Network


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

Hotspot


Dive into the research topics where Ruth A. Roberts is active.

Publication


Featured researches published by Ruth A. Roberts.


Journal of Toxicology | 2012

Drug-Induced Oxidative Stress and Toxicity

Damian G. Deavall; Elizabeth A. Martin; Judith Horner; Ruth A. Roberts

Reactive oxygen species (ROS) are a byproduct of normal metabolism and have roles in cell signaling and homeostasis. Species include oxygen radicals and reactive nonradicals. Mechanisms exist that regulate cellular levels of ROS, as their reactive nature may otherwise cause damage to key cellular components including DNA, protein, and lipid. When the cellular antioxidant capacity is exceeded, oxidative stress can result. Pleiotropic deleterious effects of oxidative stress are observed in numerous disease states and are also implicated in a variety of drug-induced toxicities. In this paper, we examine the nature of ROS-induced damage on key cellular targets of oxidative stress. We also review evidence implicating ROS in clinically relevant, drug-related side effects including doxorubicin-induced cardiac damage, azidothymidine-induced myopathy, and cisplatin-induced ototoxicity.


Toxicological Sciences | 2009

Nitrative and Oxidative Stress in Toxicology and Disease

Ruth A. Roberts; Debra L. Laskin; Charles V. Smith; Fredika M. Robertson; Erin M. G. Allen; Jonathan A. Doorn; William Slikker

Persistent inflammation and the generation of reactive oxygen and nitrogen species play pivotal roles in tissue injury during disease pathogenesis and as a reaction to toxicant exposures. The associated oxidative and nitrative stress promote diverse pathologic reactions including neurodegenerative disorders, atherosclerosis, chronic inflammation, cancer, and premature labor and stillbirth. These effects occur via sustained inflammation, cellular proliferation and cytotoxicity and via induction of a proangiogenic environment. For example, exposure to the ubiquitous air pollutant ozone leads to generation of reactive oxygen and nitrogen species in lung macrophages that play a key role in subsequent tissue damage. Similarly, studies indicate that genes involved in regulating oxidative stress are altered by anesthetic treatment resulting in brain injury, most notable during development. In addition to a role in tissue injury in the brain, inflammation, and oxidative stress are implicated in Parkinsons disease, a neurodegenerative disease characterized by the loss of dopamine neurons. Recent data suggest a mechanistic link between oxidative stress and elevated levels of 3,4-dihydroxyphenylacetaldehyde, a neurotoxin endogenous to dopamine neurons. These findings have significant implications for development of therapeutics and identification of novel biomarkers for Parkinsons disease pathogenesis. Oxidative and nitrative stress is also thought to play a role in creating the proinflammatory microenvironment associated with the aggressive phenotype of inflammatory breast cancer. An understanding of fundamental concepts of oxidative and nitrative stress can underpin a rational plan of treatment for diseases and toxicities associated with excessive production of reactive oxygen and nitrogen species.


Toxicological Sciences | 2011

Cardiotoxicity Associated with Targeting Kinase Pathways in Cancer

Howard R. Mellor; Alex R. Bell; Jean-Pierre Valentin; Ruth A. Roberts

Cardiotoxicity, also referred to as drug-induced cardiac injury, is an issue associated with the use of some small-molecule kinase inhibitors and antibody-based therapies targeting signaling pathways in cancer. Although these drugs have had a major impact on cancer patient survival, data have implicated kinase-targeting agents such as sunitinib, imatinib, trastuzumab, and sorafenib in adversely affecting cardiac function in a subset of treated individuals. In many cases, adverse cardiac events in the clinic were not anticipated based on preclinical safety evaluation of the molecule. In order to support the development of efficacious and safe kinase inhibitors for the treatment of cancer and other indications, new preclinical approaches and screens are required to predict clinical cardiotoxicity. Laboratory investigations into the underlying molecular mechanisms of heart toxicity induced by these molecules have identified potentially common themes including mitochondrial perturbation and modulation of adenosine monophosphate-activated protein kinase activity. Studies characterizing cardiac-specific kinase knockout mouse models have developed our understanding of the homeostatic role of some of these signaling mediators in the heart. Therefore, when considering kinases as potential future targets or when examining secondary pharmacological interactions of novel kinase inhibitors, these models may help to inform us of the potential adverse cardiac effects in the clinic.


Toxicologic Pathology | 2011

Induction of Heart Valve Lesions by Small-Molecule ALK5 Inhibitors

Mark J. Anderton; Howard R. Mellor; Alex R. Bell; Claire Sadler; Martin Pass; Steve Powell; Samantha J. Steele; Ruth A. Roberts; Annabelle Heier

Aberrant signaling by transforming growth factor-β (TGF-β) and its type I (ALK5) receptor has been implicated in a number of human diseases and this pathway is considered a potential target for therapeutic intervention. Transforming growth factor-β signaling via ALK5 plays a critical role during heart development, but the role of ALK5 in the adult heart is poorly understood. In the current study, the preclinical toxicology of ALK5 inhibitors from two different chemistry scaffolds was explored. Ten-week-old female Han Wistar rats received test compounds by the oral route for three to seven days. Both compounds induced histopathologic heart valve lesions characterized by hemorrhage, inflammation, degeneration, and proliferation of valvular interstitial cells. The pathology was observed in all animals, at all doses tested, and occurred in all four heart valves. Immunohistochemical analysis of ALK5 in rat hearts revealed expression in the valves, but not in the myocardium. Compared to control animals, protein levels of ALK5 were unchanged in the heart valves of treated animals. We also observed a physeal dysplasia in the femoro-tibial joint of rats treated with ALK5 inhibitors, a finding consistent with a pharmacological effect described previously with ALK5 inhibitors. Overall, these findings suggest that TGF-β signaling via ALK5 plays a critical role in maintaining heart valve integrity.


Trends in Pharmacological Sciences | 2010

Gender differences in drug toxicity

Tamara J Nicolson; Howard R. Mellor; Ruth A. Roberts

Clinical data suggest that gender dimorphic profiles are emerging in terms of both drug efficacy and adverse drug reactions (ADRs). With an increasing emphasis on individualised therapies and the need to prevent drug attrition there is a compelling need to understand the molecular basis for gender dimorphic profiles in ADRs and the consequences. Classes of agents exhibiting gender-based variation in pharmaceutical efficacy and toxicity include anaesthetics, HIV-1 therapies and antiarrhythmic drugs. Body weight differences are often cited as a reason for differences in drug pharmacokinetics and subsequent toxicity. However, some studies accounted for these factors and still found significance suggesting that dosage versus body weight does not explain the outcome. Here, we present an overview of current understanding of gender-specific drug toxicity and present rational molecular explanations for these adverse events. There is mounting evidence in support of hormonal effects underpinning the majority of the ADR differences observed between the sexes.


Toxicology Letters | 2000

Apoptosis and proliferation in nongenotoxic carcinogenesis: species differences and role of PPARα

Ruth A. Roberts; Neil H. James; Susan C. Hasmall; Peter R. Holden; K.G Lambe; Neil Macdonald; Douglas West; N.J Woodyatt; D Whitcome

Peroxisome proliferators (PPs) are nongenotoxic rodent hepatocarcinogens that cause liver enlargement and hepatocarcinogenesis associated with peroxisome proliferation, induction of hepatocyte DNA synthesis and suppression of apoptosis. Acyl CoA oxidase (ACO) is a key enzyme of peroxisomal beta-oxidation and its transcriptional activation by PPs is often used as marker for the rodent response. PPs activate the peroxisome proliferator activated receptor-alpha, PPARalpha. Recent data suggest a role for tumour necrosis factor alpha (TNFalpha). This cytokine appears to be permissive for a PPARalpha-dependent growth response to PPs. Humans and guinea pigs appear to be nonresponsive to the adverse effects of PPs noted in rodents. These species differences can be attributed to reduced quantity of full length functional PPARalpha in human liver and evidence supports the presence of a truncated form of PPARalpha, hPPARalpha8/14 in human liver. In addition, species differences could be attributed to qualitative differences in the PPARalpha-mediated response because the promoter for human ACO differs in sequence and activity from the rat equivalent. These data contribute to our understanding of how chemicals may cause tumours in rodents and how this response may differ in humans.


Archives of Toxicology | 2000

Species differences in response to diethylhexylphthalate: suppression of apoptosis, induction of DNA synthesis and peroxisome proliferator activated receptor alpha-mediated gene expression.

Susan C. Hasmall; Neil H. James; Neil Macdonald; Anthony R. Soames; Ruth A. Roberts

Abstract Diethylhexylphthalate (DEHP) is a phthalate plasticizer that belongs to the peroxisome proliferator (PP) class of rodent nongenotoxic hepatocarcinogens. Previously, we have shown that MEHP (a principal metabolite of DEHP and the proximal PP) induced DNA synthesis and suppressed apoptosis in rat but not in human hepatocytes in vitro. Here, we present further studies of species differences in response to DEHP. In rats, 4 days of exposure to DEHP (950 mg/kg per day by gavage) induced peroxisomal β-oxidation, DNA synthesis and suppressed apoptosis. In contrast, there was no response of guinea pig liver to DEHP. In rat hepatocytes in vitro, MEHP (250, 500 and 750 μM) induced peroxisomal β-oxidation, DNA synthesis and suppressed apoptosis. In contrast to the pleiotropic response noted in rat hepatocytes, there was no response of human hepatocytes to 250, 500 or 750 μM MEHP. PPs activate the peroxisome proliferator activated receptor alpha (PPARα) that binds to DNA at peroxisome proliferator response elements (PPREs) within the promoters of PP-responsive genes such as rat acyl CoA oxidase (ACO). However, the human ACO gene promoter differs at three bases within the PPRE from the rat ACO promoter and appears refractory to PPs. To address species differences in response to DEHP at the molecular level, we used promoter-reporter gene assays to compare the ability of MEHP to induce gene expression from the rat or the human ACO promoter. MEHP gave a concentration-dependent increase in reporter gene expression from the rat ACO gene promoter with either mouse or human PPARα. In contrast, the human ACO promoter was unable to drive MEHP-induced gene transcription irrespective of the species origin of PPARα. These data provide further weight of evidence at the cellular and molecular levels for a lack of risk to human health from the phthalate DEHP.


Archives of Toxicology | 1999

Peroxisome proliferators: mechanisms of adverse effects in rodents and molecular basis for species differences

Ruth A. Roberts

Abstract Peroxisome proliferators (PPs), such as diethylhexylphthalate (DEHP), constitute a diverse class of chemicals with many therapeutic, industrial and environmental applications. In rodents, PPs are nongenotoxic hepatocarcinogens, raising concerns regarding the potential of PPs to harm human health. However, humans differ from rodents in their response to PPs and the weight of evidence supports the supposition that PPs do not pose a carcinogenic risk to humans. The effects of PPs in the rodent are mediated by peroxisome proliferator activated receptor α (PPARα). PPARα predominates in the liver whereas another isoform PPARγ predominates in adipose tissue and in the immune system. This tissue-specific pattern of PPARα expression is consistent with a role for PPARα but not PPARγ or PPARβ in PP-induced rodent hepatocarcinogenesis. Humans, marmosets and guinea-pigs appear refractory or less responsive to the adverse liver effects of PPs. However, humans give a therapeutic response to the fibrate PPs via an alteration in lipid metabolism mediated by PPARα. Such marked species differences may be explained by quantity of PPARα and/or the quality of the PPARα-mediated response. The lower expression of full-length functional PPARα in humans could be attributed to the presence of a truncated, inactive form of PPARα, which appears to be present in most individuals examined to date. In addition, there are species differences in sequence and responsiveness of the acyl CoA oxidase (ACO) gene promoter, suggesting that even in the presence of sufficient PPARα, the human equivalent of rodent genes associated with peroxisome proliferation may remain inactive.


Mutation Research | 2000

Suppression of mouse hepatocyte apoptosis by peroxisome proliferators : role of PPARα and TNFα

Susan C. Hasmall; Neil H. James; Neil Macdonald; Frank J. Gonzalez; Jeffrey M. Peters; Ruth A. Roberts

Abstract Peroxisome proliferators (PPs) are a diverse group of nongenotoxic chemicals that in rodents cause hepatic peroxisome proliferation, liver enlargement, increased replicative DNA synthesis and suppression of apoptosis. The effects of PPs in vivo can be reproduced in vitro where PPs can induce mouse hepatocyte DNA synthesis and suppress both spontaneous apoptosis and that induced by transforming growth factor β (TGFβ). In vitro, high concentrations (>500 U/ml) of exogenous tumour necrosis factor α (TNFα) [M. Rolfe, N.H. James, R.A. Roberts, TNFα suppresses apoptosis and induces S-phase in rodent hepatocytes: a mediator of the hepatocarcinogenicity of peroxisome proliferators?, Carcinogenesis 18 (1997) 2277–2280] are also able to stimulate hepatocyte DNA synthesis and suppress apoptosis, implicating TNFα in mediating or permitting the liver growth response to PPs. Here, using cultured mouse hepatocytes isolated from PPARα null mice, we have examined the role of the peroxisome proliferator activated receptor α (PPARα) in mediating the suppression of apoptosis caused by PPs. In addition we have investigated further the role of TNFα in mediating the rodent response to PPs. The PP nafenopin (50 μM) was unable to stimulate DNA synthesis measured by bromodeoxyuridine incorporation in these PPARα null mouse hepatocytes (96% of control), unlike epidermal growth factor, a growth factor used as a positive control. In assays of apoptosis using H33258 staining of chromatin condensation, nafenopin was unable to suppress either spontaneous or TGFβ1-induced apoptosis. In contrast, high concentrations of TNFα (>500 U/ml) were able to both stimulate DNA synthesis (204% of control) and suppress apoptosis in PPARα null hepatocytes (40% and 38% of control for spontaneous and TGFβ1-induced apoptosis respectively). However, TNFα could not stimulate β-oxidation of palmitoyl CoA in either PPARα null mouse or B6C3F1 (PPARα wild type) mouse hepatocytes. These data confirm the dependence of the response to PPs on PPARα by demonstrating that PPARα mediates the suppression of hepatocyte apoptosis in response to PPs. In addition, the data provide evidence that high concentrations of TNFα can modulate DNA synthesis and apoptosis in the absence of PPs and PPARα. Thus, in vivo, physiological levels of TNFα may be permissive for a PPARα-dependent growth response to PPs.


Toxicology | 2002

PPARα and the regulation of cell division and apoptosis

Ruth A. Roberts; S Chevalier; S.C. Hasmall; Neil H. James; Sabina Cosulich; N Macdonald

Peroxisome proliferators (PPs) such as the hypolipidaemic drug, nafenopin and the phthalate plasticiser 2-diethylhexylphthalate induce rodent hepatocyte cell proliferation and suppress apoptosis leading to tumours. PPs act via the nuclear hormone receptor peroxisome proliferator activated receptor α (PPARα) which directly regulates genes implicated in the response to PPs such as the peroxisomal gene acyl CoA oxidase. As expected for xenobiotics that perturb proliferation, PPs alter expression of cell cycle regulatory proteins. However, the ability to alter expression of cyclins and cyclin-dependent kinases is shared by physiological hepatic mitogens such as epidermal growth factor and is thus unlikely to be specific to the PP-induced aberrant growth associated with hepatocarcinogenesis. Recent evidence suggests that the response of hepatocytes to PPs is not only dependent upon PPARα but also on the trophic environment provided by nonparenchymal cells and by cytokines such as tumour necrosis factor α. Additionally, the ability of PPs to suppress apoptosis and induce proliferation depends upon survival signalling mediated by p38 mitogen activated protein kinase. The cross talk between PPARα-mediated transcription, survival signalling and cell cycle will be discussed with particular emphasis on relevance to toxicology.

Collaboration


Dive into the Ruth A. Roberts's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Caroline Dive

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian Kimber

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge