Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Nat Commun ; 15(1): 3402, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649734

ABSTRACT

The immune mechanisms mediating COVID-19 vaccine attenuation of COVID-19 remain undescribed. We conducted comprehensive analyses detailing immune responses to SARS-CoV-2 virus in blood post-vaccination with ChAdOx1 nCoV-19 or a placebo. Samples from randomised placebo-controlled trials (NCT04324606 and NCT04400838) were taken at baseline, onset of COVID-19-like symptoms, and 7 days later, confirming COVID-19 using nucleic amplification test (NAAT test) via real-time PCR (RT-PCR). Serum cytokines were measured with multiplexed immunoassays. The transcriptome was analysed with long, short and small RNA sequencing. We found attenuation of RNA inflammatory signatures in ChAdOx1 nCoV-19 compared with placebo vaccinees and reduced levels of serum proteins associated with COVID-19 severity. KREMEN1, a putative alternative SARS-CoV-2 receptor, was downregulated in placebo compared with ChAdOx1 nCoV-19 vaccinees. Vaccination ameliorates reductions in cell counts across leukocyte populations and platelets noted at COVID-19 onset, without inducing potentially deleterious Th2-skewed immune responses. Multi-omics integration links a global reduction in miRNA expression at COVID-19 onset to increased pro-inflammatory responses at the mRNA level. This study reveals insights into the role of COVID-19 vaccines in mitigating disease severity by abrogating pro-inflammatory responses associated with severe COVID-19, affirming vaccine-mediated benefit in breakthrough infection, and highlighting the importance of clinically relevant endpoints in vaccine evaluation.


Subject(s)
Breakthrough Infections , COVID-19 Vaccines , COVID-19 , ChAdOx1 nCoV-19 , Adult , Female , Humans , Male , Middle Aged , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Cytokines/blood , Inflammation/immunology , Multiomics , Transcriptome , Vaccination
3.
Mol Syst Biol ; 16(11): e9888, 2020 11.
Article in English | MEDLINE | ID: mdl-33210468

ABSTRACT

Neisseria meningitidis is a major cause of meningitis and septicaemia. A MenB vaccine (4CMenB) was licensed by the European Medicines Agency in January 2013. Here we describe the blood transcriptome and proteome following infant immunisations with or without concomitant 4CMenB, to gain insight into the molecular mechanisms underlying post-vaccination reactogenicity and immunogenicity. Infants were randomised to receive control immunisations (PCV13 and DTaP-IPV-Hib) with or without 4CMenB at 2 and 4 months of age. Blood gene expression and plasma proteins were measured prior to, then 4 h, 24 h, 3 days or 7 days post-vaccination. 4CMenB vaccination was associated with increased expression of ENTPD7 and increased concentrations of 4 plasma proteins: CRP, G-CSF, IL-1RA and IL-6. Post-vaccination fever was associated with increased expression of SELL, involved in neutrophil recruitment. A murine model dissecting the vaccine components found the concomitant regimen to be associated with increased gene perturbation compared with 4CMenB vaccine alone with enhancement of pathways such as interleukin-3, -5 and GM-CSF signalling. Finally, we present transcriptomic profiles predictive of immunological and febrile responses following 4CMenB vaccine.


Subject(s)
Fever/genetics , Immunity/genetics , Meningococcal Vaccines/immunology , Animals , Blood Chemical Analysis , Diphtheria-Tetanus-Pertussis Vaccine/adverse effects , Diphtheria-Tetanus-Pertussis Vaccine/immunology , Female , Fever/blood , Fever/epidemiology , Fever/etiology , Gene Expression Profiling , Haemophilus Vaccines/adverse effects , Haemophilus Vaccines/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Incidence , Infant , Male , Meningococcal Infections/prevention & control , Meningococcal Vaccines/adverse effects , Mice , Mice, Inbred C57BL , Microarray Analysis , Pneumococcal Vaccines/adverse effects , Pneumococcal Vaccines/immunology , Poliovirus Vaccine, Inactivated/adverse effects , Poliovirus Vaccine, Inactivated/immunology , Proteome/analysis , Transcriptome , Vaccination/adverse effects , Vaccines, Conjugate/adverse effects , Vaccines, Conjugate/immunology
4.
PLoS One ; 14(8): e0221143, 2019.
Article in English | MEDLINE | ID: mdl-31430297

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are a class of small regulatory RNAs around 21-25 nucleotides in length which govern many aspects of immunity including the host innate and adaptive responses to infection. RT-qPCR studies of select microRNAs show that vaccination alters the expression circulating microRNAs but the effect of vaccination on the global microRNA population (i.e. micronome) has never been studied. AIM: To describe vaccine associated changes in the expression of microRNAs 21 days after vaccination in children receiving a pandemic influenza (H1N1) vaccination. METHOD: Serum samples were obtained from children aged 6 months to 12 years enrolled in an open label randomised control trial of two pandemic influenza (H1N1) vaccines, in which participants received either ASO3B adjuvanted split virion or a whole virion non-adjuvanted vaccine. MicroRNA expression was profiled in a discovery cohort of participants prior to, and 21 days after vaccination using an Agilent microarray platform. Findings were followed up by RT-qPCR in the original discovery cohort and then in a validation cohort of participants taken from the same study. RESULTS: 44 samples from 22 children were assayed in a discovery cohort. The microarray results revealed 19 microRNAs were differentially expressed after vaccination after adjustment for multiple testing. The microarray detected ubiquitous expression of several microRNAs which could not be validated by RT-qPCR, many of which have little evidence of existence in publicly available RNA sequencing data. Real time PCR (RT-qPCR) confirmed downregulation of miR-142-3p in the discovery cohort. These findings were not replicated in the subsequent validation cohort (n = 22). CONCLUSION: This study is the first study to profile microRNA expression after vaccination. An important feature of this study is many of the differentially expressed microRNAs could not be detected and validated by RT-qPCR. This study highlights the care that should be taken when interpreting omics biomarker discovery, highlighting the need for supplementary methods to validate microRNA microarray findings, and emphasises the importance of validation cohorts. Data from similar studies which do not meet these requirements should be interpreted with caution.


Subject(s)
Circulating MicroRNA/isolation & purification , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Oligonucleotide Array Sequence Analysis , Child , Child, Preschool , Circulating MicroRNA/blood , Circulating MicroRNA/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation/immunology , Humans , Immunogenicity, Vaccine , Infant , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Influenza, Human/virology , Male , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Vaccination/methods
5.
Front Immunol ; 10: 1363, 2019.
Article in English | MEDLINE | ID: mdl-31275313

ABSTRACT

Sepsis has a complex pathophysiology in which both excessive and refractory inflammatory responses are hallmark features. Pro-inflammatory cytokine responses during the early stages are responsible for significant endothelial dysfunction, loss of endothelial integrity, and organ failure. In addition, it is now well-established that a substantial number of sepsis survivors experience ongoing immunological derangement and immunosuppression following a septic episode. The underpinning mechanisms of these phenomena are incompletely understood yet they contribute to a significant proportion of sepsis-associated mortality. Epigenetic mechanisms including DNA methylation, histone modifications, and non-coding RNAs, have an increasingly clear role in modulating inflammatory and other immunological processes. Recent evidence suggests epigenetic mechanisms are extensively perturbed as sepsis progresses, and particularly play a role in endothelial dysfunction and immunosuppression. Whilst therapeutic modulation of the epigenome is still in its infancy, there is substantial evidence from animal models that this approach could reap benefits. In this review, we summarize research elucidating the role of these mechanisms in several aspects of sepsis pathophysiology including tissue injury and immunosuppression. We also evaluate pre-clinical evidence for the use of "epi-therapies" in the treatment of poly-microbial sepsis.


Subject(s)
Endothelium/pathology , Epigenesis, Genetic/genetics , Immunologic Deficiency Syndromes/immunology , Shock, Septic/immunology , Shock, Septic/pathology , DNA Methylation/genetics , Endothelial Cells/pathology , Histone Acetyltransferases/metabolism , Histone Code/genetics , Histone Deacetylases/metabolism , Humans , RNA, Untranslated/genetics
6.
Front Immunol ; 8: 1182, 2017.
Article in English | MEDLINE | ID: mdl-28993774

ABSTRACT

MicroRNAs (miRNAs) are short single-stranded non-coding RNA sequences that posttranscriptionally regulate up to 60% of protein encoding genes. Evidence is emerging that miRNAs are key mediators of the host response to infection, predominantly by regulating proteins involved in innate and adaptive immune pathways. miRNAs can govern the cellular tropism of some viruses, are implicated in the resistance of some individuals to infections like HIV, and are associated with impaired vaccine response in older people. Not surprisingly, pathogens have evolved ways to undermine the effects of miRNAs on immunity. Recognition of this has led to new experimental treatments, RG-101 and Miravirsen-hepatitis C treatments which target host miRNA. miRNAs are being investigated as novel infection biomarkers, and they are being used to design attenuated vaccines, e.g., against Dengue virus. This comprehensive review synthesizes current knowledge of miRNA in host response to infection with emphasis on potential clinical applications, along with an evaluation of the challenges still to be overcome.

7.
J Pathol ; 231(3): 388-99, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23939832

ABSTRACT

Carcinoma-associated fibroblasts (CAFs) influence the behaviour of cancer cells but the roles of microRNAs in this interaction are unknown. We report microRNAs that are differentially expressed between breast normal fibroblasts and CAFs of oestrogen receptor-positive cancers, and explore the influences of one of these, miR-26b, on breast cancer biology. We identified differentially expressed microRNAs by expression profiling of clinical samples and a tissue culture model: miR-26b was the most highly deregulated microRNA. Using qPCR, miR-26b was confirmed as down-regulated in fibroblasts from 15 of 18 further breast cancers. Next, we examined whether manipulation of miR-26b expression changed breast fibroblast behaviour. Reduced miR-26b expression caused fibroblast migration and invasion to increase by up to three-fold in scratch-closure and trans-well assays. Furthermore, in co-culture with MCF7 breast cancer epithelial cells, fibroblasts with reduced miR-26b expression enhanced both MCF7 migration in trans-well assays and MCF7 invasion from three-dimensional spheroids by up to five-fold. Mass spectrometry was used to identify expression changes associated with the reduction of miR-26b expression in fibroblasts. Pathway analyses of differentially expressed proteins revealed that glycolysis/TCA cycle and cytoskeletal regulation by Rho GTPases are downstream of miR-26b. In addition, three novel miR-26b targets were identified (TNKS1BP1, CPSF7, COL12A1) and the expression of each in cancer stroma was shown to be significantly associated with breast cancer recurrence. MiR-26b in breast CAFs is a potent regulator of cancer behaviour in oestrogen receptor-positive cancers, and we have identified key genes and molecular pathways that act downstream of miR-26b in CAFs.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Fibroblasts/metabolism , MicroRNAs/metabolism , Receptors, Estrogen/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Coculture Techniques , Down-Regulation , Female , Fibroblasts/pathology , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Paracrine Communication , Polymerase Chain Reaction , Signal Transduction , Time Factors , Transfection , Tumor Microenvironment
8.
J Pathol ; 227(4): 385-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22488672

ABSTRACT

MicroRNAs are a class of small regulatory RNAs that influence the stabilities and translational efficiencies of target mRNAs. They have been implicated in an increasing number of biological processes, including carcinogenesis. A huge body of literature exists documenting up- or down-regulation of specific microRNAs during carcinogenesis and identifying molecular pathways by which these microRNAs influence every aspect of cancer development, including proliferation, angiogenesis, and metastasis. These studies have provided many insights into basic cancer biology as well as allowing identification of novel biomarkers and potential drug targets. However, the vast bulk of this literature concerns solid epithelial tumours, while sarcomas remain relatively under-studied. The purpose of this article is to review the roles of microRNAs in sarcomas and to highlight microRNAs or related molecular pathways that demonstrate consistent roles within individual or across sarcoma subtypes, with a view to identifying the key regulatory molecules. Further insights into sarcoma biology may be particularly valuable since sarcomas represent a tumour group with a particularly poor prognosis and rather limited treatment options.


Subject(s)
MicroRNAs/physiology , Sarcoma/physiopathology , Cell Proliferation , Gene Expression Regulation, Neoplastic/physiology , Humans , Neoplasm Metastasis/physiopathology , Neovascularization, Pathologic/physiopathology , Sarcoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL