Network


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

Hotspot


Dive into the research topics where Saul R. Powell is active.

Publication


Featured researches published by Saul R. Powell.


Circulation | 2010

Ubiquitin Proteasome Dysfunction in Human Hypertrophic and Dilated Cardiomyopathies

Jaime Predmore; Ping Wang; Frank Davis; Sarah Bartolone; Margaret V. Westfall; David B. Dyke; Francis Pagani; Saul R. Powell; Sharlene M. Day

Background— The ubiquitin proteasome system maintains a dynamic equilibrium of proteins and prevents accumulation of damaged and misfolded proteins, yet its role in human cardiac dysfunction is not well understood. The present study evaluated ubiquitin proteasome system function in human heart failure and hypertrophic cardiomyopathy (HCM). Methods and Results— Proteasome function was studied in human nonfailing donor hearts, explanted failing hearts, and myectomy samples from patients with HCM. Proteasome proteolytic activities were markedly reduced in failing and HCM hearts compared with nonfailing hearts (P<0.01). This activity was partially restored after mechanical unloading in failing hearts (P<0.01) and was significantly lower in HCM hearts with pathogenic sarcomere mutations than in those lacking these mutations (P<0.05). There were no changes in the protein content of ubiquitin proteasome system subunits (ie, 11S, 20S, and 19S) or in active-site labeling of the 20S proteolytic subunit β-5 among groups to explain decreased ubiquitin proteasome system activity in HCM and failing hearts. Examination of protein oxidation revealed that total protein carbonyls, 4-hydroxynonenylated proteins, and oxidative modification to 19S ATPase subunit Rpt 5 were increased in failing compared with nonfailing hearts. Conclusions— Proteasome activity in HCM and failing human hearts is impaired in the absence of changes in proteasome protein content or availability of proteolytic active sites. These data provide strong evidence that posttranslational modifications to the proteasome may account for defective protein degradation in human cardiomyopathies.


The FASEB Journal | 2011

Enhancement of proteasome function by PA28α overexpression protects against oxidative stress.

Jie Li; Saul R. Powell; Xuejun Wang

The principal function of the proteasome is targeted degradation of intracellular proteins. Proteasome dysfunction has been observed in experimental cardiomyopathies and implicated in human congestive heart failure. Measures to enhance proteasome proteolytic function are currently lacking but would be beneficial in testing the pathogenic role of proteasome dysfunction and could have significant therapeutic potential. The association of proteasome activator 28 (PA28) with the 20S proteasome may play a role in antigen processing. It is unclear, however, whether the PA28 plays any important role outside of antigen presentation, although up‐regulation of PA28 has been observed in certain types of cardiomyopathy. Here, we show that PA28α overexpression (PA28αOE) stabilized PA28β, increased 11S proteasomes, and enhanced the degradation of a previously validated proteasome surrogate substrate (GFPu) in cultured neonatal rat cardiomyocytes. PA28αOE significantly attenuated H2O2‐induced increases in the protein carbonyls and markedly suppressed apoptosis in cultured cardiomyocytes under basal conditions or when stressed by H2O2. We conclude that PA28αOE is sufficient to up‐regulate 11S proteasomes, enhance proteasome‐mediated removal of misfolded and oxidized proteins, and protect against oxidative stress in cardiomyocytes, providing a highly sought means to increase proteasomal degradation of abnormal cellular proteins.—Li, J., Powell, S. R., Wang, X. Enhancement of proteasome function by PA28α overexpression protects against oxidative stress. FASEB J. 25, 883–893 (2011). www.fasebj.org


American Journal of Physiology-heart and Circulatory Physiology | 2011

Proteasome functional insufficiency in cardiac pathogenesis

Xuejun Wang; Jie Li; Hanqiao Zheng; Huabo Su; Saul R. Powell

The ubiquitin-proteasome system (UPS) is responsible for the degradation of most cellular proteins. Alterations in cardiac UPS, including changes in the degradation of regulatory proteins and proteasome functional insufficiency, are observed in many forms of heart disease and have been shown to play an important role in cardiac pathogenesis. In the past several years, remarkable progress in understanding the mechanisms that regulate UPS-mediated protein degradation has been achieved. A transgenic mouse model of benign enhancement of cardiac proteasome proteolytic function has been created. This has led to the first demonstration of the necessity of proteasome functional insufficiency in the genesis of important pathological processes. Cardiomyocyte-restricted enhancement of proteasome proteolytic function by overexpression of proteasome activator 28α protects against cardiac proteinopathy and myocardial ischemia-reperfusion injury. Additionally, exciting advances have recently been achieved in the search for a pharmacological agent to activate the proteasome. These breakthroughs are expected to serve as an impetus to further investigation into the involvement of UPS dysfunction in molecular pathogenesis and to the development of new therapeutic strategies for combating heart disease. An interplay between the UPS and macroautophagy is increasingly suggested in noncardiac systems but is not well understood in the cardiac system. Further investigations into the interplay are expected to provide a more comprehensive picture of cardiac protein quality control and degradation.


Journal of Molecular and Cellular Cardiology | 2008

Upregulation of myocardial 11S-activated proteasome in experimental hyperglycemia

Saul R. Powell; Samson Mathews Samuel; Ping Wang; Andras Divald; Mahesh Thirunavukkarasu; Srikanth Koneru; Xuejun Wang; Nilanjana Maulik

This study examined the hypothesis that the ubiquitin proteasome system (UPS) degrades proteins damaged by exposure to hyperglycemia. Experimental hyperglycemia was induced in male rats by treatment with streptozotocin. After 30 days, echocardiography confirmed the presence of cardiomyopathy as ejection fraction, fractional shortening, and diastolic function (E/A ratio) were decreased, and chamber diameter was increased in hyperglycemic animals. Proteasome non-ATP-dependent chymotryptic activity was increased over 2-fold in hyperglycemic hearts, but the ATP-dependent activity was decreased and levels of ubiquitinated proteins were increased. Protein levels of the PA28alpha of the 11S-activator ring were increased by 128% and the PA28beta subunit increased by 58% in the hyperglycemic hearts. The alpha3 subunit of the 20S-proteasome was increased by 82% while the catalytic beta5 subunit was increased by 68% in hyperglycemic hearts. Protein oxidation as indicated by protein carbonyls was significantly higher in hyperglycemic hearts. These studies support the conclusion that the UPS becomes dysfunctional during long term hyperglycemia. However, 11S-activated proteasome was increased suggesting a response to oxidative protein damage and a potential role for this form of the proteasome in a cardiac pathophysiology.


Progress in Molecular Biology and Translational Science | 2012

The ubiquitin-proteasome system and cardiovascular disease

Saul R. Powell; Joerg Herrmann; Amir Lerman; Cam Patterson; Xuejun Wang

Over the past decade, the role of the ubiquitin-proteasome system (UPS) has been the subject of numerous studies to elucidate its role in cardiovascular physiology and pathophysiology. There have been many advances in this field including the use of proteomics to achieve a better understanding of how the cardiac proteasome is regulated. Moreover, improved methods for the assessment of UPS function and the development of genetic models to study the role of the UPS have led to the realization that often the function of this system deviates from the norm in many cardiovascular pathologies. Hence, dysfunction has been described in atherosclerosis, familial cardiac proteinopathies, idiopathic dilated cardiomyopathies, and myocardial ischemia. This has led to numerous studies of the ubiquitin protein (E3) ligases and their roles in cardiac physiology and pathophysiology. This has also led to the controversial proposition of treating atherosclerosis, cardiac hypertrophy, and myocardial ischemia with proteasome inhibitors. Furthering our knowledge of this system may help in the development of new UPS-based therapeutic modalities for mitigation of cardiovascular disease.


American Journal of Physiology-heart and Circulatory Physiology | 2013

The ubiquitin proteasome system and myocardial ischemia

Justine Calise; Saul R. Powell

The ubiquitin proteasome system (UPS) has been the subject of intensive research over the past 20 years to define its role in normal physiology and in pathophysiology. Many of these studies have focused in on the cardiovascular system and have determined that the UPS becomes dysfunctional in several pathologies such as familial and idiopathic cardiomyopathies, atherosclerosis, and myocardial ischemia. This review presents a synopsis of the literature as it relates to the role of the UPS in myocardial ischemia. Studies have shown that the UPS is dysfunctional during myocardial ischemia, and recent studies have shed some light on possible mechanisms. Other studies have defined a role for the UPS in ischemic preconditioning which is best associated with myocardial ischemia and is thus presented here. Very recent studies have started to define roles for specific proteasome subunits and components of the ubiquitination machinery in various aspects of myocardial ischemia. Lastly, despite the evidence linking myocardial ischemia and proteasome dysfunction, there are continuing suggestions that proteasome inhibitors may be useful to mitigate ischemic injury. This review presents the rationale behind this and discusses both supportive and nonsupportive studies and presents possible future directions that may help in clarifying this controversy.


PLOS ONE | 2012

Nicotinic Acetylcholine Receptor Agonists Attenuate Septic Acute Kidney Injury in Mice by Suppressing Inflammation and Proteasome Activity

Prodyot Chatterjee; Michael M. Yeboah; Oonagh Dowling; Xiangying Xue; Saul R. Powell; Yousef Al-Abed; Christine N. Metz

Sepsis is one of the leading causes of acute kidney injury (AKI). Septic patients who develop acute kidney injury (AKI) are at increased risk of death. To date there is no effective treatment for AKI or septic AKI. Based on their anti-inflammatory properties, we examined the effects of nicotinic acetylcholine receptor agonists on renal damage using a mouse model of lipopolysaccharide (LPS)-induced AKI where localized LPS promotes inflammation-mediated kidney damage. Administration of nicotine (1 mg/kg) or GTS-21 (4 mg/kg) significantly abrogated renal leukocyte infiltration (by 40%) and attenuated kidney injury. These renoprotective effects were accompanied by reduced systemic and localized kidney inflammation during LPS-induced AKI. Consistent with these observations, nicotinic agonist treatment significantly decreased renal IκBα degradation and NFκB activation during LPS-induced AKI. Treatment of human kidney cells with nicotinic agonists, an NFκB inhibitor (Bay11), or a proteasome inhibitor (MG132) effectively inhibited their inflammatory responses following stimulation with LPS or TNFα. Renal proteasome activity, a major regulator of NFκB-mediated inflammation, was enhanced by approximately 50% during LPS-induced AKI and elevated proteasome activity was significantly blunted by nicotinic agonist administration in vivo. Taken together, our results identify enhanced renal proteasome activity during LPS-induced AKI and the suppression of both proteasome activity and inflammation by nicotinic agonists to attenuate LPS-induced kidney injury.


Free Radical Biology and Medicine | 2010

Decreased sensitivity associated with an altered formulation of a commercially available kit for detection of protein carbonyls

Ping Wang; Saul R. Powell

Carbonylation is a commonly studied form of oxidative modification to proteins which can be conveniently detected using commercially available kits. The most common of these kits is the Oxyblot Protein Oxidation Detection Kit (Chemicon/Millipore). Over the past year we have observed severely diminished sensitivity of these kits which was shown to be a result of a change in the formulation of one of the components supplied in the kit. This component, the 10X 2,4-dinitrophenylhydrazine derivatization solution, which had previously been dissolved in 100% trifluoroacetic acid (TFA), was now dissolved in 2N hydrochloric acid, which according to our results is not acid enough. Further, we observed that upon storage even DNPH dissolved in TFA is subject to degradation. Based on these studies, we make recomendations that should improve the sensitivity and reproducibilty of this assay.


Circulation-heart Failure | 2013

Impaired Assembly and Post-translational Regulation of 26S Proteasome in Human End-Stage Heart Failure

Sharlene M. Day; Andras Divald; Ping Wang; Frank M. Davis; Sarah Bartolone; Richard N. Jones; Saul R. Powell

Background—This study examined the hypothesis that 26S proteasome dysfunction in human end-stage heart failure is associated with decreased docking of the 19S regulatory particle to the 20S proteasome. Previous studies have demonstrated that 26S proteasome activity is diminished in human end-stage heart failure associated with oxidation of the 19S regulatory particle Rpt5 subunit. Docking of the 19S regulatory particle to the 20S proteasome requires functional Rpt subunit ATPase activity and phosphorylation of the &agr;-type subunits. Methods and Results—An enriched proteasome fraction was prepared from 7 human nonfailing and 10 failing heart explants. Native gel electrophoresis assessed docking of 19S to 20S proteasome revealing 3 proteasome populations (20S, 26S, and 30S proteasomes). In failing hearts, 30S proteasomes were significantly lower (P=0.048) by 37% suggesting diminished docking. Mass spectrometry–based phosphopeptide analysis demonstrated that the relative ratio of phosphorylated:non phosphorylated &agr;7 subunit (serine250) of the 20S proteasome was significantly less (P=0.011) by almost 80% in failing hearts. Rpt ATPase activity was determined in the enriched fraction and after immunoprecipitation with an Rpt6 antibody. ATPase activity (&rgr;mol PO4/&mgr;g protein per hour) of the total fraction was lowered from 291±97 to 194±27 and in the immunoprecipitated fraction from 42±12 to 3±2 (P=0.005) in failing hearts. Conclusions—These studies suggest that diminished 26S activity in failing human hearts may be related to impaired docking of the 19S to the 20S as a result of decreased Rpt subunit ATPase activity and &agr;7 subunit phosphorylation.


Cardiovascular Research | 2010

The role of the ubiquitin-proteasome pathway in cardiovascular disease

Christophe Depre; Saul R. Powell; Xuejun Wang

Whereas the mechanisms controlling cellular growth through protein synthesis have been investigated extensively in the cardiovascular system, especially in terms of cardiac cell hypertrophy and vascular smooth muscle cell proliferation, the processes by which endogenous proteins are degraded in these cells have received much less attention and have only begun to be revealed. The ubiquitin-proteasome system (UPS) is responsible for the degradation of 70–90% of intracellular proteins. The fundamental importance of the UPS in these degradation mechanisms was highlighted when Rose, Hershko, and Ciechanover were awarded the 2004 Nobel Prize in Chemistry for their seminal contributions in that field, i.e. the discovery of both ubiquitin1 and the proteasome.2 As illustrated in this Spotlight Issue, protein degradation by the UPS plays a central role in cardiovascular physiology and disease: from endothelial function, the cell cycle, and atherosclerosis to myocardial ischaemia, cardiac hypertrophy, inherited cardiomyopathies, and heart failure. Investigations on the role of the UPS in the cardiovascular system started about 15 years ago and have since increased exponentially. In the heart, the first reports demonstrated that the proteasome is responsible for the degradation of specific, stress-sensitive transcriptional regulators, such as NF-κB, HIF-1α, or inducible cAMP early repressor.3,4 It was subsequently shown that the UPS also degrades contractile proteins and components of the sarcomere.5,6 The development of several categories of proteasome-specific inhibitors further delineated a role for the UPS in multiple forms of heart disease, such as ischaemia/reperfusion, cardiac hypertrophy, cardiomyopathies, and heart failure. …

Collaboration


Dive into the Saul R. Powell's collaboration.

Top Co-Authors

Avatar

Ping Wang

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Xuejun Wang

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jie Li

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar

Andras Divald

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Huabo Su

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Jaime Predmore

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge