficiency and the low background levels of inflammation induced by the virus. In summary, we present a quick and accessible protocol for the rapid assessment of gene function in peritoneal tissue-resident macrophages in vivo.

19.
Oxf Open Immunol ; 1(1): iqaa004, 2020.
Article in English | MEDLINE | ID: mdl-34192267

ABSTRACT

The coronavirus infectious disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a world health concern and can cause severe disease and high mortality in susceptible groups. While vaccines offer a chance to treat disease, prophylactic and anti-viral treatments are still of vital importance, especially in context of the mutative ability of this group of viruses. Therefore, it is essential to elucidate the molecular mechanisms of viral entry, innate sensing and immune evasion of SARS-CoV-2, which control the triggers of the subsequent excessive inflammatory response. Viral evasion strategies directly target anti-viral immunity, counteracting host restriction factors and hijacking signalling pathways to interfere with interferon production. In Part I of this review, we examine SARS-CoV-2 viral entry and the described immune evasion mechanisms to provide a perspective on how the failure in initial viral sensing by infected cells can lead to immune dysregulation causing fatal COVID-19, discussed in Part II.

20.
Oxf Open Immunol ; 1(1): iqaa005, 2020.
Article in English | MEDLINE | ID: mdl-34192268

ABSTRACT

The current pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a global health crisis and will likely continue to impact public health for years. As the effectiveness of the innate immune response is crucial to patient outcome, huge efforts have been made to understand how dysregulated immune responses may contribute to disease progression. Here we have reviewed current knowledge of cellular innate immune responses to SARS-CoV-2 infection, highlighting areas for further investigation and suggesting potential strategies for intervention. We conclude that in severe COVID-19 initial innate responses, primarily type I interferon, are suppressed or sabotaged which results in an early interleukin (IL)-6, IL-10 and IL-1ß-enhanced hyperinflammation. This inflammatory environment is driven by aberrant function of innate immune cells: monocytes, macrophages and natural killer cells dispersing viral pathogen-associated molecular patterns and damage-associated molecular patterns into tissues. This results in primarily neutrophil-driven pathology including fibrosis that causes acute respiratory distress syndrome. Activated leukocytes and neutrophil extracellular traps also promote immunothrombotic clots that embed into the lungs and kidneys of severe COVID-19 patients, are worsened by immobility in the intensive care unit and are perhaps responsible for the high mortality. Therefore, treatments that target inflammation and coagulation are promising strategies for reducing mortality in COVID-19.

SELECTION OF CITATIONS
SEARCH DETAIL