Network


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

Hotspot


Dive into the research topics where Romana S Mughal is active.

Publication


Featured researches published by Romana S Mughal.


Clinical and Experimental Pharmacology and Physiology | 2009

PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR γ-INDEPENDENT EFFECTS OF THIAZOLIDINEDIONES ON HUMAN CARDIAC MYOFIBROBLAST FUNCTION

Romana S Mughal; Philip Warburton; David J. O’Regan; Stephen G. Ball; Neil A. Turner; Karen E. Porter

1 Thiazolidinediones (TZDs) are peroxisome proliferator‐activated receptor (PPAR) γ agonists that are used to lower insulin resistance in Type 2 diabetic patients. Although TZDs exhibit beneficial effects on the vasculature, their effects on the heart are less clear and are the subject of current clinical debate. Thiazolidinediones have been reported to reduce adverse myocardial remodelling, a pathology in which cardiac myofibroblasts (CMF) are pivotal. 2 The aim of the present study was to investigate whether TZDs modulate specific human CMF functions of importance to the myocardial remodelling process and to determine whether any of these effects were mediated via PPARγ activation. 3 Immunoblotting of cultured human CMF homogenates revealed strong expression of PPARγ (approximately 50 kDa). Three different TZDs (ciglitazone, rosiglitazone and troglitazone) and the endogenous PPARγ ligand 15‐deoxy‐δ12,14‐prostaglandin J2 (15d‐PGJ2) inhibited CMF proliferation (cell number and expression of proliferating cell nuclear antigen) in a concentration‐dependent manner (range 0.1–10 µmol/L) with similar potencies. This antiproliferative effect of TZDs was not reversed by the PPARγ antagonists GW9662 or T0070907 (10–25 µmol/L). None of the TZDs or 15d‐PGJ2 affected cell migration or invasion (Boyden chamber assays without or with Matrigel barrier), matrix metalloproteinase‐2 or ‐9 secretion (gelatin zymography) or the actin cytoskeleton (rhodamine/phalloidin fluorescent confocal microscopy). 4 In conclusion, TZDs reduce human CMF proliferation via a PPARγ‐independent mechanism. Although TZDs do not inhibit CMF invasion, their antiproliferative activity may contribute to the ability of this class of drugs to modulate adverse myocardial remodelling.


Diabetes | 2013

Diurnal variation in vascular and metabolic function in diet-induced obesity: divergence of insulin resistance and loss of clock rhythm

Madhu Prasai; Romana S Mughal; Stephen B. Wheatcroft; Mark T. Kearney; Peter J. Grant; Eleanor M. Scott

Circadian rhythms are integral to the normal functioning of numerous physiological processes. Evidence from human and mouse studies suggests that loss of rhythm occurs in obesity and cardiovascular disease and may be a neglected contributor to pathophysiology. Obesity has been shown to impair the circadian clock mechanism in liver and adipose tissue but its effect on cardiovascular tissues is unknown. We investigated the effect of diet-induced obesity in C57BL6J mice upon rhythmic transcription of clock genes and diurnal variation in vascular and metabolic systems. In obesity, clock gene function and physiological rhythms were preserved in the vasculature but clock gene transcription in metabolic tissues and rhythms of glucose tolerance and insulin sensitivity were blunted. The most pronounced attenuation of clock rhythm occurred in adipose tissue, where there was also impairment of clock-controlled master metabolic genes and both AMPK mRNA and protein. Across tissues, clock gene disruption was associated with local inflammation but diverged from impairment of insulin signaling. We conclude that vascular tissues are less sensitive to pathological disruption of diurnal rhythms during obesity than metabolic tissues and suggest that cellular disruption of clock gene rhythmicity may occur by mechanisms shared with inflammation but distinct from those leading to insulin resistance.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Haploinsufficiency of the Insulin-Like Growth Factor-1 Receptor Enhances Endothelial Repair and Favorably Modifies Angiogenic Progenitor Cell Phenotype

Nadira Yuldasheva; Sheikh Tawqeer Rashid; Natalie Haywood; Paul A. Cordell; Romana S Mughal; Hema Viswambharan; Helen Imrie; Piruthivi Sukumar; Richard M. Cubbon; Amir Aziz; Matthew Gage; Kamatamu Amanda Mbonye; Jessica Smith; S Galloway; A Skromna; D. Julian A. Scott; Mark T. Kearney; Stephen B. Wheatcroft

Objectives— Defective endothelial regeneration predisposes to adverse arterial remodeling and is thought to contribute to cardiovascular disease in type 2 diabetes mellitus. We recently demonstrated that the type 1 insulin-like growth factor receptor (IGF1R) is a negative regulator of insulin sensitivity and nitric oxide bioavailability. In this report, we examined partial deletion of the IGF1R as a potential strategy to enhance endothelial repair. Approach and Results— We assessed endothelial regeneration after wire injury in mice and abundance and function of angiogenic progenitor cells in mice with haploinsufficiency of the IGF1R (IGF1R+/−). Endothelial regeneration after arterial injury was accelerated in IGF1R+/− mice. Although the yield of angiogenic progenitor cells was lower in IGF1R+/− mice, these angiogenic progenitor cells displayed enhanced adhesion, increased secretion of insulin-like growth factor-1, and enhanced angiogenic capacity. To examine the relevance of IGF1R manipulation to cell-based therapy, we transfused IGF1R+/− bone marrow–derived CD117+ cells into wild-type mice. IGF1R+/− cells accelerated endothelial regeneration after arterial injury compared with wild-type cells and did not alter atherosclerotic lesion formation. Conclusions— Haploinsufficiency of the IGF1R is associated with accelerated endothelial regeneration in vivo and enhanced tube forming and adhesive potential of angiogenic progenitor cells in vitro. Partial deletion of IGF1R in transfused bone marrow–derived CD117+ cells enhanced their capacity to promote endothelial regeneration without altering atherosclerosis. Our data suggest that manipulation of the IGF1R could be exploited as novel therapeutic approach to enhance repair of the arterial wall after injury.


Stem Cells | 2014

Restoring Akt1 activity in outgrowth endothelial cells from south asian men rescues vascular reparative potential

Richard M. Cubbon; Nadira Yuldasheva; Hema Viswambharan; Ben Mercer; Baliga; Sam L. Stephen; J Askham; Piruthivi Sukumar; A Skromna; Romana S Mughal; Amn Walker; Alexander F. Bruns; Marc A. Bailey; S Galloway; Helen Imrie; Matthew Gage; Mark Rakobowchuk; Jing Li; Karen E. Porter; Sreenivasan Ponnambalam; Stephen B. Wheatcroft; David J. Beech; Mark T. Kearney

Recent data suggest reduced indices of vascular repair in South Asian men, a group at increased risk of cardiovascular events. Outgrowth endothelial cells (OEC) represent an attractive tool to study vascular repair in humans and may offer potential in cell‐based repair therapies. We aimed to define and manipulate potential mechanisms of impaired vascular repair in South Asian (SA) men. In vitro and in vivo assays of vascular repair and angiogenesis were performed using OEC derived from SA men and matched European controls, prior defining potentially causal molecular mechanisms. SA OEC exhibited impaired colony formation, migration, and in vitro angiogenesis, associated with decreased expression of the proangiogenic molecules Akt1 and endothelial nitric oxide synthase (eNOS). Transfusion of European OEC into immunodeficient mice after wire‐induced femoral artery injury augmented re‐endothelialization, in contrast with SA OEC and vehicle; SA OEC also failed to promote angiogenesis after induction of hind limb ischemia. Expression of constitutively active Akt1 (E17KAkt), but not green fluorescent protein control, in SA OEC increased in vitro angiogenesis, which was abrogated by a NOS antagonist. Moreover, E17KAkt expressing SA OEC promoted re‐endothelialization of wire‐injured femoral arteries, and perfusion recovery of ischemic limbs, to a magnitude comparable with nonmanipulated European OEC. Silencing Akt1 in European OEC recapitulated the functional deficits noted in SA OEC. Reduced signaling via the Akt/eNOS axis is causally linked with impaired OEC‐mediated vascular repair in South Asian men. These data prove the principle of rescuing marked reparative dysfunction in OEC derived from these men. Stem Cells 2014;32:2714–2723


Biochemical and Biophysical Research Communications | 2012

Divergent effects of 17-β-estradiol on human vascular smooth muscle and endothelial cell function diminishes TNF-α-induced neointima formation

Rungrat Nintasen; Kirsten Riches; Romana S Mughal; Parnpen Viriyavejakul; Urai Chaisri; Yaowapa Maneerat; Neil A. Turner; Karen E. Porter

Coronary heart disease (CHD) is a condition characterized by increased levels of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α). TNF-α can induce vascular endothelial cell (EC) and smooth muscle cell (SMC) dysfunction, central events in development of neointimal lesions. The reduced incidence of CHD in young women is believed to be due to the protective effects of estradiol (E2). We therefore investigated the effects of TNF-α on human neointima formation and SMC/EC functions and any modulatory effects of E2. Saphenous vein (SV) segments were cultured in the presence of TNF-α (10 ng/ml), E2 (2.5 nM) or both in combination. Neointimal thickening was augmented by incubation with TNF-α, an effect that was abolished by co-culture with E2. TNF-α increased SV-SMC proliferation in a concentration-dependent manner that was optimal at 10 ng/ml (1.5-fold increase), and abolished by E2 at all concentrations studied (1-50 nM). Surprisingly, E2 itself at low concentrations (1 and 5 nM) stimulated SV-SMC proliferation to a level comparable to that of TNF-α alone. SV-EC migration was significantly impaired by TNF-α (42% of control), and co-culture with E2 partially restored the ability of SV-EC to migrate and repair the wound. In contrast, TNF-α increased SV-SMC migration by 1.7-fold, an effect that was completely reversed by co-incubation with E2. Finally, TNF-α potently induced ICAM-1 and VCAM-1 expression in both SV-EC and SV-SMC. However there was no modulation by E2 in either cell-type. In conclusion, TNF-α induced SV neointima formation, increased SMC proliferation and migration, impaired SV-EC migration and increased expression of adhesion molecules. E2 exerted distinct cell-type and function-specific modulation, the mechanisms underlying which are worthy of further detailed study.


Endocrinology | 2018

Insulinlike growth factor – binding protein-1 improves vascular endothelial repair in male mice in the setting of insulin resistance

Amir Aziz; Natalie Haywood; Paul A. Cordell; J Smith; Nadira Yuldasheva; Anshuman Sengupta; Noman Ali; Ben Mercer; Romana S Mughal; Kirsten Riches; Richard M. Cubbon; Karen E. Porter; Mark T. Kearney; Stephen B. Wheatcroft

Insulin resistance is associated with impaired endothelial regeneration in response to mechanical injury. We recently demonstrated that insulinlike growth factor-binding protein-1 (IGFBP1) ameliorated insulin resistance and increased nitric oxide generation in the endothelium. In this study, we hypothesized that IGFBP1 would improve endothelial regeneration and restore endothelial reparative functions in the setting of insulin resistance. In male mice heterozygous for deletion of insulin receptors, endothelial regeneration after femoral artery wire injury was enhanced by transgenic expression of human IGFBP1 (hIGFBP1). This was not explained by altered abundance of circulating myeloid angiogenic cells. Incubation of human endothelial cells with hIGFBP1 increased integrin expression and enhanced their ability to adhere to and repopulate denuded human saphenous vein ex vivo. In vitro, induction of insulin resistance by tumor necrosis factor α (TNFα) significantly inhibited endothelial cell migration and proliferation. Coincubation with hIGFBP1 restored endothelial migratory and proliferative capacity. At the molecular level, hIGFBP1 induced phosphorylation of focal adhesion kinase, activated RhoA and modulated TNFα-induced actin fiber anisotropy. Collectively, the effects of hIGFBP1 on endothelial cell responses and acceleration of endothelial regeneration in mice indicate that manipulating IGFBP1 could be exploited as a putative strategy to improve endothelial repair in the setting of insulin resistance.


Archive | 2012

C-Peptide: Connecting Diabetes with Macrovascular Complications

Karen E. Porter; Romana S Mughal

The incidence of diabetes is increasing globally; the estimated prevalence in 2010 being 285 million worldwide (www.http://diabetes.org.uk). In the UK alone, the number of people diagnosed with diabetes has increased from 1.4 to 2.6 million since 1996 (www.http://diabetes.org.uk). These individuals are at high risk of cardiovascular disease and in this population coronary artery disease (CAD) is common, regularly manifesting itself earlier in life than in individuals without diabetes [1]. While strategies that maintain tight glucose control have been reported to ameliorate some of the microvascular complications of diabetes (retinopathy, nephropathy, neuropathy) [2], this approach appears to have little or no benefit in the macrovasculature [3]. A greater understanding of the complexity of the diabetic milieu may therefore be invaluable in exposing therapeutic targets to reduce the incidence of macrovascular diseases in diabetes.


Endocrinology | 2018

Endothelial Insulin Receptor Restoration Rescues Vascular Function in Male Insulin Receptor Haploinsufficient Mice

Anshuman Sengupta; Peysh A Patel; Nadira Yuldasheva; Romana S Mughal; S Galloway; Hema Viswambharan; Andrew M. Walker; Amir Aziz; Jessica Smith; Noman Ali; Ben Mercer; Helen Imrie; Piruthivi Sukumar; Stephen B. Wheatcroft; Mark T. Kearney; Richard M. Cubbon

Abstract Reduced systemic insulin signaling promotes endothelial dysfunction and diminished endogenous vascular repair. We investigated whether restoration of endothelial insulin receptor expression could rescue this phenotype. Insulin receptor knockout (IRKO) mice were crossed with mice expressing a human insulin receptor endothelial cell–specific overexpression (hIRECO) to produce IRKO-hIRECO progeny. No metabolic differences were noted between IRKO and IRKO-hIRECO mice in glucose and insulin tolerance tests. In contrast with control IRKO littermates, IRKO-hIRECO mice exhibited normal blood pressure and aortic vasodilatation in response to acetylcholine, comparable to parameters noted in wild type littermates. These phenotypic changes were associated with increased basal- and insulin-stimulated nitric oxide production. IRKO-hIRECO mice also demonstrated normalized endothelial repair after denuding arterial injury, which was associated with rescued endothelial cell migration in vitro but not with changes in circulating progenitor populations or culture-derived myeloid angiogenic cells. These data show that restoration of endothelial insulin receptor expression alone is sufficient to prevent the vascular dysfunction caused by systemically reduced insulin signaling.


Diabetes and Vascular Disease Research | 2018

Effects of obesity on insulin: insulin-like growth factor 1 hybrid receptor expression and Akt phosphorylation in conduit and resistance arteries

Romana S Mughal; Katherine I. Bridge; Irma Buza; Rita Slaaby; Jesper Worm; Gro Klitgaard-Povlsen; Henning Hvid; Marianne Schiødt; Richard M. Cubbon; Nadira Yuldasheva; A Skromna; Natallia Makava; Grith Skytte-Olsen; Mark T. Kearney

Insulin and insulin-like growth factor-1 stimulate specific responses in arteries, which may be disrupted by diet-induced obesity. We examined (1) temporal effects of high-fat diet compared to low-fat diet in mice on insulin receptor, insulin-like growth factor-1 receptor, insulin receptor/insulin-like growth factor-1 receptor hybrid receptor expression and insulin/insulin-like growth factor-1-mediated Akt phosphorylation in aorta; and (2) effects of high-fat diet on insulin and insulin-like growth factor-1-mediated Akt phosphorylation and vascular tone in resistance arteries. Medium-term high-fat diet (5 weeks) decreased insulin-like growth factor-1 receptor expression and increased hybrid expression (~30%) only. After long-term (16 weeks) high-fat diet, insulin receptor expression was reduced by ~30%, insulin-like growth factor-1 receptor expression decreased a further ~40% and hybrid expression increased a further ~60%. Independent correlates of hybrid receptor expression were high-fat diet, duration of high-fat diet and plasma insulin-like growth factor-1 (all p < 0.05). In aorta, insulin was a more potent activator of Akt than insulin-like growth factor-1, whereas in resistance arteries, insulin-like growth factor-1 was more potent than insulin. High-fat diet blunted insulin-mediated vasorelaxation (p < 0.01) but had no effect on insulin-like growth factor-1-mediated vasorelaxation in resistance arteries. Our findings support the possibility that hybrid receptor level is influenced by nutritional and metabolic cues. Moreover, vessel-dependent effects of insulin and insulin-like growth factor-1 on vascular tone and Akt activation may have implications in treating obesity-related vascular disease.


Heart | 2016

P41 Exploring the biological role of insulin and insulin-like growth factor-1 hybrid receptors in the endothelium

Romana S Mughal; Jesper Worm; Rita Slaaby; K Paradine; Nadira Yuldasheva; Marianne Schiødt; Gk Povlsen; Henning Hvid; Gs Olsen; Mark T. Kearney

Endothelial cells co-express insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF-1R) homodimers. Furthermore, association of insulin αβ half receptors and IGF-1 αβ half receptors also occur resulting in insulin/IGF-1 hybrid receptors. An abundance of hybrid receptors has been associated with reduced insulin sensitivity in metabolic tissue. The aim of this project is to study the differential expression of vascular derived insulin receptors, IGF-1 receptors and insulin/IGF-1 hybrids during insulin resistance in a diet-induced model of obesity (DIO). C57BL/6 mice were fed on high-fat or low-fat diets ad libitum for 16-weeks. Obese mice showed significant metabolic abnormalities indicative of insulin resistance when compared with lean littermates. A significant increase in hybrid receptor levels were observed in aorta from obese mice (immunoprecipitation and Western blotting). Total IR and IGF-1R protein levels were reduced in obese mice (Western blotting) however, no change was observed in IR and IGF-1R mRNA levels (real time-PCR). Insulin-stimulated (4.5 nmol/kg injected) in vivo serine phosphorylation of Akt was significantly blunted in aortae of obese mice, whereas IGF-1-stimulated (90 nmol/kg injected) remained unaffected. We have shown that IR and IGF1R homodimers are preferentially titrated into hybrid receptors in a model of DIO and this is accompanied with a reduction in insulin sensitivity but not IGF-1 sensitivity in the aorta. Hybrid receptors may provide a novel target for the prevention of CVD progression in vulnerable patients.

Collaboration


Dive into the Romana S Mughal's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge