210 results on '"Sheila E. Francis"'
Search Results
2. Transient increase in atherosclerotic plaque macrophage content following Streptococcus pneumoniae pneumonia in ApoE-deficient mice
- Author
-
Rohit Bazaz, Helen M. Marriott, Carl Wright, Janet Chamberlain, Laura E. West, Catherine Gelsthorpe, Paul R. Heath, Afsaneh Maleki-Dizaji, Sheila E. Francis, and David H. Dockrell
- Subjects
pneumococcus ,myocardial infarction ,atherosclerosis ,acute coronary syndrome ,statin ,Microbiology ,QR1-502 - Abstract
IntroductionDespite epidemiological associations between community acquired pneumonia (CAP) and myocardial infarction, mechanisms that modify cardiovascular disease during CAP are not well defined. In particular, largely due to a lack of relevant experimental models, the effect of pneumonia on atherosclerotic plaques is unclear. We describe the development of a murine model of the commonest cause of CAP, Streptococcus pneumoniae pneumonia, on a background of established atherosclerosis. We go on to use our model to investigate the effects of pneumococcal pneumonia on atherosclerosis.MethodsC57BL/6J and ApoE-/- mice were fed a high fat diet to promote atherosclerotic plaque formation. Mice were then infected with a range of S. pneumoniae serotypes (1, 4 or 14) with the aim of establishing a model to study atherosclerotic plaque evolution after pneumonia and bacteremia. Laser capture microdissection of plaque macrophages enabled transcriptomic analysis.ResultsIntratracheal instillation of S. pneumoniae in mice fed a cholate containing diet resulted in low survival rates following infection, suggestive of increased susceptibility to severe infection. Optimization steps resulted in a final model of male ApoE-/- mice fed a Western diet then infected by intranasal instillation of serotype 4 (TIGR4) S. pneumoniae followed by antibiotic administration. This protocol resulted in high rates of bacteremia (88.9%) and survival (88.5%). Pneumonia resulted in increased aortic sinus plaque macrophage content 2 weeks post pneumonia but not at 8 weeks, and no difference in plaque burden or other plaque vulnerability markers were found at either time point. Microarray and qPCR analysis of plaque macrophages identified downregulation of two E3 ubiquitin ligases, Huwe1 and Itch, following pneumonia. Treatment with atorvastatin failed to alter plaque macrophage content or other plaque features.DiscussionWithout antibiotics, ApoE-/- mice fed a high fat diet were highly susceptible to mortality following S. pneumoniae infection. The major infection associated change in plaque morphology was an early increase in plaque macrophages. Our results also hint at a role for the ubiquitin proteasome system in the response to pneumococcal infection in the plaque microenvironment.
- Published
- 2023
- Full Text
- View/download PDF
3. Tribbles 3 deficiency promotes atherosclerotic fibrous cap thickening and macrophage-mediated extracellular matrix remodelling
- Author
-
Laura Martinez-Campesino, Klaudia Kocsy, Jaime Cañedo, Jessica M. Johnston, Charlotte E. Moss, Simon A. Johnston, Stephen Hamby, Alison H. Goodall, Jessica Redgrave, Sheila E. Francis, Endre Kiss-Toth, and Heather L. Wilson
- Subjects
atherosclerosis ,TRIB3 ,macrophage ,fibrous ,cap ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Tribbles 3 (TRIB3) modulates lipid and glucose metabolism, macrophage lipid uptake, with a gain-of-function variant associated with increased cardiovascular risk. Here we set out to examine the role of this pseudokinase in atherosclerotic plaque development. Human endarterectomy atherosclerotic tissue specimens analysed by immunofluorescence showed upregulated TRIB3 in unstable plaques and an enrichment in unstable regions of stable plaques. Atherosclerosis was induced in full body Trib3KO and Trib3WT littermate mice by injecting mPCSK9 expressing adeno-associated virus and western diet feeding for 12 weeks. Trib3KO mice showed expanded visceral adipose depot while circulatory lipid levels remained unaltered compared to wildtype mice. Trib3KO mice aortae showed a reduced plaque development and improved plaque stability, with increased fibrous cap thickness and collagen content, which was accompanied by increased macrophage content. Analysis of both mouse and human macrophages with reduced TRIB3 expression showed elongated morphology, increased actin expression and altered regulation of genes involved in extracellular matrix remodelling. In summary, TRIB3 controls plaque development and may be atherogenic in vivo. Loss of TRIB3 increases fibrous cap thickness via altered metalloproteinase expression in macrophages, thus inhibiting collagen and elastic fibre degradation, suggesting a role for TRIB3 in the formation of unstable plaques.
- Published
- 2022
- Full Text
- View/download PDF
4. A therapeutic antibody targeting osteoprotegerin attenuates severe experimental pulmonary arterial hypertension
- Author
-
Nadine D. Arnold, Josephine A. Pickworth, Laura E. West, Sarah Dawson, Joana A. Carvalho, Helen Casbolt, Adam T. Braithwaite, James Iremonger, Lewis Renshall, Volker Germaschewski, Matthew McCourt, Philip Bland-Ward, Hager Kowash, Abdul G. Hameed, Alexander M. K. Rothman, Maria G. Frid, A. A. Roger Thompson, Holly R. Evans, Mark Southwood, Nicholas W. Morrell, David C. Crossman, Moira K. B. Whyte, Kurt R. Stenmark, Christopher M. Newman, David G. Kiely, Sheila E. Francis, and Allan Lawrie
- Subjects
Science - Abstract
Pulmonary arterial hypertension (PAH) is characterised by progressive pulmonary vascular remodelling. Here, Arnold et al. develop a therapeutic antibody targeting osteoprotegerin and find it attenuates pulmonary vascular remodelling in multiple rodent models of PAH, alone or in combination with standard of care vasodilator therapy.
- Published
- 2019
- Full Text
- View/download PDF
5. Neurovascular dysfunction in vascular dementia, Alzheimer’s and atherosclerosis
- Author
-
Osman Shabir, Jason Berwick, and Sheila E. Francis
- Subjects
Neurovascular coupling ,Atherosclerosis ,Dementia ,Mouse ,Neuroimaging ,Disease modelling ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurophysiology and neuropsychology ,QP351-495 - Abstract
Abstract Efficient blood supply to the brain is of paramount importance to its normal functioning and improper blood flow can result in potentially devastating neurological consequences. Cerebral blood flow in response to neural activity is intrinsically regulated by a complex interplay between various cell types within the brain in a relationship termed neurovascular coupling. The breakdown of neurovascular coupling is evident across a wide variety of both neurological and psychiatric disorders including Alzheimer’s disease. Atherosclerosis is a chronic syndrome affecting the integrity and function of major blood vessels including those that supply the brain, and it is therefore hypothesised that atherosclerosis impairs cerebral blood flow and neurovascular coupling leading to cerebrovascular dysfunction. This review will discuss the mechanisms of neurovascular coupling in health and disease and how atherosclerosis can potentially cause cerebrovascular dysfunction that may lead to cognitive decline as well as stroke. Understanding the mechanisms of neurovascular coupling in health and disease may enable us to develop potential therapies to prevent the breakdown of neurovascular coupling in the treatment of vascular brain diseases including vascular dementia, Alzheimer’s disease and stroke.
- Published
- 2018
- Full Text
- View/download PDF
6. The IL-1RI Co-Receptor TILRR (FREM1 Isoform 2) Controls Aberrant Inflammatory Responses and Development of Vascular Disease
- Author
-
Sarah A. Smith, PhD, Andriy O. Samokhin, PhD, Mabruka Alfadi, PhD, Emer C. Murphy, PhD, David Rhodes, MSc, W. Mike L. Holcombe, PhD, Endre Kiss-Toth, PhD, Robert F. Storey, BSc, BM, DM, Siu-Pok Yee, PhD, Sheila E. Francis, PhD, and Eva E. Qwarnstrom, PhD
- Subjects
heparan sulfate proteoglycan ,interleukin-1 receptor ,IL-1RI ,NF-κB ,TILRR ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Expression of the interleukin-1 receptor type I (IL-1RI) co-receptor Toll-like and interleukin-1 receptor regulator (TILRR) is significantly increased in blood monocytes following myocardial infarction and in the atherosclerotic plaque, whereas levels in healthy tissue are low. TILRR association with IL-1RI at these sites causes aberrant activation of inflammatory genes, which underlie progression of cardiovascular disease. The authors show that genetic deletion of TILRR or antibody blocking of TILRR function reduces development of atherosclerotic plaques. Lesions exhibit decreased levels of monocytes, with increases in collagen and smooth muscle cells, characteristic features of stable plaques. The results suggest that TILRR may constitute a rational target for site- and signal-specific inhibition of vascular disease.
- Published
- 2017
- Full Text
- View/download PDF
7. Shear stress induces endothelial-to-mesenchymal transition via the transcription factor Snail
- Author
-
Marwa M. Mahmoud, Jovana Serbanovic-Canic, Shuang Feng, Celine Souilhol, Rouyu Xing, Sarah Hsiao, Akiko Mammoto, Jing Chen, Markus Ariaans, Sheila E. Francis, Kim Van der Heiden, Victoria Ridger, and Paul C. Evans
- Subjects
Medicine ,Science - Abstract
Abstract Blood flow influences atherosclerosis by generating wall shear stress, which alters endothelial cell (EC) physiology. Low shear stress induces dedifferentiation of EC through a process termed endothelial-to-mesenchymal transition (EndMT). The mechanisms underlying shear stress-regulation of EndMT are uncertain. Here we investigated the role of the transcription factor Snail in low shear stress-induced EndMT. Studies of cultured EC exposed to flow revealed that low shear stress induced Snail expression. Using gene silencing it was demonstrated that Snail positively regulated the expression of EndMT markers (Slug, N-cadherin, α-SMA) in EC exposed to low shear stress. Gene silencing also revealed that Snail enhanced the permeability of endothelial monolayers to macromolecules by promoting EC proliferation and migration. En face staining of the murine aorta or carotid arteries modified with flow-altering cuffs demonstrated that Snail was expressed preferentially at low shear stress sites that are predisposed to atherosclerosis. Snail was also expressed in EC overlying atherosclerotic plaques in coronary arteries from patients with ischemic heart disease implying a role in human arterial disease. We conclude that Snail is an essential driver of EndMT under low shear stress conditions and may promote early atherogenesis by enhancing vascular permeability.
- Published
- 2017
- Full Text
- View/download PDF
8. X-ray Micro-Computed Tomography: An Emerging Technology to Analyze Vascular Calcification in Animal Models
- Author
-
Samantha J. Borland, Julia Behnsen, Nick Ashton, Sheila E. Francis, Keith Brennan, Michael J. Sherratt, Philip J. Withers, and Ann E. Canfield
- Subjects
vascular calcification ,micro-CT ,mouse models ,histology ,correlative microscopy ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Vascular calcification describes the formation of mineralized tissue within the blood vessel wall, and it is highly associated with increased cardiovascular morbidity and mortality in patients with chronic kidney disease, diabetes, and atherosclerosis. In this article, we briefly review different rodent models used to study vascular calcification in vivo, and critically assess the strengths and weaknesses of the current techniques used to analyze and quantify calcification in these models, namely 2-D histology and the o-cresolphthalein assay. In light of this, we examine X-ray micro-computed tomography (µCT) as an emerging complementary tool for the analysis of vascular calcification in animal models. We demonstrate that this non-destructive technique allows us to simultaneously quantify and localize calcification in an intact vessel in 3-D, and we consider recent advances in µCT sample preparation techniques. This review also discusses the potential to combine 3-D µCT analyses with subsequent 2-D histological, immunohistochemical, and proteomic approaches in correlative microscopy workflows to obtain rich, multifaceted information on calcification volume, calcification load, and signaling mechanisms from within the same arterial segment. In conclusion we briefly discuss the potential use of µCT to visualize and measure vascular calcification in vivo in real-time.
- Published
- 2020
- Full Text
- View/download PDF
9. Author Correction: Shear stress induces endothelial-to-mesenchymal transition via the transcription factor Snail
- Author
-
Marwa M. Mahmoud, Jovana Serbanovic-Canic, Shuang Feng, Celine Souilhol, Rouyu Xing, Sarah Hsiao, Akiko Mammoto, Jing Chen, Markus Ariaans, Sheila E. Francis, Kim Van der Heiden, Victoria Ridger, and Paul C. Evans
- Subjects
Medicine ,Science - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
10. Dietary Docosahexaenoic Acid Reduces Oscillatory Wall Shear Stress, Atherosclerosis, and Hypertension, Most Likely Mediated via an IL‐1–Mediated Mechanism
- Author
-
Mabruka A. Alfaidi, Janet Chamberlain, Alexander Rothman, David Crossman, Maria‐Cruz Villa‐Uriol, Patrick Hadoke, Junxi Wu, Torsten Schenkel, Paul C. Evans, and Sheila E. Francis
- Subjects
docosahexaenoic acid ,endothelium ,hypertension ,inflammation ,interleukin 1 ,wall shear stress ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background Hypertension is a complex condition and a common cardiovascular risk factor. Dietary docosahexaenoic acid (DHA) modulates atherosclerosis and hypertension, possibly via an inflammatory mechanism. IL‐1 (interleukin 1) has an established role in atherosclerosis and inflammation, although whether IL‐1 inhibition modulates blood pressure is unclear. Methods and Results Male apoE−/− (apolipoprotein E–null) mice were fed either a high fat diet or a high fat diet plus DHA (300 mg/kg per day) for 12 weeks. Blood pressure and cardiac function were assessed, and effects of DHA on wall shear stress and atherosclerosis were determined. DHA supplementation improved left ventricular function, reduced wall shear stress and oscillatory shear at ostia in the descending aorta, and significantly lowered blood pressure compared with controls (119.5±7 versus 159.7±3 mm Hg, P
- Published
- 2018
- Full Text
- View/download PDF
11. Differential IL-1 signaling induced by BMPR2 deficiency drives pulmonary vascular remodeling
- Author
-
Josephine Pickworth, Alexander Rothman, James Iremonger, Helen Casbolt, Kay Hopkinson, Peter M. Hickey, Santhi Gladson, Sheila Shay, Nicholas W. Morrell, Sheila E. Francis, James D. West, and Allan Lawrie
- Subjects
Diseases of the circulatory (Cardiovascular) system ,RC666-701 ,Diseases of the respiratory system ,RC705-779 - Abstract
Bone morphogenetic protein receptor type 2 (BMPR2) mutations are present in patients with heritable and idiopathic pulmonary arterial hypertension (PAH). Circulating levels of interleukin-1 (IL-1) are raised in patients and animal models. Whether interplay between BMP and IL-1 signaling can explain the local manifestation of PAH in the lung remains unclear. Cell culture, siRNA, and mRNA microarray analysis of RNA isolated from human pulmonary artery (PASMC) and aortic (AoSMC) smooth muscle cells were used. R899X +/– BMPR2 transgenic mice fed a Western diet for six weeks were given daily injections of IL-1ß prior to assessment for PAH and tissue collection. PASMC have reduced inflammatory activation in response to IL-1ß compared with AoSMCs; however, PASMC with reduced BMPR2 demonstrated an exaggerated response. Mice treated with IL-1ß had higher white blood cell counts and significantly raised serum protein levels of IL-6 and osteoprotegerin (OPG) plasma levels recapitulating in vitro data. Phenotypically, IL-1ß treated mice demonstrated increased pulmonary vascular remodeling. IL-1ß induces an exaggerated pulmonary artery specific transcriptomic inflammatory response when BMPR2 signaling is reduced.
- Published
- 2017
- Full Text
- View/download PDF
12. TRAIL Deficient Mice Are Protected from Sugen/Hypoxia Induced Pulmonary Arterial Hypertension
- Author
-
Sarah H. Dawson, Nadine D. Arnold, Josephine A. Pickworth, Sheila E. Francis, and Allan Lawrie
- Subjects
TRAIL ,murine model ,pulmonary arterial hypertension ,sugen/hypoxia ,Medicine - Abstract
Pulmonary arterial hypertension (PAH) is a progressive lung disease diagnosed by an increase in pulmonary arterial blood pressure that is driven by a progressive vascular remodelling of small pulmonary arterioles. We have previously reported that tumor necrosis factor apoptosis-inducing ligand (TRAIL) protein expression is increased in pulmonary vascular lesions and pulmonary artery smooth muscle cells (PASMC) of patients with idiopathic PAH. The addition of recombinant TRAIL induces the proliferation and migration of PASMCs in vitro. TRAIL is required for hypoxia-induced pulmonary hypertension in mice, and blockade of TRAIL prevents and reduces disease development in other rodent models of PAH. Due to the availability of knockout and transgenic mice, murine models of disease are key to further advances in understanding the complex and heterogeneous pathogenesis of PAH. However, murine models vary in their disease severity, and are often criticized for lacking the proliferative pulmonary vascular lesions characteristic of PAH. The murine Sugen-hypoxic (SuHx) mouse model has recently been reported to have a more severe PAH phenotype consisting advanced pulmonary vascular remodelling. We therefore aimed to determine whether TRAIL was also required for the development of PAH in this model. C57BL/6 and TRAIL−/− mice were exposed to normoxia, Sugen5416 alone, hypoxia or both Sugen5416 and hypoxia (SuHx). We report here that SuHx treated C57BL/6 mice developed more severe PAH than hypoxia alone, and that TRAIL−/− mice were protected from disease development. These data further emphasise the importance of this pathway and support the use of the SuHx mouse model for investigating the importance of potential mediators in PAH pathogenesis.
- Published
- 2014
- Full Text
- View/download PDF
13. Enhanced Macrophage Tribbles-1 Expression in Murine Experimental Atherosclerosis
- Author
-
Adrienn Angyal, Endre Kiss-Toth, Vanessa Ernst, Karen Holland, Nadine D. Arnold, Sheila E. Francis, and Hye Youn Sung
- Subjects
Tribbles ,atherosclerosis ,macrophages ,mouse ,Biology (General) ,QH301-705.5 - Abstract
Development of the atherosclerotic plaque involves a complex interplay between a number of cell types and an extensive inter-cellular communication via cell bound as well as soluble mediators. The family of tribbles proteins has recently been identified as novel controllers of pro-inflammatory signal transduction. The objective of this study was to address the expression pattern of all three tribbles proteins in atherosclerotic plaques from a mouse model of atherosclerosis. Each tribbles were expressed in vascular smooth muscle cells, endothelial cells as well as in resident macrophages of mouse atherosclerotic plaques. The role of IL-1 mediated inflammatory events in controlling tribbles expression was also addressed by inducing experimental atherosclerosis in ApoE−/−IL1R1−/− (double knockout) mice. Immunohistochemical analysis of these mice showed a selective decrease in the percentage of trb-1 expressing macrophages, compared to the ApoE−/− cohort (14.7% ± 1.55 vs. 26.3% ± 1.19). The biological significance of this finding was verified in vitro where overexpression of trb-1 in macrophages led to a significant attenuation (~70%) of IL-6 production as well as a suppressed IL-12 expression induced by a proinflammatory stimulus. In this in vitro setting, expression of truncated trb-1 mutants suggests that the kinase domain of this protein is sufficient to exert this inhibitory action.
- Published
- 2012
- Full Text
- View/download PDF
14. IL-1B drives opposing responses in primary tumours and bone metastases; harnessing combination therapies to improve outcome in breast cancer
- Author
-
Sheila E. Francis, Charlotte K. Moore, Victoria J. Cookson, Diane V. Lefley, Russell Hughes, Paul R. Heath, Catherine A. Evans, Amy R. Spicer-Hadlington, Khawla Ahmed, A. Graham Pockley, Jayakumar Vadakekolathu, Ana E. Amariutei, Penelope D. Ottewell, Emmanuel Pinteaux, Lewis A. Quayle, and Claudia Tulotta
- Subjects
0301 basic medicine ,Combination therapy ,medicine.medical_treatment ,Article ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Immune system ,Medicine ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging ,RC254-282 ,business.industry ,Bone metastasis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell migration ,medicine.disease ,030104 developmental biology ,Zoledronic acid ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Tumour immunology ,business ,medicine.drug - Abstract
Breast cancer bone metastasis is currently incurable, ~75% of patients with late-stage breast cancer develop disease recurrence in bone and available treatments are only palliative. We have previously shown that production of the pro-inflammatory cytokine interleukin-1B (IL-1B) by breast cancer cells drives bone metastasis in patients and in preclinical in vivo models. In the current study, we have investigated how IL-1B from tumour cells and the microenvironment interact to affect primary tumour growth and bone metastasis through regulation of the immune system, and whether targeting IL-1 driven changes to the immune response improves standard of care therapy for breast cancer bone metastasis. Using syngeneic IL-1B/IL1R1 knock out mouse models in combination with genetic manipulation of tumour cells to overexpress IL-1B/IL1R1, we found that IL-1B signalling elicited an opposite response in primary tumours compared with bone metastases. In primary tumours, IL-1B inhibited growth, by impairing the infiltration of innate immune cell subsets with potential anti-cancer functions but promoted enhanced tumour cell migration. In bone, IL-1B stimulated the development of osteolytic metastases. In syngeneic models of breast cancer, combining standard of care treatments (Doxorubicin and Zoledronic acid) with the IL-1 receptor antagonist Anakinra inhibited both primary tumour growth and metastasis. Anakinra had opposite effects on the immune response compared to standard of care treatment, and its anti-inflammatory signature was maintained in the combination therapy. These data suggest that targeting IL-1B signalling may provide a useful therapeutic approach to inhibit bone metastasis and improve efficacy of current treatments for breast cancer patients.
- Published
- 2021
15. Cell-specific conditional deletion of interleukin-1 (IL-1) ligands and its receptors: a new toolbox to study the role of IL-1 in health and disease
- Author
-
Neil E. Humphreys, Werner Müller, Emmanuel Pinteaux, Ari Waisman, Ilgiz A. Mufazalov, Wesam H. Abdulaal, Sheila E. Francis, and Maj Simonsen-Jackson
- Subjects
Cell signaling ,medicine.medical_treatment ,Cre recombinase ,Review ,Interleukin 1 receptor, type II ,Biology ,Ligands ,03 medical and health sciences ,0302 clinical medicine ,Cre/loxP ,Drug Discovery ,medicine ,Animals ,Humans ,Conditional deletion ,Receptor ,Genetics (clinical) ,IL-1 receptors ,030304 developmental biology ,Inflammation ,0303 health sciences ,IL-1 ,Immunity ,Receptors, Interleukin-1 ,Cell biology ,Cytokine ,Genetically Engineered Mouse ,Molecular Medicine ,Cre-Lox recombination ,Interleukin 1 receptor, type I ,030217 neurology & neurosurgery ,Interleukin-1 ,Signal Transduction - Abstract
The pro-inflammatory cytokine interleukin-1 (IL-1) plays a key role in many physiological processes and during the inflammatory and immune response to most common diseases. IL-1 exists as two agonists, IL-1α and IL-1β that bind to the only signaling IL-1 type 1 receptor (IL-1R1), while a second decoy IL-1 type 2 receptor (IL-1R2) binds both forms of IL-1 without inducing cell signaling. The field of immunology and inflammation research has, over the past 35 years, unraveled many mechanisms of IL-1 actions, through in vitro manipulation of the IL-1 system or by using genetically engineered mouse models that lack either member of the IL-1 family in ubiquitous constitutive manner. However, the limitation of global mouse knockout technology has significantly hampered our understanding of the precise mechanisms of IL-1 actions in animal models of disease. Here we report and review the recent generation of new conditional mouse mutants in which exons of Il1a, Il1b, Il1r1, and Il1r2 genes flanked by loxP sites (fl/fl) can be deleted in cell-/tissue-specific constitutive or inducible manner by Cre recombinase expression. Hence, IL-1αfl/fl, IL-1βfl/fl, IL-1R1fl/fl, and IL-1R2fl/fl mice constitute a new toolbox that will provide a step change in our understanding of the cell-specific role of IL-1 and its receptor in health and disease and the potential development of targeted IL-1 therapies.
- Published
- 2020
16. Assessment of neurovascular coupling and cortical spreading depression in mixed mouse models of atherosclerosis and Alzheimer’s disease
- Author
-
Osman Shabir, Ben Pendry, Llywelyn Lee, Beth Eyre, Paul S Sharp, Monica A Rebollar, David Drew, Clare Howarth, Paul R Heath, Stephen B Wharton, Sheila E Francis, and Jason Berwick
- Subjects
Male ,Neurovascular ,Mouse ,QH301-705.5 ,Science ,General Biochemistry, Genetics and Molecular Biology ,CSD ,Amyloid beta-Protein Precursor ,Mice ,Alzheimer Disease ,Animals ,Biology (General) ,General Immunology and Microbiology ,General Neuroscience ,Cortical Spreading Depression ,Hemodynamics ,Brain ,General Medicine ,Alzheimer's disease ,Mice, Inbred C57BL ,Disease Models, Animal ,comorbidity ,Cerebrovascular Circulation ,Neurovascular Coupling ,Medicine ,atherosclerosis ,Alzheimer’s disease ,Research Article ,Neuroscience ,dementia - Abstract
Neurovascular coupling is a critical brain mechanism whereby changes to blood flow accompany localised neural activity. The breakdown of neurovascular coupling is linked to the development and progression of several neurological conditions including dementia. In this study, we examined cortical haemodynamics in mouse preparations that modelled Alzheimer’s disease (J20-AD) and atherosclerosis (PCSK9-ATH) between 9 and 12 m of age. We report novel findings with atherosclerosis where neurovascular decline is characterised by significantly reduced blood volume, altered levels of oxyhaemoglobin and deoxyhaemoglobin, in addition to global neuroinflammation. In the comorbid mixed model (J20-PCSK9-MIX), we report a 3 x increase in hippocampal amyloid-beta plaques. A key finding was that cortical spreading depression (CSD) due to electrode insertion into the brain was worse in the diseased animals and led to a prolonged period of hypoxia. These findings suggest that systemic atherosclerosis can be detrimental to neurovascular health and that having cardiovascular comorbidities can exacerbate pre-existing Alzheimer’s-related amyloid-plaques.
- Published
- 2022
17. Author response: Assessment of neurovascular coupling and cortical spreading depression in mixed mouse models of atherosclerosis and Alzheimer’s disease
- Author
-
Osman Shabir, Ben Pendry, Llywelyn Lee, Beth Eyre, Paul S Sharp, Monica A Rebollar, David Drew, Clare Howarth, Paul R Heath, Stephen B Wharton, Sheila E Francis, and Jason Berwick
- Published
- 2021
18. BS1 Reprogramming human macrophages with drug X: potential mechanisms for stabilisation of atherosclerotic plaques
- Author
-
Endre Kiss-Toth, Jessica Redgrave, Arshad Majid, Sheila E. Francis, and Klaudia Kocsy
- Subjects
business.industry ,CD14 ,Gene expression ,OLR1 ,Medicine ,Macrophage ,Pharmacology ,Scavenger receptor ,business ,In vitro ,Mannose receptor ,MSR1 - Abstract
Introduction Ischemic stroke is commonly caused by large artery atherosclerosis. Patients with a high atherosclerotic burden (stenosis) and inflamed or ulcerated plaque are at increased risk of early recurrent ischaemic events. Treatment with anti-inflammatory agents may therefore reduce stroke incidence and recurrence in patients with this condition, but in vitro human studies of the possible mechanisms are lacking. We hypothesised that an anti-inflammatory compound (denoted drug X*) alters the expression of macrophage specific genes, including scavenger receptors in in vitro models of human carotid plaque macrophages. Methods CD14+ monocytes were collected from whole blood donations (volunteers) and differentiated to monocyte derived macrophages (MDMs). Macrophage polarisation with M1 (100 ng/ml LPS + 20 ng/ml INF-γ) and M2a (20 ng/ml IL-4) markers was performed in complete media supplemented with 3 different clinically relevant concentrations of drug X. Gene expression (RT-qPCR) analysis (n=10) was carried out in the differentiated hMDMs to test for macrophage specific genes and scavenger receptors. Western Blot analysis (n=7) and immunofluorescence stain (IF) (n=5) was performed on drug X- treated MDMs to validate changes in the expression of Oxidized Low Density Lipoprotein Receptor 1 (OLR1). The physiological effect of drug X was also tested on oxLDL (25 μg/ml) uptake by macrophages (n=5). Results Our data suggest that drug X may re-program macrophages to a less inflammatory state (an M2 state) by significantly altering Mannose Receptor C-type 1 (MRC1) gene expression (p:0.0183). Drug X also alters the expression level of Macrophage Scavenger Receptor 1 (MSR1) gene, provoking the pacification of the M1 state and resulting in a beneficial effect on LDL uptake (p: 0.0845). Using OLR1 gene expression as a readout, in both M1 and M2a macrophages, the average OLR1 expression was reduced by treatment with drug X (p: 0.0003). This implies that M1 macrophages can bind less oxLDL which could reduce the formation of atherosclerotic plaque in the vessel wall. OLR1 protein expression was also reduced (p:0.0003) following 24-hour exposure of the macrophages to drug X. In functional tests, oxLDL uptake by drug X treated hMDMs was reduced by 44% (p:0.003). Conclusions The gene expression of macrophage specific genes and scavenger receptors in human M1 and M2a macrophages was significantly altered following 24-hour exposure to clinically relevant concentrations of drug X in vitro. Drug X reduced ‘active’ and pathogenic cell behaviours e.g., lipid uptake and promoted the polarisation to the M2 state. These data demonstrate that human macrophages can be reprogrammed to a less pathogenic state in vitro and they provide a potential mechanism for the effectiveness of drug X in the stabilisation of carotid atherosclerotic plaques in humans. * Drug X is used for ongoing IP/patent assessment Conflict of Interest No
- Published
- 2021
19. Frataxin and endothelial cell senescence in pulmonary hypertension
- Author
-
Allan Lawrie and Sheila E. Francis
- Subjects
0301 basic medicine ,Drug ,Senescence ,Male ,media_common.quotation_subject ,Hypertension, Pulmonary ,Pharmacology ,Vascular Remodeling ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Iron-Binding Proteins ,medicine ,Animals ,Humans ,Senolytic ,Cellular Senescence ,media_common ,Endothelial Progenitor Cells ,Mice, Knockout ,Navitoclax ,biology ,business.industry ,Endothelial Cells ,General Medicine ,medicine.disease ,Pulmonary hypertension ,Endothelial stem cell ,030104 developmental biology ,Blood pressure ,chemistry ,Friedreich Ataxia ,030220 oncology & carcinogenesis ,Frataxin ,biology.protein ,Commentary ,Female ,Endothelium, Vascular ,business ,Research Article - Abstract
The dynamic regulation of endothelial pathophenotypes in pulmonary hypertension (PH) remains undefined. Cellular senescence is linked to PH with intracardiac shunts; however, its regulation across PH subtypes is unknown. Since endothelial deficiency of iron-sulfur (Fe-S) clusters is pathogenic in PH, we hypothesized that a Fe-S biogenesis protein, frataxin (FXN), controls endothelial senescence. An endothelial subpopulation in rodent and patient lungs across PH subtypes exhibited reduced FXN and elevated senescence. In vitro, hypoxic and inflammatory FXN deficiency abrogated activity of endothelial Fe-S–containing polymerases, promoting replication stress, DNA damage response, and senescence. This was also observed in stem cell–derived endothelial cells from Friedreich’s ataxia (FRDA), a genetic disease of FXN deficiency, ataxia, and cardiomyopathy, often with PH. In vivo, FXN deficiency–dependent senescence drove vessel inflammation, remodeling, and PH, whereas pharmacologic removal of senescent cells in Fxn-deficient rodents ameliorated PH. These data offer a model of endothelial biology in PH, where FXN deficiency generates a senescent endothelial subpopulation, promoting vascular inflammatory and proliferative signals in other cells to drive disease. These findings also establish an endothelial etiology for PH in FRDA and left heart disease and support therapeutic development of senolytic drugs, reversing effects of Fe-S deficiency across PH subtypes.
- Published
- 2021
20. BS30 Light sheet imaging to analyse the spatial distribution of proteins in atherosclerotic plaques
- Author
-
Nicholas Van-Hateren, Maria Fragiadaki, Sheila E. Francis, Paul C. Evans, and Ziqi Zhou
- Subjects
biology ,Enos ,Confocal microscopy ,law ,Optical clearing ,Microscopy ,Biophysics ,Shear stress ,Plaque growth ,biology.organism_classification ,Clearance ,law.invention ,Staining - Abstract
The mechanisms linking shear stress, endothelial physiology and plaque biology are currently poorly understood, but their elucidation could identify new strategies to reduce plaque growth and rupture. To address this, we use eNOS as high shear stress marker coupled to immunofluorescent staining, optical clearing and light-sheet microscopy, to develop a system for analysing the spatial distribution of proteins in murine plaques and correlate them with local WSS. Results Confocal microscopy of concavity mounted slides revealed strong eNOS staining at the outer curvature of WT mice but significantly reduced staining at the inner curvature (N=5; P Light-sheet microscopy of cleared samples with preserved 3D structure confirmed elevated expression of eNOS at HSS regions of the outer curvature (figure 1A; N=5 WT; P Conflict of Interest Atherosclerosis
- Published
- 2021
21. High-resolution and sensitivity bi-directional x-ray phase contrast imaging using 2D Talbot array illuminators
- Author
-
Christian Petrich, Pierre Thibault, Alex Gustschin, Jörg U. Hammel, Madleen Busse, Julia Herzen, Mirko Riedel, Wolfgang Noichl, Wolfgang Gottwald, Sheila E. Francis, Julian Moosmann, Felix Beckmann, Kirsten Taphorn, Gustschin, Alex, Riedel, Mirko, Taphorn, Kirsten, Petrich, Christian, Gottwald, Wolfgang, Noichl, Wolfgang, Busse, Madleen, Francis, Sheila E., Beckmann, Felix, Hammel, J??rg U., Moosmann, Julian, Thibault, Pierre, and Herzen, Julia
- Subjects
Materials science ,differential X-ray phase contrast ,business.industry ,High resolution ,02 engineering and technology ,021001 nanoscience & nanotechnology ,01 natural sciences ,Atomic and Molecular Physics, and Optics ,Electronic, Optical and Magnetic Materials ,010309 optics ,X-ray ,Optics ,X-ray phase contrast ,2D Talbot array ,X-Ray Phase-Contrast Imaging ,0103 physical sciences ,Sensitivity (control systems) ,ddc:620 ,0210 nano-technology ,business - Abstract
Optica 8(12), 1588 - 1595 (2021). doi:10.1364/OPTICA.441004, Two-dimensional (2D) Talbot array illuminators (TAIs) were designed, fabricated, and evaluated for high-resolution high-contrast x-ray phase imaging of soft tissue at 10���20 keV. The TAIs create intensity modulations with a high compression ratio on the micrometer scale at short propagation distances. Their performance was compared with various other wavefront markers in terms of period, visibility, flux efficiency, and flexibility to be adapted for limited beam coherence and detector resolution. Differential x-ray phase contrast and dark-field imaging were demonstrated with a one-dimensional, linear phase stepping approach yielding 2D phase sensitivity using unified modulated pattern analysis (UMPA) for phase retrieval. The method was employed for x-ray phase computed tomography reaching a resolution of 3 ��m on an unstained murine artery. It opens new possibilities for three-dimensional, non-destructive, and quantitative imaging of soft matter such as virtual histology. The phase modulators can also be used for various other x-ray applications such as dynamic phase imaging, super-resolution structured illumination microscopy, or wavefront sensing., Published by OSA, Washington, DC
- Published
- 2021
22. Preclinical models of disease and multimorbidity with focus upon cardiovascular disease and dementia
- Author
-
Jason Berwick, Manmohi D Dake, Tobias Moll, Beth Eyre, Osman Shabir, Sheila E. Francis, and Martyna M. Matuszyk
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Aging ,Drug Evaluation, Preclinical ,Context (language use) ,Disease ,03 medical and health sciences ,0302 clinical medicine ,Metabolic Diseases ,Diabetes mellitus ,Medicine ,Dementia ,Animals ,Humans ,Amyotrophic lateral sclerosis ,Intensive care medicine ,Vascular dementia ,Stroke ,business.industry ,Multimorbidity ,Neurodegenerative Diseases ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Cardiovascular Diseases ,business ,030217 neurology & neurosurgery ,Developmental Biology ,Frontotemporal dementia - Abstract
The use of animal models is fundamental to furthering our understanding of human disease mechanisms, as well as identifying potential therapeutic targets. Diseases of ageing often involve multiple body systems; however, multi-systemic features are not fully recapitulated in the many of the animal models available. Therefore, combining pre-clinical models to better reflect the multimorbidities observed at the clinical level is critical. This review will highlight some of the key pre-clinical experimental models associated with cardiovascular (atherosclerosis, coronary heart disease), cerebrovascular (stroke, vascular dementia), metabolic (obesity, type-2 diabetes mellitus) and neurological (amyotrophic lateral sclerosis, frontotemporal dementia, Parkinson's, epilepsy) diseases, and whether these models encompass known multimorbidities. In addition to this, we discuss established pre-clinical models that combine two or more conditions, within the context of dementia.
- Published
- 2020
23. Loss of PKCα increases arterial medial calcification in a uremic mouse model of chronic kidney disease
- Author
-
Neil E. Humphreys, Keith Brennan, Philip J. Withers, Cecilia Facchi, Antony Adamson, Michael J. Sherratt, Sheila E. Francis, Julia Behnsen, Nick Ashton, Samantha Borland, and Ann E. Canfield
- Subjects
Aortic arch ,medicine.medical_specialty ,Vascular smooth muscle ,business.industry ,medicine.disease ,Aortic arches ,medicine.anatomical_structure ,Endocrinology ,In vivo ,Internal medicine ,medicine.artery ,medicine ,business ,Blood urea nitrogen ,Immunostaining ,Calcification ,Kidney disease - Abstract
Arterial medial calcification is an independent risk factor for mortality in chronic kidney disease. We previously reported that knock-down of PKCα expression increases high phosphate-induced mineral deposition by vascular smooth muscle cellsin vitro. This new study tests the hypothesis that PKCα regulates uremia-induced medial calcificationin vivo. Female wild-type and PKCα−/−mice underwent a two-stage subtotal nephrectomy and were fed a high phosphate diet for 8 weeks. X-ray micro computed tomography demonstrated that uremia-induced medial calcification was increased in the abdominal aorta and aortic arch of PKCα−/−mice compared to wild-types. Blood urea nitrogen was also increased in PKCα−/−mice compared to wild-types; there was no correlation between blood urea nitrogen and calcification in PKCα−/−mice. Phosphorylated SMAD2 immunostaining was detected in calcified aortic arches from uremic PKCα−/−mice; the osteogenic marker Runx2 was also detected in these areas. No phosphorylated SMAD2 staining were detected in calcified arches from uremic wild-types. PKCα knock-down increased TGF-β1-induced SMAD2 phosphorylation in vascular smooth muscle cellsin vitro, whereas the PKCα activator prostratin decreased SMAD2 phosphorylation. In conclusion, loss of PKCα increases uremia-induced medial calcification. The PKCα/TGF-β signaling axis could therefore represent a new therapeutic target for arterial medial calcification in chronic kidney disease.
- Published
- 2020
24. JAG1-NOTCH4 Mechanosensing Drives Atherosclerosis
- Author
-
Hannah Roddie, Victoria Ridger, Blanca Tardajos Ayllon, Jovana Serbanovic-Canic, Emily V. Chambers, Sheila E. Francis, Mark P. Ariaans, Paul C. Evans, Mark J Dunning, Sarah De Val, Jin Li, Celine Souilhol, Maria Fragiadaki, Yun Fang, Daniela Pirri, Lindsay Canham, Timothy James Chico, and Xiuying Li
- Subjects
Endothelial stem cell ,JAG1 ,medicine.anatomical_structure ,Downregulation and upregulation ,Chemistry ,medicine ,Functional studies ,Therapeutic targeting ,medicine.disease ,Receptor ,Stroke ,Artery ,Cell biology - Abstract
Endothelial cell (EC) sensing of fluid shear stress regulates atherosclerosis, a disease of arteries that causes heart attack and stroke. Atherosclerosis preferentially develops at regions of arteries exposed to low oscillatory shear stress (LOSS), whereas high shear regions are protected. We show using inducible EC-specific genetic deletion in hyperlipidaemic mice that the Notch ligands JAG1 and DLL4 have opposing roles in atherosclerosis. While endothelial Jag1 promoted atherosclerosis at sites of LOSS, endothelial Dll4 was atheroprotective. Analysis of porcine and murine arteries and cultured human coronary artery EC exposed to experimental flow revealed that JAG1 and its receptor NOTCH4 are strongly upregulated by LOSS. Functional studies in cultured cells and in mice with EC-specific deletion of Jag1 show that JAG1-NOTCH4 signalling drives vascular dysfunction by repressing endothelial repair. These data demonstrate a fundamental role for JAG1-NOTCH4 in sensing LOSS during disease, and suggest therapeutic targeting of this pathway to treat atherosclerosis.
- Published
- 2020
25. Enhanced Cerebral Blood Volume under Normobaric Hyperoxia in the J20-hAPP Mouse Model of Alzheimer’s Disease
- Author
-
Luke Boorman, Stephen B. Wharton, Clare Howarth, Osman Shabir, Sheila E. Francis, Jason Berwick, Monica Alejandra Rebollar, and Paul S. Sharp
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Heterozygote ,lcsh:Medicine ,Hemodynamics ,Blood volume ,Sensory system ,Mice, Transgenic ,Disease ,Hyperoxia ,Hippocampus ,Article ,Normobaric hyperoxia ,03 medical and health sciences ,Amyloid beta-Protein Precursor ,Mice ,0302 clinical medicine ,Alzheimer Disease ,medicine ,Animals ,Cerebral Blood Volume ,lcsh:Science ,030304 developmental biology ,Neurons ,0303 health sciences ,Multidisciplinary ,business.industry ,lcsh:R ,Neuro-vascular interactions ,Brain ,Neurovascular bundle ,Immunohistochemistry ,Mice, Inbred C57BL ,Oxygen ,Disease Models, Animal ,Cerebral blood volume ,Cerebral blood flow ,Diseases of the nervous system ,lcsh:Q ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Early impairments to neurovascular coupling have been proposed to be a key pathogenic factor in the onset and progression of Alzheimer’s disease (AD). Studies have shown impaired neurovascular function in several mouse models of AD, including the J20-hAPP mouse. In this study, we aimed to investigate early neurovascular changes using wild-type (WT) controls and J20-hAPP mice at 6-9 months of age, by measuring cerebral haemodynamics and neural activity to physiological sensory stimulations. A thinned cranial window was prepared to allow access to cortical vasculature and imaged using 2D-optical imaging spectroscopy (2D-OIS). After chronic imaging sessions where the skull was intact, a terminal acute imaging session was performed where an electrode was inserted into the brain to record simultaneous neural activity. We found that cerebral haemodynamic changes were significantly enhanced in J20-hAPP mice compared with controls in response to physiological stimulations, potentially due to the significantly higher neural activity (hyperexcitability) seen in the J20-hAPP mice. Thus, neurovascular coupling remained preserved under a chronic imaging preparation. Further, under hyperoxia, the baseline blood volume and saturation of all vascular compartments in the brains of J20-hAPP mice were substantially enhanced compared to WT controls, but this effect disappeared under normoxic conditions. This study highlights novel findings not previously seen in the J20-hAPP mouse model, and may point towards a potential therapeutic strategy by driving an increased baseline blood flow to the brain, thereby potentially enhancing the clearance of beta-amyloid.
- Published
- 2020
26. The Anti-Osteoprotegerin (OPG) Antibody Ky3 Attenuates OPG-Fas Mediated Pulmonary Artery Smooth Muscle Cell Proliferation, Migration and Pro-Inflammatory Signalling In Vitro and In Vivo
- Author
-
Laura West, Josephine A. Pickworth, J. Papworth, Nadine Arnold, Allan Lawrie, Volker Germaschewski, Sheila E. Francis, and Alfred A.R. Thompson
- Subjects
biology ,Cell growth ,Chemistry ,In vitro ,Signalling ,Osteoprotegerin ,Smooth muscle ,In vivo ,medicine.artery ,Pulmonary artery ,Cancer research ,medicine ,biology.protein ,Antibody - Published
- 2020
27. Neurovascular Function in a Novel Model of Experimental Atherosclerosis
- Author
-
Monica Alejandra Rebollar, Clare Howarth, Sheila E. Francis, Paul R. Heath, Ben Pendry, Jason Berwick, Osman Shabir, and Stephen B. Wharton
- Subjects
0303 health sciences ,medicine.medical_specialty ,biology ,business.industry ,Hemodynamics ,Blood volume ,medicine.disease ,Neurovascular bundle ,biology.organism_classification ,Cortex (botany) ,Astrogliosis ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Downregulation and upregulation ,Enos ,Internal medicine ,medicine ,medicine.symptom ,business ,Hypercapnia ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
ObjectiveAtherosclerosis is a major risk factor for dementia. The aims of this study were to determine if experimental atherosclerosis leads to altered neurovascular function and causes neurovascular damage.Approach and ResultsWe analysed cerebral blood volume in male C57BL6/J mice injected with an adeno-associated virus (AAV) vector for mutated proprotein convertase subtilisin/kexin type 9 (PCSK9D377Y) fed a Western diet for 35 weeks to induce atherosclerosis (ATH) and 9-12m male wild-type (WT) C57BL/6J. We imaged blood volume responses to sensory stimulation and vascular reactivity gas challenges in the cortex of the brain through a thinned cranial window using 2D-optical imaging spectroscopy (2D-OIS). Neural activity was also recorded with multi-channel electrodes. Stimulation-evoked cortical haemodynamics, in terms of cerebral blood volume, were significantly reduced in ATH mice compared to WT and evoked neural activity was also significantly lower. However, vascular reactivity as assessed by 10% hypercapnia, remained intact in ATH mice. Immunohistochemistry in ATH mice revealed a reduced number of cortical neurons and pericytes in the cortex, but increased astrogliosis. qRT-PCR revealed significantly enhanced TNFα & IL1β in ATH mice compared to WT as well as significant upregulation of eNOS.ConclusionSystemic atherosclerosis causes significant neurovascular decline by 9m in atherosclerotic mice characterised by reduced neural activity, associated with loss of neurons and subsequent reduced cortical haemodynamics in response to physiological stimulations. The altered neurovascular function in ATH mice is chiefly mediated by TNFα.HighlightsSystemic atherosclerosis leads to significantly reduced stimulus-evoked hemodynamic responses in the cortex by 9m of age in the rAAV8-mPCSK9-D377Y mouse model of atherosclerosis compared to wild-type controls.Reduced cerebral haemodynamics are related to reduced neural activity in the cortex that could be due to a loss of cortical neurons potentially caused by significant TNFa-mediated neuroinflammation.
- Published
- 2020
- Full Text
- View/download PDF
28. Neutrophil microvesicles drive atherosclerosis by delivering miR-155 to atheroprone endothelium
- Author
-
Victoria Ridger, Chiara Recarti, Marwa Mahmoud, Andreas Schober, Amanda Burnett, Merete Long, Ingrid Gomez, Ben Ward, Jessica Johnston, Marc A. M. J. van Zandvoort, Sheila E. Francis, Siôn A Parry, Paul G. Hellewell, Endre Kiss-Toth, Mark P. Ariaans, Celine Souilhol, Le Anh Luong, Rohit Bazaz, Laura West, Rachel M Woods, Birke J. Benedikter, Chiara Niespolo, Paul C. Evans, Carl J. Hulston, Med Microbiol, Infect Dis & Infect Prev, RS: NUTRIM - R2 - Liver and digestive health, RS: Carim - B06 Imaging, and Moleculaire Celbiologie
- Subjects
0301 basic medicine ,Mice, Knockout, ApoE ,Neutrophils ,General Physics and Astronomy ,030204 cardiovascular system & hematology ,PATHWAY ,Pathogenesis ,Mice ,0302 clinical medicine ,Macrophage ,Platelet ,Lymphocytes ,lcsh:Science ,Multidisciplinary ,Smooth-muscle-cells ,NF-kappa B ,ASSOCIATION ,Plaque, Atherosclerotic ,3. Good health ,FACTOR-KAPPA-B ,medicine.anatomical_structure ,miRNAs ,medicine.symptom ,EXPRESSION ,Adhesion molecule ,Endothelium ,Cell-derived microparticles ,Science ,Activation ,Inflammation ,Diet, High-Fat ,Article ,General Biochemistry, Genetics and Molecular Biology ,miR-155 ,03 medical and health sciences ,microRNA ,medicine ,Animals ,Humans ,business.industry ,Macrophages ,Endothelial Cells ,General Chemistry ,Atherosclerosis ,Microvesicles ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,Gene Expression Regulation ,Cancer research ,lcsh:Q ,business - Abstract
Neutrophils are implicated in the pathogenesis of atherosclerosis but are seldom detected in atherosclerotic plaques. We investigated whether neutrophil-derived microvesicles may influence arterial pathophysiology. Here we report that levels of circulating neutrophil microvesicles are enhanced by exposure to a high fat diet, a known risk factor for atherosclerosis. Neutrophil microvesicles accumulate at disease-prone regions of arteries exposed to disturbed flow patterns, and promote vascular inflammation and atherosclerosis in a murine model. Using cultured endothelial cells exposed to disturbed flow, we demonstrate that neutrophil microvesicles promote inflammatory gene expression by delivering miR-155, enhancing NF-κB activation. Similarly, neutrophil microvesicles increase miR-155 and enhance NF-κB at disease-prone sites of disturbed flow in vivo. Enhancement of atherosclerotic plaque formation and increase in macrophage content by neutrophil microvesicles is dependent on miR-155. We conclude that neutrophils contribute to vascular inflammation and atherogenesis through delivery of microvesicles carrying miR-155 to disease-prone regions., The role of neutrophils in the development of atherosclerosis has long been an enigma, with few neutrophils detected within the plaque. Here, the authors show that microvesicles released from neutrophils increase vascular inflammation and enhance atherosclerotic plaque formation through delivery of miR-155.
- Published
- 2020
29. Effect of Hypoglycemia on Inflammatory Responses and the Response to Low-Dose Endotoxemia in Humans
- Author
-
Linda J Kay, Ahmed Iqbal, Simon Heller, Alan Bernjak, Peter Novodvorsky, Richard Jacques, Rory J. McCrimmon, Ian Sabroe, Lynne R. Prince, Fiona Wright, Danielle Lambert, Lewis Birch, Mark R Thomas, Sheila E. Francis, Robert F. Storey, and Ian A. Macdonald
- Subjects
Lipopolysaccharides ,Male ,Platelet Aggregation ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,030204 cardiovascular system & hematology ,Biochemistry ,Monocytes ,0302 clinical medicine ,Endocrinology ,Insulin ,Platelet ,Prospective Studies ,Healthy Volunteers ,3. Good health ,medicine.anatomical_structure ,Injections, Intravenous ,Female ,medicine.symptom ,Adult ,Agonist ,medicine.medical_specialty ,Diabetes, Pancreatic and Gastrointestinal Hormones ,medicine.drug_class ,Dose-Response Relationship, Immunologic ,030209 endocrinology & metabolism ,Context (language use) ,Inflammation ,Hypoglycemia ,Proinflammatory cytokine ,Young Adult ,03 medical and health sciences ,Internal medicine ,Diabetes mellitus ,Escherichia coli ,medicine ,Humans ,Clinical Research Articles ,business.industry ,Monocyte ,Biochemistry (medical) ,medicine.disease ,Endotoxemia ,Immunity, Innate ,Glucose ,Human Experimentation ,Hyperglycemia ,Glucose Clamp Technique ,business - Abstract
Context Hypoglycemia is emerging as a risk for cardiovascular events in diabetes. We hypothesized that hypoglycemia activates the innate immune system, which is known to increase cardiovascular risk. Objective To determine whether hypoglycemia modifies subsequent innate immune system responses. Design and Setting Single-blinded, prospective study of three independent parallel groups. Participants and Interventions Twenty-four healthy participants underwent either a hyperinsulinemic-hypoglycemic (2.5 mmol/L), euglycemic (6.0 mmol/L), or sham-saline clamp (n = 8 for each group). After 48 hours, all participants received low-dose (0.3 ng/kg) intravenous endotoxin. Main Outcome Measures We studied in-vivo monocyte mobilization and monocyte-platelet interactions. Results Hypoglycemia increased total leukocytes (9.98 ± 1.14 × 109/L vs euglycemia 4.38 ± 0.53 × 109/L, P < 0.001; vs sham-saline 4.76 ± 0.36 × 109/L, P < 0.001) (mean ± SEM), mobilized proinflammatory intermediate monocytes (42.20 ± 7.52/μL vs euglycemia 20.66 ± 3.43/μL, P < 0.01; vs sham-saline 26.20 ± 3.86/μL, P < 0.05), and nonclassic monocytes (36.16 ± 4.66/μL vs euglycemia 12.72 ± 2.42/μL, P < 0.001; vs sham-saline 19.05 ± 3.81/μL, P < 0.001). Following hypoglycemia vs euglycemia, platelet aggregation to agonist (area under the curve) increased (73.87 ± 7.30 vs 52.50 ± 4.04, P < 0.05) and formation of monocyte-platelet aggregates increased (96.05 ± 14.51/μL vs 49.32 ± 6.41/μL, P < 0.05). Within monocyte subsets, hypoglycemia increased aggregation of intermediate monocytes (10.51 ± 1.42/μL vs euglycemia 4.19 ± 1.08/μL, P < 0.05; vs sham-saline 3.81± 1.42/μL, P < 0.05) and nonclassic monocytes (9.53 ± 1.08/μL vs euglycemia 2.86 ± 0.72/μL, P < 0.01; vs sham-saline 3.08 ± 1.01/μL, P < 0.05), with platelets compared with controls. Hypoglycemia led to greater leukocyte mobilization in response to subsequent low-dose endotoxin challenge (10.96 ± 0.97 vs euglycemia 8.21 ± 0.85 × 109/L, P < 0.05). Conclusions Hypoglycemia mobilizes monocytes, increases platelet reactivity, promotes interaction between platelets and proinflammatory monocytes, and potentiates the subsequent immune response to endotoxin. These changes may contribute to increased cardiovascular risk observed in people with diabetes., Using an in vivo human experimental model, we show that hypoglycemia primes the innate immune system, leading to a more profound inflammatory response to a subsequent inflammatory stimulus.
- Published
- 2018
30. Differential IL-1 signaling induced by BMPR2 deficiency drives pulmonary vascular remodeling
- Author
-
Sheila Shay, James Iremonger, Peter M. Hickey, Josephine A. Pickworth, Helen Casbolt, Allan Lawrie, Nicholas W. Morrell, Kay Hopkinson, Sheila E. Francis, James West, Alexander M.K. Rothman, Santhi Gladson, Morrell, Nicholas [0000-0001-5700-9792], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,lcsh:Diseases of the circulatory (Cardiovascular) system ,Inflammation ,Vascular remodelling in the embryo ,03 medical and health sciences ,White blood cell ,Internal medicine ,medicine.artery ,pulmonary hypertension ,medicine ,Research Articles ,lcsh:RC705-779 ,Lung ,business.industry ,BMPR-II ,lcsh:Diseases of the respiratory system ,medicine.disease ,Pulmonary hypertension ,BMPR2 ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Cell culture ,lcsh:RC666-701 ,inflammation ,Pulmonary artery ,interleukin-1ß ,medicine.symptom ,business - Abstract
Background: Bone morphogenetic protein receptor type 2 (BMPR2) mutations are present in patients with heritable and idiopathic pulmonary arterial hypertension (PAH). Circulating levels of Interleukin-1 (IL-1) are raised in patients and animal models. Whether interplay between BMP and IL- 1 signalling can explain the local manifestation of PAH in the lung remains unclear. Methods: Cell culture, siRNA and mRNA microarray analysis of RNA isolated from human Pulmonary artery (PASMC) and Aortic (AoSMC) smooth muscle cells were used. R899X+/- BMPR2 transgenic mice fed western diet for six weeks were given daily injections of IL-1s prior to assessment for PAH and tissue collection. Results: PASMC have reduced inflammatory activation in response to IL-1s compared with AoSMCs, however PASMC with reduced BMPR2 demonstrated an exaggerated response. Mice treated with IL-1s had higher white blood cell counts, and significantly raised serum protein levels of IL-6 and OPG plasma levels recapitulating in vitro data. Phenotypically, IL-1s treated mice demonstrated increased pulmonary vascular remodelling. Conclusions: IL-1s induces an exaggerated pulmonary artery specific transcriptomic inflammatory response when BMPR2 signalling is reduced.
- Published
- 2017
31. Transforming growth factor-beta signaling via ALK1 and ALK5 regulates distinct functional pathways in vein graft intimal hyperplasia
- Author
-
Martin W. McBride, David J. Kelly, P. ten Dijke, Nicholas W. Morrell, Angela C. Bradshaw, Christian Delles, S. Arias-Rivas, M. Pek, A. Shaw, Stuart A. Nicklin, Emma L. Low, Andy Baker, John D. McClure, J. T. Schwartze, Menzo J. E. Havenga, Adam Kurkiewicz, Sheila E. Francis, Pawel Herzyk, and Midory Thorikay
- Subjects
Neointima ,Intimal hyperplasia ,biology ,Chemistry ,Cell migration ,Transforming growth factor beta ,medicine.disease ,Cell biology ,Transactivation ,Fibrosis ,medicine ,biology.protein ,Beta (finance) ,Transforming growth factor - Abstract
RationaleTransforming growth factor-beta (TGFβ) is tightly regulated at multiple levels, with regulation at the receptor level now recognized as a key determinant of the cellular response to this pleiotropic cytokine. TGFβ promotes saphenous vein graft neointima formation after coronary artery bypass graft (CABG) surgery, inducing smooth muscle cell (SMC) hyperplasia and fibrosis by signaling via activin receptor-like kinase 5(ALK5). However, the role of the alternate TGFβ receptor ALK1 remains completely unknown.ObjectiveTo define the receptor pathways activated by TGFβ in SMCs and their mechanistic importance during CABG neointima formation.Methods and resultsRadioligand co-IP assays revealed direct interactions between TGFβ, ALK5 and ALK1 in primary saphenous vein graft SMC (HSVSMC) from patients undergoing CABG. Knockdown and pharmacological inhibition of ALK5 or ALK1 in HSVSMC significantly attenuated TGFβ-induced phosphorylation of receptor-regulated (R)-Smads 2/3 and 1/5, respectively. Microarray profiling followed by qRT-PCR validation showed that TGFβ induced distinct transcriptional networks downstream of ALK5 or ALK1, associated with HSVSMC contractility and migration, respectively and confirmed using migration assays as well as qRT-PCR and western blot assays of contractile SMC markers. scRNAseq analysis of TGFβ-treated HSVSMC identified distinct subgroups of cells showing ALK5 or ALK1 transcriptional responses, while RNA velocity analyses indicated divergence in differentiation towards ALK5 or ALK1-dominant lineages. ALK1, ALK5 and their downstream effectors pSmad1/5 and pSmad2/3 were localized to αSMA+ neointimal SMCs in remodelled mouse vein grafts. Pharmacological inhibition or genetic ablation of Smad1/5 substantially reducing neointima formation following acute vascular injury. Notably, expression and activation of ALK1, ALK5 and their respective downstream R-Smads was already evident in hyperplastic saphenous veins prior to grafting.ConclusionsWhilst canonical TGFβ signaling via ALK5 promotes a contractile HSVSMC phenotype, transactivation of ALK1 by TGFβ induces neointima formation by driving cell migration. Restoring the balance between ALK1 and ALK5 in HSVSMC may represent a novel therapeutic strategy for vein graft failure.
- Published
- 2019
32. Transfer of complex regional pain syndrome to mice via human autoantibodies is mediated by interleukin-1–induced mechanisms
- Author
-
Andreas Goebel, Tamas Kiss, Nikolett Szentes, Emmanuel Pinteaux, Krisztina Tóth, Sheila E. Francis, Hajnalka Ábrahám, Krisztina Pohóczky, Ágnes Kemény, Nikolett Lénárt, Bálint Botz, Adam Denes, Valéria Tékus, Zsuzsanna Helyes, Serena Sensi, and Zsuzsanna Környei
- Subjects
Inflammation ,CRPS ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Sensitization ,030304 developmental biology ,0303 health sciences ,Anakinra ,Multidisciplinary ,business.industry ,complex regional pain syndrome ,Interleukin ,medicine.disease ,3. Good health ,Complex regional pain syndrome ,medicine.anatomical_structure ,Immunology ,Hyperalgesia ,Prednisolone ,medicine.symptom ,business ,autoantibody ,interleukin-1 ,030217 neurology & neurosurgery ,Glucocorticoid ,anakinra ,medicine.drug - Abstract
Neuroimmune interactions may contribute to severe pain and regional inflammatory and autonomic signs in complex regional pain syndrome (CRPS), a posttraumatic pain disorder. Here, we investigated peripheral and central immune mechanisms in a translational passive transfer trauma mouse model of CRPS. Small plantar skin-muscle incision was performed in female C57BL/6 mice treated daily with purified serum immunoglobulin G (IgG) from patients with longstanding CRPS or healthy volunteers followed by assessment of paw edema, hyperalgesia, inflammation, and central glial activation. CRPS IgG significantly increased and prolonged swelling and induced stable hyperalgesia of the incised paw compared with IgG from healthy controls. After a short-lasting paw inflammatory response in all groups, CRPS IgG-injected mice displayed sustained, profound microglia and astrocyte activation in the dorsal horn of the spinal cord and pain-related brain regions, indicating central sensitization. Genetic deletion of interleukin-1 (IL-1) using IL-1αβ knockout (KO) mice and perioperative IL-1 receptor type 1 (IL-1R1) blockade with the drug anakinra, but not treatment with the glucocorticoid prednisolone, prevented these changes. Anakinra treatment also reversed the established sensitization phenotype when initiated 8 days after incision. Furthermore, with the generation of an IL-1β floxed(fl/fl) mouse line, we demonstrated that CRPS IgG-induced changes are in part mediated by microglia-derived IL-1β, suggesting that both peripheral and central inflammatory mechanisms contribute to the transferred disease phenotype. These results indicate that persistent CRPS is often contributed to by autoantibodies and highlight a potential therapeutic use for clinically licensed antagonists, such as anakinra, to prevent or treat CRPS via blocking IL-1 actions.
- Published
- 2019
33. Myeloid Tribbles 1 induces early atherosclerosis via enhanced foam cell expansion
- Author
-
Sheila E. Francis, Adrienn Angyal, Robert C. Bauer, S. Kim Suvarna, T. Neil Dear, Jessica Johnston, Endre Kiss-Toth, Stephen E. Hamby, Zoltan Hegedus, Alison H. Goodall, Martin R Turner, Kajus Baidžajevas, Heather L. Wilson, Carol C. Shoulders, Daniel J. Rader, Johnston, Jessica M [0000-0003-4877-0680], Angyal, Adrienn [0000-0001-8483-1749], Bauer, Robert C [0000-0001-9752-9645], Baidžajevas, Kajus [0000-0002-8133-2861], Turner, Martin [0000-0002-3801-9896], Wilson, Heather L [0000-0002-7892-3425], Goodall, Alison H [0000-0002-7883-9645], Rader, Daniel J [0000-0002-9245-9876], Shoulders, Carol C [0000-0003-2218-1095], Francis, Sheila E [0000-0001-6552-0339], Kiss-Toth, Endre [0000-0003-4406-4017], and Apollo - University of Cambridge Repository
- Subjects
Male ,Myeloid ,Transgene ,Diseases and Disorders ,Protein Serine-Threonine Kinases ,030204 cardiovascular system & hematology ,Models, Biological ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genetics ,OLR1 ,medicine ,Animals ,Humans ,Myeloid Cells ,Health and Medicine ,Scavenger receptor ,Receptor ,Research Articles ,030304 developmental biology ,Foam cell ,0303 health sciences ,Multidisciplinary ,Triglyceride ,Chemistry ,Intracellular Signaling Peptides and Proteins ,SciAdv r-articles ,Middle Aged ,Atherosclerosis ,Scavenger Receptors, Class E ,medicine.disease ,Phenotype ,Plaque, Atherosclerotic ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,Cholesterol ,medicine.anatomical_structure ,Atheroma ,Low-density lipoprotein ,Cancer research ,Lipoprotein ,Research Article ,Foam Cells - Abstract
Trib1 controls atherosclerotic plaque macrophage function by up-regulating OLR1, promoting foam cell formation and atherosclerosis., Macrophages drive atherosclerotic plaque progression and rupture; hence, attenuating their atherosclerosis-inducing properties holds promise for reducing coronary heart disease (CHD). Recent studies in mouse models have demonstrated that Tribbles 1 (Trib1) regulates macrophage phenotype and shows that Trib1 deficiency increases plasma cholesterol and triglyceride levels, suggesting that reduced TRIB1 expression mediates the strong genetic association between the TRIB1 locus and increased CHD risk in man. However, we report here that myeloid-specific Trib1 (mTrib1) deficiency reduces early atheroma formation and that mTrib1 transgene expression increases atherogenesis. Mechanistically, mTrib1 increased macrophage lipid accumulation and the expression of a critical receptor (OLR1), promoting oxidized low-density lipoprotein uptake and the formation of lipid-laden foam cells. As TRIB1 and OLR1 RNA levels were also strongly correlated in human macrophages, we suggest that a conserved, TRIB1-mediated mechanism drives foam cell formation in atherosclerotic plaque and that inhibiting mTRIB1 could be used therapeutically to reduce CHD.
- Published
- 2019
- Full Text
- View/download PDF
34. BS61 PKCαα knock-down enhances intimal calcification in a murine model of atherosclerosis
- Author
-
Cathy M. Holt, Sheila E. Francis, Julia Behnsen, Keith Brennan, Michael J. Sherratt, Ann E. Canfield, Samantha Borland, and Abimbola Akerele
- Subjects
medicine.medical_specialty ,Vascular smooth muscle ,business.industry ,medicine.disease ,In vitro ,Lesion ,chemistry.chemical_compound ,Endocrinology ,chemistry ,In vivo ,Internal medicine ,Medicine ,Oil Red O ,medicine.symptom ,business ,Protein kinase A ,Transforming growth factor ,Calcification - Abstract
Intimal calcification is the formation of mineralised tissue within atherosclerotic lesions and can lead to an increased risk of plaque rupture and mortality in man. Even though experimental data are sparse, it had been suggested that inhibiting protein kinase Cα (PKCα) may be of therapeutic benefit in atherosclerosis. However, we recently discovered that loss of PKCα increases mineral deposition by vascular smooth muscle cells (VSMCs) in a transforming growth factor-β (TGFβ)-dependent manner in vitro, suggesting that vascular calcification and its devastating consequences could be increased if patients with cardiovascular disease are treated with PKCα inhibitors. This study tests the hypothesis that PKCα regulates atherosclerosis and intimal calcification in vivo. Nine to ten week-old male PKCα knock-out (PKCα-/-) mice crossed with ApoE-/- mice (PKCα-/-ApoE-/-) were fed a high-fat, high-cholesterol diet (Western diet) for 8, 18 and 28 weeks. Male ApoE-/- mice fed the same diet were used as controls. En face Oil Red O staining was significantly increased (∼1.5-fold, P Aortic sinus lesion size was not significantly different between PKCα-/-ApoE-/- and ApoE-/- mice at any time point. However, a significant increase in intimal calcification (∼2-fold, P In conclusion, our study has identified that PKCα may play a protective role in both atherosclerosis and intimal calcification. This suggests that inhibiting PKCα may not be of any therapeutic benefit in atherosclerosis. Activation of PKCα could instead represent a new therapeutic target for atherosclerosis-induced intimal calcification. Conflict of interest None
- Published
- 2019
35. BS50 PKCα knock-down increases medial calcification in a murine model of chronic kidney disease
- Author
-
Julia Behnsen, Neil Humphrey, Keith Brennan, Cecilia Facchi, Ann E. Canfield, Antony Adamson, Samantha Borland, Michael J. Sherratt, Nick Ashton, and Sheila E. Francis
- Subjects
Aortic arch ,medicine.medical_specialty ,Vascular smooth muscle ,Kinase ,business.industry ,Abdominal aorta ,medicine.disease ,Endocrinology ,In vivo ,medicine.artery ,Internal medicine ,medicine ,business ,Protein kinase C ,Calcification ,Transforming growth factor - Abstract
Medial calcification is the formation of mineralised tissue within the smooth muscle layer of the vessel wall, and frequently occurs in patients with chronic kidney disease. Calcification within the medial layer of the vessel wall can reduce aortic and arterial elasticity, which impairs cardiovascular haemodynamics and results in a significantly elevated risk of morbidity and mortality in the form of hypertension, cardiac hypertrophy and sudden cardiac death. Protein kinase Cα (PKCα) belongs to the PKC family of serine/threonine kinases and we recently discovered that knocking-down PKCα expression increases high phosphate-induced mineral deposition by vascular smooth muscle cells (VSMCs) in vitro. This study tests the hypothesis that PKCα regulates uraemia-induced medial calcification in vivo. PKCα-/- mice were generated on the calcification-susceptible DBA/2 background (PKCα-/-) using CRISPR/Cas9 technology. To induce uraemia, wild-type DBA/2 and PKCα-/- mice underwent a two-stage sub-total nephrectomy and were fed a high phosphate (1.5%) diet for 8 weeks. Renal function was measured by blood urea nitrogen (BUN). Calcification in the ascending aorta/aortic arch and abdominal aorta were analysed and quantified by micro CT and histology. On average, 68.6 ± 3% (SD, n=3) of renal mass was removed from wild-type and 65 ± 3.2% (n=5) was removed from PKCα-/- mice (P>0.05). Loss of PKCα significantly increased uraemia-induced medial calcification in the abdominal aorta (∼20-fold increase, P We have shown previously that inhibiting transforming growth factor-β (TGF-β) signalling with SB431542 prevents the increase in calcification observed in PKCα-siRNA treated VSMCs. Therefore, to determine the mechanism by which loss of PKCα exerts its effects we examined the relationship between PKCα and TGF-β signalling in vitro and in vivo. Our results show that knock-down of PKCα using siRNA increased TGF-β1-induced Smad2 phosphorylation in VSMCs in vitro (P In conclusion, our study suggests that PKCα may play a protective role in uraemia-induced medial calcification. The PKCα/TGFβ signalling axis could therefore represent a new therapeutic target for uraemia-induced medial calcification. Conflict of interest None
- Published
- 2019
36. BS22 Double positive (CD86+ MRC1+) inflammatory macrophages in the pathogenesis of carotid atherosclerosis
- Author
-
Arshad Majid, Sheila E. Francis, Robert Lonsdale, Endre Kiss-Toth, Jessica Redgrave, and Klaudia Kocsy
- Subjects
education.field_of_study ,Pathology ,medicine.medical_specialty ,CD68 ,business.industry ,medicine.medical_treatment ,Population ,Fibrous cap ,Double negative ,Carotid endarterectomy ,Proinflammatory cytokine ,Pathogenesis ,medicine.anatomical_structure ,medicine.artery ,medicine ,Internal carotid artery ,education ,business - Abstract
Introduction Atherosclerosis is a complex inflammatory disease in which major arteries narrow, and an atherosclerotic plaque develops, modulated by genetic and environmental factors. Macrophages participate in all stages of the plaque formation and progression. Different macrophage subtypes within carotid plaques have been shown to be altered in unstable vs. stable plaques. M1 proinflammatory macrophages are mostly abundant in symptomatic and unstable plaques. In contrast, M2 macrophages are associated with regulatory and wound-healing properties and generally found in stable lesions. In the future, the characterisation of these changes in situ in the carotid plaques, associated with in vitro macrophage studies, and blood composition may lead to the identification of diagnostic markers to direct the best treatment for stroke patients. Methods Carotid plaques were obtained from patients with a recent stroke or TIA and with a greater than 50% stenosis of the internal carotid artery. Plaques were removed during carotid endarterectomy and fixed in 10% (v/v) neutral buffered formalin for maximum 36h. Macrophage phenotypes within the shoulder regions were characterised with immunofluorescence microscopy. A novel immunofluorescence staining, image acquisition and analysis technique was developed to identify single M1 (CD68+CD86+MRC1-), single M2 (CD68+CD86-MRC1+), double positive (CD68+CD86+MRC1+) and double negative (CD68+CD86-MRC1-) macrophages in the human carotid atherosclerotic plaques. Results 20 carotid plaques were collected from patients with recent stroke or TIA (73% male, 70% smokers, 87% hypertensive). In 17 plaques the shoulder region (either side of a thinned fibrous cap) was sufficiently intact for histological analysis. Dense infiltration with macrophages was seen in all 17 specimens. The population comprised double positive (CD86+MRC1+) (71%), M1 single positive (CD86+MRC1-) (19%), and M2 single positive (CD86-MRC1+) (4%). Double negative (CD86-MRC1-) macrophages represented 6% of all macrophages. Amongst 10 unstable plaques (defined as AHA Grade 6 or above) the majority (80%) of shoulder macrophages were M1 single positive. In contrast, amongst 7 stable plaques, the majority of plaque shoulder macrophages (65%) were M2 single positive. Conclusions Macrophages within symptomatic carotid plaques can express both M1 (CD86) and M2 (MRC1) markers, or can express neither. Pro-inflammatory macrophage phenotypes cluster in areas of unstable carotid plaque. Larger studies are needed to confirm the role of dual staining macrophages and double negative macrophages. If such in situ macrophages can be manipulated towards one phenotype with pharmaceutical interventions, this could offer a potential new approach to plaque stabilising therapies. Conflict of interest No
- Published
- 2019
37. Neurovascular dysfunction in vascular dementia, Alzheimer’s and atherosclerosis
- Author
-
Jason Berwick, Sheila E. Francis, and Osman Shabir
- Subjects
0301 basic medicine ,Mouse ,Neuroimaging ,Review ,Disease ,Vascular dementia ,lcsh:RC321-571 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Alzheimer Disease ,medicine ,Animals ,Humans ,Dementia ,cardiovascular diseases ,Cognitive decline ,Stroke ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,business.industry ,Dementia, Vascular ,General Neuroscience ,lcsh:QP351-495 ,Brain ,medicine.disease ,Neurovascular bundle ,Atherosclerosis ,3. Good health ,030104 developmental biology ,lcsh:Neurophysiology and neuropsychology ,Cerebral blood flow ,Disease modelling ,business ,Neuroscience ,Neurovascular coupling ,Alzheimer’s ,030217 neurology & neurosurgery - Abstract
Efficient blood supply to the brain is of paramount importance to its normal functioning and improper blood flow can result in potentially devastating neurological consequences. Cerebral blood flow in response to neural activity is intrinsically regulated by a complex interplay between various cell types within the brain in a relationship termed neurovascular coupling. The breakdown of neurovascular coupling is evident across a wide variety of both neurological and psychiatric disorders including Alzheimer’s disease. Atherosclerosis is a chronic syndrome affecting the integrity and function of major blood vessels including those that supply the brain, and it is therefore hypothesised that atherosclerosis impairs cerebral blood flow and neurovascular coupling leading to cerebrovascular dysfunction. This review will discuss the mechanisms of neurovascular coupling in health and disease and how atherosclerosis can potentially cause cerebrovascular dysfunction that may lead to cognitive decline as well as stroke. Understanding the mechanisms of neurovascular coupling in health and disease may enable us to develop potential therapies to prevent the breakdown of neurovascular coupling in the treatment of vascular brain diseases including vascular dementia, Alzheimer’s disease and stroke.
- Published
- 2018
38. Author Correction: Shear stress induces endothelial-to-mesenchymal transition via the transcription factor Snail
- Author
-
Markus Ariaans, Sheila E. Francis, Jovana Serbanovic-Canic, Paul C. Evans, Sarah Hsiao, Celine Souilhol, Shuang Feng, Marwa Mahmoud, Rouyu Xing, Akiko Mammoto, Victoria Ridger, Jing Chen, and Kim Van der Heiden
- Subjects
Multidisciplinary ,Transition (genetics) ,biology.animal ,Science ,Mesenchymal stem cell ,Shear stress ,Medicine ,Snail ,Biology ,Transcription factor ,Cell biology - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
39. Interleukin-1β has atheroprotective effects in advanced atherosclerotic lesions of mice
- Author
-
Delphine Gomez, Gabriel F. Alencar, Richard A. Baylis, Gwendalyn J. Randolph, Gary K. Owens, Ari Waisman, Hermann Gram, Brittany G Durgin, Emmanuel Pinteaux, Sheila E. Francis, Werner Müller, Cynthia St. Hilaire, Alexandra A. C. Newman, and Sidney Mahan
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Infarction ,Down-Regulation/drug effects ,Inflammation ,030204 cardiovascular system & hematology ,Antibodies, Neutralizing/pharmacology ,General Biochemistry, Genetics and Molecular Biology ,Lesion ,Cell Proliferation/drug effects ,03 medical and health sciences ,0302 clinical medicine ,Macrophages/drug effects ,Atherosclerosis/metabolism ,Monocytes/drug effects ,Neutralization Tests ,Signal Transduction/drug effects ,medicine ,Macrophage ,Myocyte ,Animals ,Cell Polarity/drug effects ,business.industry ,Fibrous cap ,Apoptosis/drug effects ,General Medicine ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,Canakinumab ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,Interleukin-1beta/metabolism ,medicine.symptom ,business ,Interleukin 1 receptor, type I ,Myocytes, Smooth Muscle/drug effects ,medicine.drug ,Inflammation/pathology - Abstract
Despite decades of research, our understanding of the processes controlling late-stage atherosclerotic plaque stability remains poor. A prevailing hypothesis is that reducing inflammation may improve advanced plaque stability, as recently tested in the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial, in which post-myocardial infarction subjects were treated with an IL-1β antibody. Here, we performed intervention studies in which smooth muscle cell (SMC) lineage-tracing Apoe-/- mice with advanced atherosclerosis were treated with anti-IL-1β or IgG control antibodies. Surprisingly, we found that IL-1β antibody treatment between 18 and 26 weeks of Western diet feeding induced a marked reduction in SMC and collagen content, but increased macrophage numbers in the fibrous cap. Moreover, although IL-1β antibody treatment had no effect on lesion size, it completely inhibited beneficial outward remodeling. We also found that SMC-specific knockout of Il1r1 (encoding IL-1 receptor type 1) resulted in smaller lesions nearly devoid of SMCs and lacking a fibrous cap, whereas macrophage-selective loss of IL-1R1 had no effect on lesion size or composition. Taken together, these results show that IL-1β has multiple beneficial effects in late-stage murine atherosclerosis, including promotion of outward remodeling and formation and maintenance of an SMC- and collagen-rich fibrous cap.
- Published
- 2018
40. Dietary Docosahexaenoic Acid Reduces Oscillatory Wall Shear Stress, Atherosclerosis, and Hypertension, Most Likely Mediated via an IL-1-Mediated Mechanism
- Author
-
Patrick W. F. Hadoke, Alexander M.K. Rothman, Maria-Cruz Villa-Uriol, Sheila E. Francis, David C. Crossman, Junxi Wu, Janet Chamberlain, Torsten Schenkel, Mabruka Alfaidi, Paul C. Evans, University of St Andrews. School of Medicine, and University of St Andrews. Office of the Principal
- Subjects
0301 basic medicine ,Male ,Apolipoprotein B ,Mice, Knockout, ApoE ,Interleukin-1beta ,Anti-Inflammatory Agents ,030204 cardiovascular system & hematology ,Interleukin 1 ,Wall shear stress ,0302 clinical medicine ,Aorta ,Original Research ,biology ,docosahexaenoic acid ,Plaque, Atherosclerotic ,3. Good health ,Docosahexaenoic acid ,medicine.anatomical_structure ,RB Pathology ,Hypertension ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,Signal Transduction ,Cardiac function curve ,medicine.medical_specialty ,Endothelium ,Docosahexaenoic Acids ,endothelium ,NDAS ,Aortic Diseases ,Inflammation ,Diet, High-Fat ,interleukin 1 ,Proinflammatory cytokine ,03 medical and health sciences ,Internal medicine ,TA164 ,medicine ,Animals ,Arterial Pressure ,business.industry ,medicine.disease ,Atherosclerosis ,wall shear stress ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Blood pressure ,Atheroma ,Animal Models of Human Disease ,inflammation ,Growth Factors/Cytokines ,Dietary Supplements ,biology.protein ,Stress, Mechanical ,business ,RB ,Basic Science Research - Abstract
Background Hypertension is a complex condition and a common cardiovascular risk factor. Dietary docosahexaenoic acid ( DHA ) modulates atherosclerosis and hypertension, possibly via an inflammatory mechanism. IL‐1 (interleukin 1) has an established role in atherosclerosis and inflammation, although whether IL ‐1 inhibition modulates blood pressure is unclear. Methods and Results Male apoE −/− (apolipoprotein E–null) mice were fed either a high fat diet or a high fat diet plus DHA (300 mg/kg per day) for 12 weeks. Blood pressure and cardiac function were assessed, and effects of DHA on wall shear stress and atherosclerosis were determined. DHA supplementation improved left ventricular function, reduced wall shear stress and oscillatory shear at ostia in the descending aorta, and significantly lowered blood pressure compared with controls (119.5±7 versus 159.7±3 mm Hg, P DHA feeding in mice demonstrated a 4‐fold reduction in lesion burden in distal aortas and in brachiocephalic arteries ( P DHA treatment selectively decreased plaque endothelial IL ‐1β ( P Conclusions Our findings revealed that raised blood pressure can be reduced by inhibiting IL ‐1 indirectly by administration of DHA in the diet through a mechanism that involves a reduction in wall shear stress and local expression of the proinflammatory cytokine IL ‐1β.
- Published
- 2018
41. 123 Myeloid TRIB1 controls experimental atherosclerosis
- Author
-
Carol C. Shoulders, Daniel J. Rader, Jessica Johnston, Robert C. Bauer, Adrienn Angyal, Endre Kiss-Toth, and Sheila E. Francis
- Subjects
medicine.medical_specialty ,Aorta ,Myeloid ,business.industry ,Regulator ,Genome-wide association study ,medicine.anatomical_structure ,Endocrinology ,Internal medicine ,medicine.artery ,Knockout mouse ,Medicine ,Macrophage ,Bone marrow ,business ,Foam cell - Abstract
Introduction Genome Wide Association Studies have identified Tribbles-1 (TRIB1) as a regulator of plasma lipid levels and a risk factor for MI. Studies using Trib1 full body- and liver specific knockout mice have shown that hepatic expression of Trib1 reduces circulating lipids1. Additionally, Trib1 has been shown to be a regulator of alternatively activated macrophage polarisation2. However, there has been no study to directly evaluate the role of myeloid Trib1 (mTrib1) in atherogenesis. Methods To determine the role of mTrib1 in atherosclerosis, we developed myeloid-Trib1 deficient (mTrib1KO) and overexpression (mTrib1Tg) mouse strains. To distinguish between metabolic and inflammatory drivers of atherosclerosis, bone marrow from these strains were transplanted into lethally irradiated ApoE-/- mice and fed on a western diet for 12 weeks. Additionally, we also induced atherosclerosis in mTrib1Tg and mTrib1WT strains by injecting rAAV8/mPCSK9 and fed western diet for 12 weeks. Results mTrib1KO→ApoE-/- mice were protected while mTrib1Tg→ApoE-/- mice presented with increased atherosclerotic burden in both the aorta (p Conclusion We conclude that Trib1 is a potent regulator of atherosclerosis, the over-expression of which promotes atherogenesis through elevated oxLDL uptake and subsequent foam cell formation in plaque macrophages. References . Burkhardt et al. JCI (2010);120:4410–4414. . Satoh et al. Nature (2013);7442:524–8.
- Published
- 2018
42. 116 Semi-quantitative imaging of macrophages in human carotid atherosclerotic plaques
- Author
-
Sheila E. Francis, Jessica Johnston, Arshad Majid, Kevin Tang, Endre Kiss-Toth, Jessica Redgrave, Klaudia Kocsy, and Shavinthi Wadanamby
- Subjects
Pathology ,medicine.medical_specialty ,CD68 ,business.industry ,medicine.medical_treatment ,Fibrous cap ,H&E stain ,Inflammation ,Histology ,Carotid endarterectomy ,Proinflammatory cytokine ,medicine.anatomical_structure ,medicine ,medicine.symptom ,business ,Mannose receptor - Abstract
Introduction Atherosclerosis is a pathological chronic inflammatory process where lipoprotein, cholesterol, calcium and apoptotic cells build up a lipid-rich plaque within an artery. Inflammatory cells such as macrophages are a major component of the atherosclerotic plaque and participate in all stages of plaque formation and progression. The plaque can be classified as histologically stable or unstable, depending on the extent of inflammation, composition and the thickness of fibrous cap. Macrophage subtypes within carotid plaques have been shown to be altered in unstable vs. stable plaques. Broadly, M1 macrophages are proinflammatory and promote atherogenesis as well as enhancing plaque instability. In contrast, M2 macrophages have regulatory and wound-healing properties including collagen synthesis and promote tissue repair. Detailed characterisation of these macrophage subtypes and correlative analysis with in vivo carotid plaque MRI and blood composition may lead to the identification of diagnostic markers to direct the best treatment for stroke patients. Methodology Carotid plaques were obtained from patients undergoing carotid endarterectomy for recently symptomatic carotid stenosis. A validated clinical histology grading was used to define degree of overall plaque instability. The plaque ’shoulder’ regions were identified on H and E stained sections and were deemed the regions of interest (ROI). Macrophage phenotypes within the shoulder regions were characterised with immunofluorescence (IF) microscopy. An IF protocol to simultaneously stain for M1 and M2 macrophages alongside an image acquisition and analysis strategy was then developed. We used single IF with a pan macrophage polarisation marker (CD68), and dual IF with M1 (CD86) and M2 marker (MR: Mannose Receptor). Plaques were imaged using multi-colour fluorescence microscopy (Leica AF6000). After image registration, the regions of positive staining for CD68 were overlapped with the dual staining (CD86/MR) section and the prevalence of CD86 +and MR +macrophages were measured. Results Of the 16 collected plaques, an intact shoulder was identified in 13 samples and in these regions, macrophages were abundant. A median of 31.1% (4.5%–70.6%) macrophages stained for both M1 (CD86) and M2 (MR) markers. There was a higher prevalence of CD86 +MR + macrophages in shoulder regions from unstable versus stable plaques but this did not reach significance. Single positive M2 (MR +CD86-) cells were of higher prevalence in stable plaques. Hemosiderin-laden macrophages were also identified via their strong auto-fluorescence. Conclusions M1 (CD86+) and dual staining (CD86 +MR+) macrophage populations were predominant in the unstable plaques whereas single positive M2 (MR +CD86-) cells were predominant in stable plaques. Future analysis on a larger number of carotid plaques is required to better understand the prevalence and potential influence of different macrophage populations on plaque instability.
- Published
- 2018
43. 140 Acetylated ldl induces IL-1β release from human coronary endothelial cells
- Author
-
Janet Chamberlain, Majid Almansouri, and Sheila E. Francis
- Subjects
biology ,business.industry ,Cholesterol ,Molecular biology ,Pathogenesis ,chemistry.chemical_compound ,chemistry ,Neutrophil elastase ,Lactate dehydrogenase ,Toxicity ,Extracellular ,biology.protein ,Medicine ,Secretion ,Viability assay ,business - Abstract
Background Atherosclerosis is a chronic vascular inflammatory disease characterised by disturbed arterial blood flow due to the atheromatous plaque build-up within arterial layers. Lipid rich plaques contain various forms of cholesterol, such as acetylated low density lipoprotein (AcLDL), that undergo many modification processes and which enter vascular cells, initiating over exuberant repair processes leading to arterial occlusion and life threatening myocardial infarction or stroke. Endothelial cells (EC) strongly drive the pathogenesis of atherosclerosis by producing pro-inflammatory cytokines, such as interleukin-1 beta (IL-1β). We have recently shown that the release of IL-1β from EC occurs via extracellular lysosomal derived vesicles, but the stimuli and mechanisms by which IL-1β is released remain to be fully elucidated. We hypothesise that AcLDL enters the arterial endothelium and induces IL-1 secretion, potentially via a caspase-1/NLRP3 mechanism. Methods Human coronary artery endothelial cells (HCAEC) isolated from three different donors were cultured and stimulated with pro-inflammatory cytokines TNF-α and IL-1β (10 ng/ml each, for 48 hours), followed by incubation with native human AcLDL cholesterol at multiple concentrations (10–200 ug/ml) for 6 hours. Cell lysates and culture supernatants were collected and analysed for IL-1β using ELISA. Cell viability was also measured. Results AcLDL induced the release of IL-1β from stimulated HCAECs in a dose dependent manner with maximum release (155.4 pg/ml, n=3) at concentration of 50 ug/ml. This was 4 fold greater than that released by cytokine-stimulated (38.49 pg/ml, n=3) and 3 fold greater than that released by neutrophil elastase (49.45 pg/ml, n=3). This release was not caused by toxicity of the AcLDL: cell viability was confirmed by lactate dehydrogenase cell viability assay. Conclusion AcLDL is capable of eliciting IL-1β release in activated HCAECs, without causing toxicity.
- Published
- 2018
44. Neutrophil microvesicles drive atherosclerosis by deliveringmiR-155to atheroprone endothelium
- Author
-
Chiara Recarti, Le Anh Luong, Amanda Burnett, Celine Souilhol, Ben Ward, Endre Kiss-Toth, Jessica Johnston, Victoria Ridger, Merete Long, Marwa Mahmoud, Sheila E. Francis, Andreas Schober, Siôn A Parry, Paul Hellewell, Marc A. M. J. van Zandvoort, Mark P. Ariaans, Carl J. Hulston, Birke J. Benedikter, Ingrid Gomez, Rachel M Woods, Laura West, Paul C. Evans, and Rohit Bazaz
- Subjects
miR-155 ,Pathogenesis ,medicine.anatomical_structure ,Endothelium ,business.industry ,Vascular inflammation ,microRNA ,Cancer research ,Medicine ,Disturbed flow ,business ,Microvesicles ,Pathophysiology - Abstract
Neutrophils have been implicated in the pathogenesis of atherosclerosis, a lipid-driven disease of arteries, but they are seldom found in atherosclerotic plaques. To resolve this longstanding paradox, we investigated whether neutrophil-derived microvesicles may influence arterial pathophysiology. Clinical and pre-clinical studies revealed that levels of circulating neutrophil microvesicles were enhanced by exposure to a high fat diet, a known risk factor for atherosclerosis. Neutrophil microvesicles accumulated at disease-prone regions of arteries that are exposed to complex flow patterns, and they promoted vascular inflammation and atherosclerosis in a murine model. Using cultured endothelial cells exposed to disturbed flow, it was demonstrated that neutrophil microvesicles promoted inflammatory gene expression by delivering a microRNA (miR-155) that enhanced NF-κB activation. Similary, neutrophil microvesicles increased miR-155 and enhanced NF-κB at disease-prone sites of disturbed flow in arteries of mice. We conclude that delivery of microvesicles carrying miR-155 to disease-prone regions of arteries provides a novel mechanism by which neutrophils contribute to vascular inflammation and atherogenesis.
- Published
- 2018
- Full Text
- View/download PDF
45. Experimental models of murine atherosclerosis: does perception match reality?
- Author
-
Jessica M, Johnston, Sheila E, Francis, and Endre, Kiss-Toth
- Subjects
Time Factors ,Mice, Knockout, ApoE ,Arteries ,Atherosclerosis ,Diet, High-Fat ,Lipids ,Severity of Illness Index ,Plaque, Atherosclerotic ,Disease Models, Animal ,Phenotype ,Receptors, LDL ,Disease Progression ,Animals ,Genetic Predisposition to Disease ,Proprotein Convertase 9 - Published
- 2018
46. Neutrophil Elastase Promotes Interleukin-1β Secretion from Human Coronary Endothelium
- Author
-
Amanda Burnett, Victoria Ridger, Marc Daigneault, Janet Chamberlain, Mabruka Alfaidi, Sheila E. Francis, and Heather L. Wilson
- Subjects
Male ,Endothelium ,Cell Survival ,Interleukin-1beta ,Immunology ,Apoptosis ,Mice, Transgenic ,Inflammation ,Pharmacology ,Biochemistry ,Gene Expression Regulation, Enzymologic ,Microcirculation ,Pathogenesis ,Mice ,Apolipoproteins E ,In vivo ,medicine ,Animals ,Humans ,Secretion ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,biology ,Endothelial Cells ,Cell Biology ,Coronary Vessels ,Immunohistochemistry ,Plaque, Atherosclerotic ,medicine.anatomical_structure ,Neutrophil elastase ,biology.protein ,Calcium ,Endothelium, Vascular ,medicine.symptom ,Leukocyte Elastase ,Intracellular - Abstract
The endothelium is critically involved in the pathogenesis of atherosclerosis by producing proinflammatory mediators, including interleukin-1 beta (IL-1β). Coronary arteries from patients with ischaemic heart disease express large amounts of IL-1β in endothelium. However, the mechanism by which endothelial cells (ECs) release IL-1β remains to be elucidated. We investigated neutrophil elastase (NE), a potent serine protease detected in vulnerable areas of human carotid plaques, as a potential ′trigger′ for IL-1β processing and release. This study tested the hypothesis that NE potentiates the processing and release of IL-1β from human coronary endothelium. We found that NE cleaves the pro-isoform of IL-1β in ECs and causes significant secretion of bioactive IL-1β via extracellular vesicles. This release was significantly attenuated by inhibition of neutrophil elastase, but not caspase-1. Transient increases in intracellular Ca2+ levels were observed prior to secretion. Inside ECs, and after NE treatment only, IL-1β was detected within LAMP-1 positive multivesicular bodies (MVBs). The released vesicles contained bioactive IL-1β. In vivo, in experimental atherosclerosis, NE was detected in mature atherosclerotic plaques, predominantly in the endothelium, alongside IL-1β. This study reveals a novel mechanistic link between NE expression in atherosclerotic plaques and concomitant pro-inflammatory bioactive IL-1β secretion from ECs; this could reveal additional potential anti-IL-1β therapeutic targets and provide further insight into the inflammatory process by which vascular disease develops.
- Published
- 2015
47. Coronary stents seeded with human trophoblastic endovascular progenitor cells show accelerated strut coverage without excessive neointimal proliferation in a porcine model
- Author
-
Tushar Raina, Behrouz Aflatoonian, Sheila E. Francis, Javaid Iqbal, Harry Moore, Kadem Al-Lamee, N Arnold, Julian Gunn, James Walsh, and Sam Whitehouse
- Subjects
Bare-metal stent ,Pathology ,medicine.medical_specialty ,Swine ,medicine.medical_treatment ,Coronary Angiography ,Restenosis ,Neointima ,Intravascular ultrasound ,medicine ,Animals ,Progenitor cell ,Cells, Cultured ,Embryonic Stem Cells ,Ultrasonography, Interventional ,Cell Proliferation ,Neointimal hyperplasia ,medicine.diagnostic_test ,Guided Tissue Regeneration ,business.industry ,Stem Cells ,Percutaneous coronary intervention ,Stent ,medicine.disease ,Trophoblasts ,Endothelial stem cell ,Models, Animal ,Microscopy, Electron, Scanning ,Stents ,Endothelium, Vascular ,Radiology ,Cardiology and Cardiovascular Medicine ,business - Abstract
The success of percutaneous coronary intervention (PCI) has been limited by restenosis and stent thrombosis. Delayed or incomplete endothelial regeneration is believed to be a key factor responsible for these events. Developing a stent with an accelerated healing profile may be of benefit. We aimed to evaluate the feasibility and safety of seeding a bare metal stent (BMS) with human trophoblastic endovascular progenitor cells (hTEC) derived from human embryonic stem cells. A porcine coronary artery model was used to compare the rate and extent of endothelial regeneration and the degree of neointimal proliferation. Characterisation of hTEC confirmed a mixed progenitor and endothelial cell phenotype. The biodistribution and fate of hTEC were studied using radiolabelled 111Indium oxine and fluorescent in situ hybridisation. Scanning electron microscopy showed earlier endothelial coverage in hTEC-seeded stents as compared to similar BMS. hTEC-seeded BMS achieved complete stent coverage in three days. Quantitative coronary angiography, intravascular ultrasound assessment and histomorphometry showed no difference in neointimal hyperplasia between hTEC-seeded and control BMS. hTEC seeding of coronary stents is a novel and safe approach to accelerate endothelial regeneration without increasing neointimal proliferation.
- Published
- 2014
48. Streptococcus pneumoniaeworsens cerebral ischemia via interleukin 1 and platelet glycoprotein Ibα
- Author
-
Helen Casbolt, Katie N. Murray, David H. Dockrell, Peter Warn, Bazaz Rohit, Bernadett Martinecz, Janet Chamberlain, Adam Denes, Bernhard Nieswandt, Sheila E. Francis, Caroline Drake, Nancy J. Rothwell, Jesús M. Pradillo, Andrew Sharp, and Stuart M. Allan
- Subjects
0303 health sciences ,business.industry ,Ischemia ,Interleukin ,Platelet Glycoprotein GPIb-IX Complex ,Inflammation ,medicine.disease ,Systemic inflammation ,3. Good health ,Brain ischemia ,03 medical and health sciences ,0302 clinical medicine ,Neurology ,Immunology ,medicine ,Neurology (clinical) ,Platelet activation ,medicine.symptom ,business ,Stroke ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
OBJECTIVE: Bacterial infection contributes to diverse noninfectious diseases and worsens outcome after stroke. Streptococcus pneumoniae, the most common infection in patients at risk of stroke, is a major cause of prolonged hospitalization and death of stroke patients, but how infection impacts clinical outcome is not known. METHODS: We induced sustained pulmonary infection by a human S. pneumoniae isolate in naive and comorbid rodents to investigate the effect of infection on vascular and inflammatory responses prior to and after cerebral ischemia. RESULTS: S. pneumoniae infection triggered atherogenesis, led to systemic induction of interleukin (IL) 1, and profoundly exacerbated (50-90%) ischemic brain injury in rats and mice, a response that was more severe in combination with old age and atherosclerosis. Systemic blockade of IL-1 with IL-1 receptor antagonist (IL-1Ra) fully reversed infection-induced exacerbation of brain injury and functional impairment caused by cerebral ischemia. We show that infection-induced systemic inflammation mediates its effects via increasing platelet activation and microvascular coagulation in the brain after cerebral ischemia, as confirmed by reduced brain injury in response to blockade of platelet glycoprotein (GP) Ib?. IL-1 and platelet-mediated signals converge on microglia, as both IL-1Ra and GPIb? blockade reversed the production of IL-1? by microglia in response to cerebral ischemia in infected animals. INTERPRETATION: S. pneumoniae infection augments atherosclerosis and exacerbates ischemic brain injury via IL-1 and platelet-mediated systemic inflammation. These mechanisms may contribute to diverse cardio- and cerebrovascular pathologies in humans.
- Published
- 2014
49. Vessel wall, not platelet, P2Y12 potentiates early atherogenesis
- Author
-
Laura West, Tanja Steiner, Robert F. Storey, Heather M Judge, and Sheila E. Francis
- Subjects
Blood Platelets ,Male ,Ticagrelor ,medicine.medical_specialty ,Adenosine ,Ticlopidine ,P-selectin ,Physiology ,Mice ,Apolipoproteins E ,P2Y12 ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Platelet ,Platelet activation ,Receptor ,Mice, Knockout ,business.industry ,Atherosclerosis ,medicine.disease ,Clopidogrel ,Receptors, Purinergic P2Y12 ,Mice, Inbred C57BL ,P-Selectin ,Endocrinology ,Atheroma ,Immunology ,Blood Vessels ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
Aims Platelets have a fundamental role in atherothrombosis, but their role in early atherogenesis is unclear. The P2Y12 receptor is responsible for amplifying and sustaining platelet activation and P2Y12 inhibition is crucial in modulating the vessel wall response to injury. We therefore examined the role of platelet vs. vessel wall P2Y12 in early atherogenesis and considered the use of P2Y12 antagonists ticagrelor and clopidogrel in modulating this process. Methods and results ApoE−/− and ApoE−/−P2Y12−/− male mice underwent bone marrow transplantation and were fed a western diet for 4 weeks before assessing atherosclerotic burden. Compared with ApoE−/− controls, platelet P2Y12 deficiency profoundly reduced platelet reactivity but had no effect on atheroma formation, whereas vessel wall P2Y12 deficiency significantly attenuated atheroma in the aortic sinus and brachiocephalic artery (both P < 0.001). ApoE−/− and ApoE−/−P2Y12−/− male mice fed western diet plus either twice-daily doses of ticagrelor (100 mg/kg) or daily clopidogrel (20 mg/kg) for 4 weeks exhibited no significant reduction in atheroma compared with control mice fed mannitol. Attenuated P-selectin expression confirmed platelet P2Y12 inhibition in drug-treated mice. Conclusions Despite its major contribution to platelet reactivity, platelet P2Y12 has no effect on early atheroma formation, whereas vessel wall P2Y12 is important in this process. Ticagrelor and clopidogrel effectively reduced platelet reactivity but were unable to inhibit early atherogenesis, demonstrating that these P2Y12 inhibitors may not be effective in preventing early disease.
- Published
- 2014
50. P16 HUMAN MACROPHAGE SUBSETS IN THE PATHOGENESIS OF CAROTID ATHEROSCLEROSIS
- Author
-
Jessica Redgrave, K Baidzajevas, Endre Kiss-Toth, Sheila E. Francis, Arshad Majid, R Alqurashi, Heather L. Wilson, Klaudia Kocsy, and Jessica Johnston
- Subjects
0301 basic medicine ,Pathogenesis ,Carotid atherosclerosis ,03 medical and health sciences ,030102 biochemistry & molecular biology ,Physiology ,business.industry ,Physiology (medical) ,Immunology ,Medicine ,Macrophage ,Cardiology and Cardiovascular Medicine ,business - Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.