Network


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

Hotspot


Dive into the research topics where Violetta Steeples is active.

Publication


Featured researches published by Violetta Steeples.


PLOS Genetics | 2010

A mutation in the mitochondrial fission gene Dnm1l leads to cardiomyopathy

Houman Ashrafian; Louise Docherty; Vincenzo C. Leo; Christopher Towlson; Monica Neilan; Violetta Steeples; Craig A. Lygate; Tertius Hough; Stuart Townsend; Debbie Williams; Sara Wells; Dominic P. Norris; Sarah Glyn-Jones; John M. Land; Ivana Barbaric; Zuzanne Lalanne; Paul Denny; Dorota Szumska; Shoumo Bhattacharya; Julian L. Griffin; Iain Hargreaves; Narcis Fernandez-Fuentes; Michael Cheeseman; Hugh Watkins; T. Neil Dear

Mutations in a number of genes have been linked to inherited dilated cardiomyopathy (DCM). However, such mutations account for only a small proportion of the clinical cases emphasising the need for alternative discovery approaches to uncovering novel pathogenic mutations in hitherto unidentified pathways. Accordingly, as part of a large-scale N-ethyl-N-nitrosourea mutagenesis screen, we identified a mouse mutant, Python, which develops DCM. We demonstrate that the Python phenotype is attributable to a dominant fully penetrant mutation in the dynamin-1-like (Dnm1l) gene, which has been shown to be critical for mitochondrial fission. The C452F mutation is in a highly conserved region of the M domain of Dnm1l that alters protein interactions in a yeast two-hybrid system, suggesting that the mutation might alter intramolecular interactions within the Dnm1l monomer. Heterozygous Python fibroblasts exhibit abnormal mitochondria and peroxisomes. Homozygosity for the mutation results in the death of embryos midway though gestation. Heterozygous Python hearts show reduced levels of mitochondria enzyme complexes and suffer from cardiac ATP depletion. The resulting energy deficiency may contribute to cardiomyopathy. This is the first demonstration that a defect in a gene involved in mitochondrial remodelling can result in cardiomyopathy, showing that the function of this gene is needed for the maintenance of normal cellular function in a relatively tissue-specific manner. This disease model attests to the importance of mitochondrial remodelling in the heart; similar defects might underlie human heart muscle disease.


Cardiovascular Research | 2011

The role of vascular myoglobin in nitrite-mediated blood vessel relaxation

Julian O.M. Ormerod; Houman Ashrafian; Abdul R. Maher; Sayqa Arif; Violetta Steeples; Gustav V.R. Born; Stuart Egginton; Martin Feelisch; Hugh Watkins; Michael P. Frenneaux

Aims This work investigates the role of myoglobin in mediating the vascular relaxation induced by nitrite. Nitrite, previously considered an inert by-product of nitric oxide metabolism, is now believed to play an important role in several areas of pharmacology and physiology. Myoglobin can act as a nitrite reductase in the heart, where it is plentiful, but it is present at a far lower level in vascular smooth muscle—indeed, its existence in the vessel wall is controversial. Haem proteins have been postulated to be important in nitrite-induced vasodilation, but the specific role of myoglobin is unknown. The current study was designed to confirm the presence of myoglobin in murine aortic tissue and to test the hypothesis that vascular wall myoglobin is important for nitrite-induced vasodilation. Methods and results Aortic rings from wild-type and myoglobin knockout mice were challenged with nitrite, before and after exposure to the haem-protein inhibitor carbon monoxide (CO). CO inhibited vasodilation in wild-type rings but not in myoglobin-deficient rings. Restitution of myoglobin using a genetically modified adenovirus both increased vasodilation to nitrite and reinstated the wild-type pattern of response to CO. Conclusion Myoglobin is present in the murine vasculature and contributes significantly to nitrite-induced vasodilation.


Cell Metabolism | 2016

Chronic Activation of γ2 AMPK Induces Obesity and Reduces β Cell Function

Arash Yavari; Claire J. Stocker; Sahar Ghaffari; Edward T. Wargent; Violetta Steeples; Gabor Czibik; Katalin Pinter; Mohamed Bellahcene; Angela Woods; Pablo Blanco Martinez de Morentin; Celine Cansell; Brian Yee Hong Lam; André Chuster; Kasparas Petkevicius; Marie-Sophie Nguyen-Tu; Aida Martinez-Sanchez; Timothy J. Pullen; Peter L. Oliver; A Stockenhuber; Chinh Nguyen; Merzaka Lazdam; Jacqueline F. O’Dowd; Parvathy E. Harikumar; Mónika Tóth; Craig Beall; Theodosios Kyriakou; Julia Parnis; Dhruv Sarma; George Katritsis; Diana D.J. Wortmann

Summary Despite significant advances in our understanding of the biology determining systemic energy homeostasis, the treatment of obesity remains a medical challenge. Activation of AMP-activated protein kinase (AMPK) has been proposed as an attractive strategy for the treatment of obesity and its complications. AMPK is a conserved, ubiquitously expressed, heterotrimeric serine/threonine kinase whose short-term activation has multiple beneficial metabolic effects. Whether these translate into long-term benefits for obesity and its complications is unknown. Here, we observe that mice with chronic AMPK activation, resulting from mutation of the AMPK γ2 subunit, exhibit ghrelin signaling-dependent hyperphagia, obesity, and impaired pancreatic islet insulin secretion. Humans bearing the homologous mutation manifest a congruent phenotype. Our studies highlight that long-term AMPK activation throughout all tissues can have adverse metabolic consequences, with implications for pharmacological strategies seeking to chronically activate AMPK systemically to treat metabolic disease.


Leukemia | 2016

Cryptic splicing events in the iron transporter ABCB7 and other key target genes in SF3B1 -mutant myelodysplastic syndromes

Hamid Dolatshad; Andrea Pellagatti; Fabio Liberante; Miriam Llorian; Emmanouela Repapi; Violetta Steeples; Swagata Roy; L Scifo; Richard N. Armstrong; J Shaw; Bon Ham Yip; Sally Killick; Rajko Kusec; Stephen Taylor; Ken I. Mills; Kienan Savage; Christopher W. J. Smith; Jacqueline Boultwood

The splicing factor SF3B1 is the most frequently mutated gene in myelodysplastic syndromes (MDS), and is strongly associated with the presence of ring sideroblasts (RS). We have performed a systematic analysis of cryptic splicing abnormalities from RNA sequencing data on hematopoietic stem cells (HSCs) of SF3B1-mutant MDS cases with RS. Aberrant splicing events in many downstream target genes were identified and cryptic 3′ splice site usage was a frequent event in SF3B1-mutant MDS. The iron transporter ABCB7 is a well-recognized candidate gene showing marked downregulation in MDS with RS. Our analysis unveiled aberrant ABCB7 splicing, due to usage of an alternative 3′ splice site in MDS patient samples, giving rise to a premature termination codon in the ABCB7 mRNA. Treatment of cultured SF3B1-mutant MDS erythroblasts and a CRISPR/Cas9-generated SF3B1-mutant cell line with the nonsense-mediated decay (NMD) inhibitor cycloheximide showed that the aberrantly spliced ABCB7 transcript is targeted by NMD. We describe cryptic splicing events in the HSCs of SF3B1-mutant MDS, and our data support a model in which NMD-induced downregulation of the iron exporter ABCB7 mRNA transcript resulting from aberrant splicing caused by mutant SF3B1 underlies the increased mitochondrial iron accumulation found in MDS patients with RS.


Journal of Biological Chemistry | 2015

Resistance of dynamin-related protein 1 oligomers to disassembly impairs mitophagy, resulting in myocardial inflammation and heart failure.

Thomas J. Cahill; Vincenzo C. Leo; Matthew Kelly; A Stockenhuber; Nolan W. Kennedy; Leyuan Bao; G Cereghetti; Andrew R. Harper; Gabor Czibik; C Lao; Mohamed Bellahcene; Violetta Steeples; Sahar Ghaffari; Arash Yavari; Alice Mayer; Joanna Poulton; Ferguson Djp.; Luca Scorrano; Nishani T. Hettiarachchi; Chris Peers; John P. Boyle; R B Hill; Alison Simmons; Hugh Watkins; T N Dear; Houman Ashrafian

Background: The C452F mutation in the mitochondrial fission protein Drp1 leads to heart failure through an unknown mechanism. Results: C452F impairs Drp1 disassembly, leading to impaired mitophagy, failed bioenergetics, and inflammation. Conclusion: Drp1-mediated mitochondrial fission is essential for normal cardiac function. Significance: Mutations in mitochondrial quality control proteins are a likely cause of human cardiomyopathy. We have reported previously that a missense mutation in the mitochondrial fission gene Dynamin-related protein 1 (Drp1) underlies the Python mouse model of monogenic dilated cardiomyopathy. The aim of this study was to investigate the consequences of the C452F mutation on Drp1 protein function and to define the cellular sequelae leading to heart failure in the Python monogenic dilated cardiomyopathy model. We found that the C452F mutation increased Drp1 GTPase activity. The mutation also conferred resistance to oligomer disassembly by guanine nucleotides and high ionic strength solutions. In a mouse embryonic fibroblast model, Drp1 C452F cells exhibited abnormal mitochondrial morphology and defective mitophagy. Mitochondria in C452F mouse embryonic fibroblasts were depolarized and had reduced calcium uptake with impaired ATP production by oxidative phosphorylation. In the Python heart, we found a corresponding progressive decline in oxidative phosphorylation with age and activation of sterile inflammation. As a corollary, enhancing autophagy by exposure to a prolonged low-protein diet improved cardiac function in Python mice. In conclusion, failure of Drp1 disassembly impairs mitophagy, leading to a downstream cascade of mitochondrial depolarization, aberrant calcium handling, impaired ATP synthesis, and activation of sterile myocardial inflammation, resulting in heart failure.


Journal of Clinical Investigation | 2017

The U2AF1S34F mutation induces lineage-specific splicing alterations in myelodysplastic syndromes

Bon Ham Yip; Violetta Steeples; Emmanouela Repapi; Richard N. Armstrong; Miriam Llorian; Swagata Roy; J Shaw; Hamid Dolatshad; Stephen Taylor; Amit Verma; Matthias Bartenstein; Paresh Vyas; Nicholas C.P. Cross; Luca Malcovati; Mario Cazzola; Eva Hellström-Lindberg; Seishi Ogawa; Christopher W. J. Smith; Andrea Pellagatti; Jacqueline Boultwood

Mutations of the splicing factor–encoding gene U2AF1 are frequent in the myelodysplastic syndromes (MDS), a myeloid malignancy, and other cancers. Patients with MDS suffer from peripheral blood cytopenias, including anemia, and an increasing percentage of bone marrow myeloblasts. We studied the impact of the common U2AF1S34F mutation on cellular function and mRNA splicing in the main cell lineages affected in MDS. We demonstrated that U2AF1S34F expression in human hematopoietic progenitors impairs erythroid differentiation and skews granulomonocytic differentiation toward granulocytes. RNA sequencing of erythroid and granulomonocytic colonies revealed that U2AF1S34F induced a higher number of cassette exon splicing events in granulomonocytic cells than in erythroid cells. U2AF1S34F altered mRNA splicing of many transcripts that were expressed in both cell types in a lineage-specific manner. In hematopoietic progenitors, the introduction of isoform changes identified in the U2AF1S34F target genes H2AFY, encoding an H2A histone variant, and STRAP, encoding serine/threonine kinase receptor–associated protein, recapitulated phenotypes associated with U2AF1S34F expression in erythroid and granulomonocytic cells, suggesting a causal link. Furthermore, we showed that isoform modulation of H2AFY and STRAP rescues the erythroid differentiation defect in U2AF1S34F MDS cells, suggesting that splicing modulators could be used therapeutically. These data have critical implications for understanding MDS phenotypic heterogeneity and support the development of therapies targeting splicing abnormalities.


Circulation-cardiovascular Genetics | 2014

Citric Acid Cycle Intermediates in Cardioprotection

Gabor Czibik; Violetta Steeples; Arash Yavari; Houman Ashrafian

Over the last decade, there has been a concerted clinical effort to deliver on the laboratory promise that a variety of maneuvers can profoundly increase cardiac tolerance to ischemia and/or reduce additional damage consequent upon reperfusion. Here we will review the proximity of the metabolic approach to clinical practice. Specifically, we will focus on how the citric acid cycle is involved in cardioprotection. Inspired by cross-fertilization between fundamental cancer biology and cardiovascular medicine, a set of metabolic observations have identified novel metabolic pathways, easily manipulable in man, which can harness metabolism to robustly combat ischemia-reperfusion injury.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Mutation of Fnip1 is associated with B-cell deficiency, cardiomyopathy, and elevated AMPK activity

Owen M. Siggs; A Stockenhuber; Mukta Deobagkar-Lele; Katherine R. Bull; Tanya L. Crockford; Bethany L. Kingston; Greg Crawford; Consuelo Anzilotti; Violetta Steeples; Sahar Ghaffari; Gabor Czibik; Mohamed Bellahcene; Hugh Watkins; Houman Ashrafian; Benjamin Davies; Angela Woods; David Carling; Arash Yavari; Bruce Beutler; Richard J. Cornall

Significance Cellular metabolism is tightly regulated by AMP-activated protein kinase (AMPK): the function of which is influenced by folliculin (FLCN), folliculin-interacting protein (FNIP)1, and FNIP2. FLCN is a known tumor-suppressor protein that is mutated in Birt–Hogg–Dubé syndrome, whereas FNIP1 and FNIP2 are binding partners of FLCN. Previous reports have suggested that the FLCN/FNIP1/FNIP2 complex acts a positive regulator of AMPK, whereas other reports suggest the opposite. Using a new mouse model of FNIP1 deficiency, our findings support the latter: we found that mutation of Fnip1 leads to B-cell deficiency and the development of a cardiomyopathy similar to mice and humans with gain-of-function mutations in AMPK. Folliculin (FLCN) is a tumor-suppressor protein mutated in the Birt–Hogg–Dubé (BHD) syndrome, which associates with two paralogous proteins, folliculin-interacting protein (FNIP)1 and FNIP2, forming a complex that interacts with the AMP-activated protein kinase (AMPK). Although it is clear that this complex influences AMPK and other metabolic regulators, reports of its effects have been inconsistent. To address this issue, we created a recessive loss-of-function variant of Fnip1. Homozygous FNIP1 deficiency resulted in profound B-cell deficiency, partially restored by overexpression of the antiapoptotic protein BCL2, whereas heterozygous deficiency caused a loss of marginal zone B cells. FNIP1-deficient mice developed cardiomyopathy characterized by left ventricular hypertrophy and glycogen accumulation, with close parallels to mice and humans bearing gain-of-function mutations in the γ2 subunit of AMPK. Concordantly, γ2-specific AMPK activity was elevated in neonatal FNIP1-deficient myocardium, whereas AMPK-dependent unc-51–like autophagy activating kinase 1 (ULK1) phosphorylation and autophagy were increased in FNIP1-deficient B-cell progenitors. These data support a role for FNIP1 as a negative regulator of AMPK.


Nature Communications | 2017

Mammalian γ2 AMPK regulates intrinsic heart rate

Arash Yavari; Mohamed Bellahcene; Annalisa Bucchi; S Sirenko; Katalin Pinter; Neil Herring; Julia Jeannine Jung; Kirill V. Tarasov; Emily J. Sharpe; Markus Wolfien; Gabor Czibik; Violetta Steeples; Sahar Ghaffari; C Nguyen; A Stockenhuber; Clair Jrs.; Christian Rimmbach; Y Okamoto; Da Yang; Min Wang; B D Ziman; J M Moen; Riordon; C Ramirez; M Paina; J. Lee; Jianzhi Zhang; Ismayil Ahmet; Michael G. Matt; Y S Tarasova

AMPK is a conserved serine/threonine kinase whose activity maintains cellular energy homeostasis. Eukaryotic AMPK exists as αβγ complexes, whose regulatory γ subunit confers energy sensor function by binding adenine nucleotides. Humans bearing activating mutations in the γ2 subunit exhibit a phenotype including unexplained slowing of heart rate (bradycardia). Here, we show that γ2 AMPK activation downregulates fundamental sinoatrial cell pacemaker mechanisms to lower heart rate, including sarcolemmal hyperpolarization-activated current (If) and ryanodine receptor-derived diastolic local subsarcolemmal Ca2+ release. In contrast, loss of γ2 AMPK induces a reciprocal phenotype of increased heart rate, and prevents the adaptive intrinsic bradycardia of endurance training. Our results reveal that in mammals, for which heart rate is a key determinant of cardiac energy demand, AMPK functions in an organ-specific manner to maintain cardiac energy homeostasis and determines cardiac physiological adaptation to exercise by modulating intrinsic sinoatrial cell behavior.AMPK regulates cellular energy balance using its γ subunit as an energy sensor of cellular AMP and ADP to ATP ratios. Here, the authors show that γ2 AMPK activation lowers heart rate by reducing the activity of pacemaker cells, whereas loss of γ2 AMPK increases heart rate and prevents the adaptive bradycardia of endurance training in mice.


Advances in biological regulation | 2017

Splicing factor mutations in the myelodysplastic syndromes: target genes and therapeutic approaches

Richard N. Armstrong; Violetta Steeples; Shalini Singh; Andrea Sanchi; Jacqueline Boultwood; Andrea Pellagatti

Mutations in splicing factor genes (SF3B1, SRSF2, U2AF1 and ZRSR2) are frequently found in patients with myelodysplastic syndromes (MDS), suggesting that aberrant spliceosome function plays a key role in the pathogenesis of MDS. Splicing factor mutations have been shown to result in aberrant splicing of many downstream target genes. Recent functional studies have begun to characterize the splicing dysfunction in MDS, identifying some key aberrantly spliced genes that are implicated in disease pathophysiology. These findings have led to the development of therapeutic strategies using splicing-modulating agents and rapid progress is being made in this field. Splicing inhibitors are promising agents that exploit the preferential sensitivity of splicing factor-mutant cells to these compounds. Here, we review the known target genes associated with splicing factor mutations in MDS, and discuss the potential of splicing-modulating therapies for these disorders.

Collaboration


Dive into the Violetta Steeples'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