Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Myriam Chimen is active.

Publication


Featured researches published by Myriam Chimen.


Diabetologia | 2012

What are the health benefits of physical activity in type 1 diabetes mellitus? A literature review

Myriam Chimen; Amy Kennedy; Krishnarajah Nirantharakumar; T. T. Pang; Rob C Andrews; Parth Narendran

Physical activity improves well-being and reduces the risk of heart disease, cancer and type 2 diabetes mellitus in the general population. In individuals with established type 2 diabetes, physical activity improves glucose and lipid levels, reduces weight and improves insulin resistance. In type 1 diabetes mellitus, however, the benefits of physical activity are less clear. There is poor evidence for a beneficial effect of physical activity on glycaemic control and microvascular complications, and significant risk of harm through hypoglycaemia. Here we review the literature relating to physical activity and health in type 1 diabetes. We examine its effect on a number of outcomes, including glycaemic control, lipids, blood pressure, diabetic complications, well-being and overall mortality. We conclude that whilst there is sufficient evidence to recommend physical activity in the management of type 1 diabetes, it is still unclear as to what form, duration and intensity should be recommended and whether there is benefit for many of the outcomes examined.


Science Translational Medicine | 2014

A Zebrafish Compound Screen Reveals Modulation of Neutrophil Reverse Migration as an Anti-Inflammatory Mechanism

Anne L. Robertson; Geoffrey R. Holmes; Aleksandra Bojarczuk; Joseph Burgon; Catherine A. Loynes; Myriam Chimen; Amy Sawtell; Bashar Hamza; Joseph Willson; Sarah R. Walmsley; Sean R. Anderson; Mark Coles; Stuart N. Farrow; Roberto Solari; Simon Jones; Lynne R. Prince; Daniel Irimia; G. Ed Rainger; Visakan Kadirkamanathan; Moira K. B. Whyte; Stephen A. Renshaw

The proresolution therapeutic tanshinone IIA drives inflammation resolution by reverse migration. An Anti-Inflammatory Fish Story Inflammation is one way the body tries to protect itself from injury and begin the healing process. However, as with any good thing, too much inflammation can be harmful, causing bystander injuries to healthy tissue. Hence, there is an active mechanism to resolve inflammation; failed resolution contributes to diseases of chronic inflammation such as atherosclerosis and rheumatoid arthritis. Now, Robertson et al. use a zebrafish screening platform to identify new means of resolving inflammation. The authors used a transgenic zebrafish model of sterile tissue injury to screen potential factors involved in inflammation resolution. They found that tanshinone IIA, which is derived from a Chinese medicinal herb, had proresolving activity by both inducing neutrophil apoptosis and promoting reverse migration of neutrophils. What’s more, these effects were not limited to their zebrafish model but held true in human neutrophils. Although efficacy remains to be tested in actual patients, these data support “fishing” for new drug candidates for resolving inflammation. Diseases of failed inflammation resolution are common and largely incurable. Therapeutic induction of inflammation resolution is an attractive strategy to bring about healing without increasing susceptibility to infection. However, therapeutic targeting of inflammation resolution has been hampered by a lack of understanding of the underlying molecular controls. To address this drug development challenge, we developed an in vivo screen for proresolution therapeutics in a transgenic zebrafish model. Inflammation induced by sterile tissue injury was assessed for accelerated resolution in the presence of a library of known compounds. Of the molecules with proresolution activity, tanshinone IIA, derived from a Chinese medicinal herb, potently induced inflammation resolution in vivo both by induction of neutrophil apoptosis and by promoting reverse migration of neutrophils. Tanshinone IIA blocked proinflammatory signals in vivo, and its effects are conserved in human neutrophils, supporting a potential role in treating human inflammation and providing compelling evidence of the translational potential of this screening strategy.


PLOS ONE | 2013

Does exercise improve glycaemic control in type 1 diabetes? A systematic review and meta-analysis.

Amy Kennedy; Krishnarajah Nirantharakumar; Myriam Chimen; Terence T. Pang; Karla Hemming; Rob C Andrews; Parth Narendran

Objective Whilst regular exercise is advocated for people with type 1 diabetes, the benefits of this therapy are poorly delineated. Our objective was to review the evidence for a glycaemic benefit of exercise in type 1 diabetes. Research Design and Methods Electronic database searches were carried out in MEDLINE, Embase, Cochrane’s Controlled Trials Register and SPORTDiscus. In addition, we searched for as yet unpublished but completed trials. Glycaemic benefit was defined as an improvement in glycosylated haemoglobin (HbA1c). Both randomised and non-randomised controlled trials were included. Results Thirteen studies were identified in the systematic review. Meta-analysis of twelve of these (including 452 patients) demonstrated an HbA1c reduction but this was not statistically significant (standardised mean difference (SMD) −0.25; 95% CI, −0.59 to 0.09). Conclusions This meta-analysis does not reveal evidence for a glycaemic benefit of exercise as measured by HbA1c. Reasons for this finding could include increased calorie intake, insulin dose reductions around the time of exercise or lack of power. We also suggest that HbA1c may not be a sensitive indicator of glycaemic control, and that improvement in glycaemic variability may not be reflected in this measure. Exercise does however have other proven benefits in type 1 diabetes, and remains an important part of its management.


Platelets | 2015

The role of platelets in the recruitment of leukocytes during vascular disease

G. Ed Rainger; Myriam Chimen; Matthew J. Harrison; Clara M. Yates; Paul Harrison; Stephen P. Watson; Marie Lordkipanidzé; Gerard B. Nash

Abstract Besides their role in the formation of thrombus during haemostasis, it is becoming clear that platelets contribute to a number of other processes within the vasculature. Indeed, the integrated function of the thrombotic and inflammatory systems, which results in platelet-mediated recruitment of leukocytes, is now considered to be of great importance in the propagation, progression and pathogenesis of atherosclerotic disease of the arteries. There are three scenarios by which platelets can interact with leukocytes: (1) during haemostasis, when platelets adhere to and are activated on sub-endothelial matrix proteins exposed by vascular damage and then recruit leukocytes to a growing thrombus. (2) Platelets adhere to and are activated on stimulated endothelial cells and then bridge blood borne leukocytes to the vessel wall and. (3) Adhesion between platelets and leukocytes occurs in the blood leading to formation of heterotypic aggregates prior to contact with endothelial cells. In the following review we will not discuss leukocyte recruitment during haemostasis, as this represents a physiological response to tissue trauma that can progress, at least in its early stages, in the absence of inflammation. Rather we will deal with scenarios 2 and 3, as these pathways of platelet–leukocyte interactions are important during inflammation and in chronic inflammatory diseases such as atherosclerosis. Indeed, these interactions mean that leukocytes possess means of adhesion to the vessel wall under conditions that may not normally be permissive of leukocyte–endothelial cell adhesion, meaning that the disease process may be able to bypass the regulatory pathways which would ordinarily moderate the inflammatory response.


Nature Medicine | 2015

Homeostatic regulation of T cell trafficking by a B cell-derived peptide is impaired in autoimmune and chronic inflammatory disease

Myriam Chimen; Helen M. McGettrick; Bonita H. R. Apta; Sahithi J. Kuravi; Clara M. Yates; Amy Kennedy; Arjun Odedra; Mohammed Alassiri; Matthew J. Harrison; Ashley Martin; Francesca Barone; Saba Nayar; Jessica Hitchcock; Adam F. Cunningham; Karim Raza; Andrew Filer; David A. Copland; Andrew D. Dick; Joseph Robinson; Neena Kalia; Lucy S. K. Walker; Christopher D. Buckley; Gerard B. Nash; Parth Narendran; G. Ed Rainger

During an inflammatory response, lymphocyte recruitment into tissue must be tightly controlled because dysregulated trafficking contributes to the pathogenesis of chronic disease. Here we show that during inflammation and in response to adiponectin, B cells tonically inhibit T cell trafficking by secreting a peptide (PEPITEM) proteolytically derived from 14.3.3 zeta delta (14.3.3.ζδ) protein. PEPITEM binds cadherin-15 on endothelial cells, promoting synthesis and release of sphingosine-1 phosphate, which inhibits trafficking of T cells without affecting recruitment of other leukocytes. Expression of adiponectin receptors on B cells and adiponectin-induced PEPITEM secretion wanes with age, implying immune senescence of the pathway. Additionally, these changes are evident in individuals with type 1 diabetes or rheumatoid arthritis, and circulating PEPITEM in patient serum is reduced compared to that of healthy age-matched donors. In both diseases, tonic inhibition of T cell trafficking across inflamed endothelium is lost. Control of patient T cell trafficking is re-established by treatment with exogenous PEPITEM. Moreover, in animal models of peritonitis, hepatic ischemia-reperfusion injury, Salmonella infection, uveitis and Sjögrens syndrome, PEPITEM reduced T cell recruitment into inflamed tissues.


The Journal of Clinical Endocrinology and Metabolism | 2013

Inhibition of Islet Immunoreactivity by Adiponectin Is Attenuated in Human Type 1 Diabetes

Terence T L Pang; Myriam Chimen; Edward Goble; Natalie C. Dixon; Aled Benbow; Suzy Eldershaw; Dylan Thompson; S. C. L. Gough; P. Narendran

CONTEXT Adiponectin is an adipocyte-derived cytokine with insulin-sensitizing and antiinflammatory properties. These dual actions have not previously been examined in the context of human disease. OBJECTIVES Our objective was to examine the adiponectin axis in type 1 diabetes (T1D). T1D is an autoimmune inflammatory disease resulting from pancreatic β-cell destruction, in which insulin resistance associates with progression to disease. DESIGN, PATIENTS, AND INTERVENTIONS We measured circulating adiponectin and adiponectin receptor expression on blood-immune cells from 108 matched healthy, T1D, and type 2 diabetic subjects. We tested adiponectin effect on T cell proliferation to islet antigens and antigen-presenting function of monocyte-derived dendritic cells (mDCs). Lastly, we assessed the effect of a 3-week lifestyle intervention program on immune cell adiponectin receptor expression in 18 healthy subjects. RESULTS Circulating concentrations of adiponectin were not affected by T1D. However, expression of adiponectin receptors on blood monocytes was markedly reduced and inversely associated with insulin resistance. Reduced adiponectin receptor expression resulted in increased T cell proliferation to islet-antigen presented by autologous mDCs. We demonstrated a critical role for adiponectin in down-regulating the costimulatory molecule CD86 on mDCs, and this function was impaired in T1D. We proceeded to show that lifestyle intervention increased adiponectin receptor but reduced CD86 expression on monocytes. CONCLUSIONS These data indicate that T cells are released from the antiinflammatory effects of adiponectin in T1D and suggest a mechanism linking insulin resistance and islet immunity. Furthermore, we suggest that interventions that reduce insulin resistance could modulate the inflammatory process in T1D.


Journal of Immunology | 2017

Monocyte subsets coregulate inflammatory responses by integrated signaling through TNF and IL-6 at the endothelial cell interface.

Myriam Chimen; Clara M. Yates; Helen M. McGettrick; Lewis S. C. Ward; Matthew J. Harrison; Bonita H. R. Apta; Lea H. Dib; Beat A. Imhof; Paul Harrison; Gerard B. Nash; G. Ed Rainger

Two major monocyte subsets, CD14+CD16− (classical) and CD14+/dimCD16+ (nonclassical/intermediate), have been described. Each has different functions ascribed in its interactions with vascular endothelial cells (EC), including migration and promoting inflammation. Although monocyte subpopulations have been studied in isolated systems, their influence on EC and on the course of inflammation has been ignored. In this study, using unstimulated or cytokine-activated EC, we observed significant differences in the recruitment, migration, and reverse migration of human monocyte subsets. Associated with this, and based on their patterns of cytokine secretion, there was a difference in their capacity to activate EC and support the secondary recruitment of flowing neutrophils. High levels of TNF were detected in cocultures with nonclassical/intermediate monocytes, the blockade of which significantly reduced neutrophil recruitment. In contrast, classical monocytes secreted high levels of IL-6, the blockade of which resulted in increased neutrophil recruitment. When cocultures contained both monocyte subsets, or when conditioned supernatant from classical monocytes cocultures (IL-6hi) was added to nonclassical/intermediate monocyte cocultures (TNFhi), the activating effects of TNF were dramatically reduced, implying that when present, the anti-inflammatory activities of IL-6 were dominant over the proinflammatory activities of TNF. These changes in neutrophil recruitment could be explained by regulation of E-selectin on the cocultured EC. This study suggests that recruited human monocyte subsets trigger a regulatory pathway of cytokine-mediated signaling at the EC interface, and we propose that this is a mechanism for limiting the phlogistic activity of newly recruited monocytes.


Journal of Immunology | 2017

ADAM10-Interacting Tetraspanins Tspan5 and Tspan17 Regulate VE-Cadherin Expression and Promote T Lymphocyte Transmigration

Jasmeet S. Reyat; Myriam Chimen; Peter J. Noy; Justyna Szyroka; G. Ed Rainger; Michael G. Tomlinson

The recruitment of blood leukocytes across the endothelium to sites of tissue infection is central to inflammation, but also promotes chronic inflammatory diseases. A disintegrin and metalloproteinase 10 (ADAM10) is a ubiquitous transmembrane molecular scissor that is implicated in leukocyte transmigration by proteolytically cleaving its endothelial substrates. These include VE-cadherin, a homotypic adhesion molecule that regulates endothelial barrier function, and transmembrane chemokines CX3CL1 and CXCL16, which have receptors on leukocytes. However, a definitive role for endothelial ADAM10 in transmigration of freshly isolated primary leukocytes under flow has not been demonstrated, and the relative importance of distinct ADAM10 substrates is unknown. Emerging evidence suggests that ADAM10 can be regarded as six different molecular scissors with different substrate specificities, depending on which of six TspanC8 tetraspanins it is associated with, but TspanC8s remain unstudied in leukocyte transmigration. In the current study, ADAM10 knockdown on primary HUVECs was found to impair transmigration of freshly isolated human peripheral blood T lymphocytes, but not neutrophils or B lymphocytes, in an in vitro flow assay. This impairment was due to delayed transmigration rather than a complete block, and was overcome in the presence of neutrophils. Transmigration of purified lymphocytes was dependent on ADAM10 regulation of VE-cadherin, but not CX3CL1 and CXCL16. Tspan5 and Tspan17, the two most closely related TspanC8s by sequence, were the only TspanC8s that regulated VE-cadherin expression and were required for lymphocyte transmigration. Therefore endothelial Tspan5- and Tspan17-ADAM10 complexes may regulate inflammation by maintaining normal VE-cadherin expression and promoting T lymphocyte transmigration.


Cardiovascular Research | 2018

Nitrite circumvents platelet resistance to nitric oxide in patients with heart failure preserved ejection fraction and chronic atrial fibrillation

Alessandra Borgognone; Eduard Shantsila; Sophie M Worrall; Eakkapote Prompunt; Thomas Loka; Brodie L. Loudon; Myriam Chimen; G. Ed Rainger; Janet M. Lord; Ashley Turner; Peter Nightingale; Martin Feelisch; Paulus Kirchhof; Gregory Y.H. Lip; Steve P. Watson; Michael P. Frenneaux; Melanie Madhani

Abstract Aims Heart failure (HF) is a pro-thrombotic state. Both platelet and vascular responses to nitric oxide (NO) donors are impaired in HF patients with reduced ejection fraction (HFrEF) compared with healthy volunteers (HVs) due to scavenging of NO, and possibly also reduced activity of the principal NO sensor, soluble guanylate cyclase (sGC), limiting the therapeutic potential of NO donors as anti-aggregatory agents. Previous studies have shown that nitrite inhibits platelet activation presumptively after its reduction to NO, but the mechanism(s) involved remain poorly characterized. Our aim was to compare the effects of nitrite on platelet function in HV vs. HF patients with preserved ejection fraction (HFpEF) and chronic atrial fibrillation (HFpEF–AF), vs. patients with chronic AF without HF, and to assess whether these effects occur independent of the interaction with other formed elements of blood. Methods and results Platelet responses to nitrite and the NO donor sodium nitroprusside (SNP) were compared in age-matched HV controls (n = 12), HFpEF–AF patients (n = 29), and chronic AF patients (n = 8). Anti-aggregatory effects of nitrite in the presence of NO scavengers/sGC inhibitor were determined and vasodilator-stimulated phosphoprotein (VASP) phosphorylation was assessed using western blotting. In HV and chronic AF, both nitrite and SNP inhibited platelet aggregation in a concentration-dependent manner. Inhibition of platelet aggregation by the NO donor SNP was impaired in HFpEF-AF patients compared with healthy and chronic AF individuals, but there was no impairment of the anti-aggregatory effects of nitrite. Nitrite circumvented platelet NO resistance independently of other blood cells by directly activating sGC and phosphorylating VASP. Conclusion We here show for the first time that HFpEF-AF (but not chronic AF without HF) is associated with marked impairment of platelet NO responses due to sGC dysfunction and nitrite circumvents the ‘platelet NO resistance’ phenomenon in human HFpEF, at least partly, by acting as a direct sGC activator independent of NO.


Archive | 2017

Introduction: T Cell Trafficking in Inflammation and Immunity

Myriam Chimen; Bonita H. R. Apta; Helen M. McGettrick

T cell migration across vascular endothelium is essential for T cell responses, as through the expression of specific tissue-homing receptors, these cells then access peripheral tissues, with the goal of eliminating invading pathogens and/or tumor cells. However, aberrant trafficking of T cells to peripheral tissues contributes to the development of most chronic inflammatory diseases. Very little is known about the mechanisms by which T cell trafficking is regulated during inflammation, and it is thus difficult to target this aspect of pathology for the development of new therapies. It is therefore important to understand the pathways involved in regulating the recruitment of immune cells.

Collaboration


Dive into the Myriam Chimen's collaboration.

Top Co-Authors

Avatar

Clara M. Yates

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar

Gerard B. Nash

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

G. Ed Rainger

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar

G.E. Rainger

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar

Amy Kennedy

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge