Network


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

Hotspot


Dive into the research topics where Andrea L. Portbury is active.

Publication


Featured researches published by Andrea L. Portbury.


Molecular and Cellular Biology | 2008

CHIP deficiency decreases longevity, with accelerated aging phenotypes accompanied by altered protein quality control.

Jin Na Min; Ryan A. Whaley; Norman E. Sharpless; Pamela Lockyer; Andrea L. Portbury; Cam Patterson

ABSTRACT During the course of biological aging, there is a gradual accumulation of damaged proteins and a concomitant functional decline in the protein degradation system. Protein quality control is normally ensured by the coordinated actions of molecular chaperones and the protein degradation system that collectively help to maintain protein homeostasis. The carboxyl terminus of Hsp70-interacting protein (CHIP), a ubiquitin ligase/cochaperone, participates in protein quality control by targeting a broad range of chaperone substrates for proteasome degradation via the ubiquitin-proteasome system, demonstrating a broad involvement of CHIP in maintaining cytoplasmic protein quality control. In the present study, we have investigated the influence that protein quality control exerts on the aging process by using CHIP−/− mice. CHIP deficiency in mice leads to a markedly reduced life span, along with accelerated age-related pathophysiological phenotypes. These features were accompanied by indications of accelerated cellular senescence and increased indices of oxidative stress. In addition, CHIP−/− mice exhibit a deregulation of protein quality control, as indicated by elevated levels of toxic oligomer proteins and a decline in proteasome activity. Taken together, these data reveal that impaired protein quality control contributes to cellular senescence and implicates CHIP-dependent quality control mechanisms in the regulation of mammalian longevity in vivo.


Cardiovascular Research | 2008

Build it up-Tear it down: protein quality control in the cardiac sarcomere.

Monte S. Willis; Jonathan C. Schisler; Andrea L. Portbury; Cam Patterson

The assembly and maintenance of the cardiac sarcomere, which contains the basic contractile components of actin and myosin, are essential for cardiac function. While often described as a static structure, the sarcomere is actually dynamic and undergoes constant turnover, allowing it to adapt to physiological changes while still maintaining function. A host of new factors have been identified that play a role in the regulation of protein quality control in the sarcomere, including chaperones that mediate the assembly of sarcomere components and ubiquitin ligases that control their specific degradation. There is clear evidence of sarcomere disorganization in animal models lacking muscle-specific chaperone proteins, illustrating the importance of these molecules in sarcomere structure and function. Although ubiquitin ligases have been found within the sarcomere structure itself, the role of the ubiquitin proteasome system in cardiac sarcomere regulation, and the factors that control its activity, are only just now being elucidated. The number of ubiquitin ligases identified with specificity for sarcomere proteins, each with distinct target substrates, is growing, allowing for tight regulation of this system. In this review, we highlight the dynamic interplay between sarcomere-specific chaperones and ubiquitin-dependent degradation of sarcomere proteins that is necessary in order to maintain structure and function of the cardiac sarcomere.


Human Molecular Genetics | 2014

Ataxia and hypogonadism caused by the loss of ubiquitin ligase activity of the U box protein CHIP

Chang He Shi; Jonathan C. Schisler; Carrie Rubel; Song Tan; Bo Song; Holly McDonough; Lei Xu; Andrea L. Portbury; Cheng Yuan Mao; Cadence True; Rui Hao Wang; Qing Zhi Wang; Shi lei Sun; Stephanie B. Seminara; Cam Patterson; Yu Ming Xu

Gordon Holmes syndrome (GHS) is a rare Mendelian neurodegenerative disorder characterized by ataxia and hypogonadism. Recently, it was suggested that disordered ubiquitination underlies GHS though the discovery of exome mutations in the E3 ligase RNF216 and deubiquitinase OTUD4. We performed exome sequencing in a family with two of three siblings afflicted with ataxia and hypogonadism and identified a homozygous mutation in STUB1 (NM_005861) c.737C→T, p.Thr246Met, a gene that encodes the protein CHIP (C-terminus of HSC70-interacting protein). CHIP plays a central role in regulating protein quality control, in part through its ability to function as an E3 ligase. Loss of CHIP function has long been associated with protein misfolding and aggregation in several genetic mouse models of neurodegenerative disorders; however, a role for CHIP in human neurological disease has yet to be identified. Introduction of the Thr246Met mutation into CHIP results in a loss of ubiquitin ligase activity measured directly using recombinant proteins as well as in cell culture models. Loss of CHIP function in mice resulted in behavioral and reproductive impairments that mimic human ataxia and hypogonadism. We conclude that GHS can be caused by a loss-of-function mutation in CHIP. Our findings further highlight the role of disordered ubiquitination and protein quality control in the pathogenesis of neurodegenerative disease and demonstrate the utility of combining whole-exome sequencing with molecular analyses and animal models to define causal disease polymorphisms.


Journal of Biological Chemistry | 2011

Tearin' Up My Heart: Proteolysis in the Cardiac Sarcomere

Andrea L. Portbury; Monte S. Willis; Cam Patterson

Proteolysis within the cardiac sarcomere is a constantly evolving area of research. Three major pathways of proteolysis have been identified as being active within the cardiac sarcomere, namely the ubiquitin-proteasome system, autophagy, and the calpain system. The role of ubiquitin-proteasome system-mediated proteolysis in cardiovascular health and disease has been known for some time; however, it is now apparent that other proteolytic systems also aid in the stabilization of cardiac sarcomere structure and function. This minireview focuses on the individual as well as cooperative involvement of each of these three major pathways of proteolysis within the cardiac sarcomere.


Circulation Research | 2008

Brothers and sisters: molecular insights into arterial-venous heterogeneity.

Julius Aitsebaomo; Andrea L. Portbury; Jonathan C. Schisler; Cam Patterson

The molecular differences between arteries and veins are genetically predetermined and are evident even before the first embryonic heart beat. Although ephrinB2 and EphB4 are expressed in cells that will ultimately differentiate into arteries and veins, respectively, many other genes have been shown to play a significant role in cell fate determination. The expression patterns of ephrinB2 and EphB4 are restricted to arterial-venous boundaries, and Eph/ephrin signaling provides repulsive cues at arterial-venous boundaries that are thought to prevent intermixing of arterial- and venous-fated cells. However, the maintenance of arterial-venous fate is susceptible to some degree of plasticity. Thus, in response to signals from the ambient microenvironment and shear stress, there is flow-mediated intercalation of the arteries and veins that ultimately leads to the formation of a functional, closed-loop circulation. In addition, cells in the blood vessels of each organ undergo epigenetic, morphological, and functional adaptive changes that are specific to the proximate function of their cognate organ(s). These adaptive changes result in an interorgan and intraorgan vessel heterogeneity that manifest clinically in a disparate response of different organs to identical risk factors and injury in the same animal. In this review, we focus on the molecular and physiological factors influencing arterial-venous heterogeneity between and within different organ(s). We explore arterial-venous differences in selected organs, as well as their respective endothelial cell architectural organization that results in their inter- and intraorgan heterogeneity.


Circulation Research | 2012

LRP1-Dependent Endocytic Mechanism Governs the Signaling Output of the Bmp System in Endothelial Cells and in Angiogenesis

Xinchun Pi; Christopher E. Schmitt; Liang Xie; Andrea L. Portbury; Yaxu Wu; Pamela Lockyer; Laura A. Dyer; Martin Moser; Guojun Bu; Edward J. Flynn; Suk-Won Jin; Cam Patterson

Rationale: Among the extracellular modulators of Bmp (bone morphogenetic protein) signaling, Bmper (Bmp endothelial cell precursor-derived regulator) both enhances and inhibits Bmp signaling. Recently we found that Bmper modulates Bmp4 activity via a concentration-dependent, endocytic trap-and–sink mechanism. Objective: To investigate the molecular mechanisms required for endocytosis of the Bmper/Bmp4 and signaling complex and determine the mechanism of Bmpers differential effects on Bmp4 signaling. Methods and Results: Using an array of biochemical and cell biology techniques, we report that LRP1 (LDL receptor-related protein 1), a member of the LDL receptor family, acts as an endocytic receptor for Bmper and a coreceptor of Bmp4 to mediate the endocytosis of the Bmper/Bmp4 signaling complex. Furthermore, we demonstrate that LRP1-dependent Bmper/Bmp4 endocytosis is essential for Bmp4 signaling, as evidenced by the phenotype of lrp1-deficient zebrafish, which have abnormal cardiovascular development and decreased Smad1/5/8 activity in key vasculogenic structures. Conclusions: Together, these data reveal a novel role for LRP1 in the regulation of Bmp4 signaling by regulating receptor complex endocytosis. In addition, these data introduce LRP1 as a critical regulator of vascular development. These observations demonstrate Bmpers ability to fine-tune Bmp4 signaling at the single-cell level, unlike the spatial regulatory mechanisms applied by other Bmp modulators.


Circulation Research | 2011

Tear Me Down Role of Calpain in the Development of Cardiac Ventricular Hypertrophy

Cam Patterson; Andrea L. Portbury; Jonathan C. Schisler; Monte S. Willis

Cardiac hypertrophy develops most commonly in response to hypertension and is an independent risk factor for the development of heart failure. The mechanisms by which cardiac hypertrophy may be reversed to reduce this risk have not been fully determined to the point where mechanism-specific therapies have been developed. Recently, proteases in the calpain family have been implicated in the regulation of the development of cardiac hypertrophy in preclinical animal models. In this review, we summarize the molecular mechanisms by which calpain inhibition has been shown to modulate the development of cardiac (specifically ventricular) hypertrophy. The context within which calpain inhibition might be developed for therapeutic intervention of cardiac hypertrophy is then discussed.


Journal of Biological Chemistry | 2009

Stress-dependent Daxx-CHIP interaction suppresses the p53 apoptotic program.

Holly McDonough; Peter C. Charles; Eleanor Hilliard; Shu-Bing Qian; Jin-na Min; Andrea L. Portbury; Douglas M. Cyr; Cam Patterson

Our previous studies have implicated CHIP (carboxyl terminus of Hsp70-interacting protein) as a co-chaperone/ubiquitin ligase whose activities yield protection against stress-induced apoptotic events. In this report, we demonstrate a stress-dependent interaction between CHIP and Daxx (death domain-associated protein). This interaction interferes with the stress-dependent association of HIPK2 with Daxx, blocking phosphorylation of serine 46 in p53 and inhibiting the p53-dependent apoptotic program. Microarray analysis confirmed suppression of the p53-dependent transcriptional portrait in CHIP+/+ but not in CHIP−/− heat shocked mouse embryonic fibroblasts. The interaction between CHIP and Daxx results in ubiquitination of Daxx, which is then partitioned to an insoluble compartment of the cell. In vitro ubiquitination of Daxx by CHIP revealed that ubiquitin chain formation utilizes non-canonical lysine linkages associated with resistance to proteasomal degradation. The ubiquitination of Daxx by CHIP utilizes lysines 630 and 631 and competes with the sumoylation machinery of the cell at these residues. These studies implicate CHIP as a stress-dependent regulator of Daxx that counters the pro-apoptotic influence of Daxx in the cell. By abrogating p53-dependent apoptotic pathways and by ubiquitination competitive with Daxx sumoylation, CHIP integrates the proteotoxic stress response of the cell with cell cycle pathways that influence cell survival.


BMC Medical Genomics | 2008

Tobacco use induces anti-apoptotic, proliferative patterns of gene expression in circulating leukocytes of Caucasian males

Peter C. Charles; Brian D. Alder; Eleanor Hilliard; Jonathan C. Schisler; Robert E. Lineberger; Joel S. Parker; Sabeen Mapara; Samuel S. Wu; Andrea L. Portbury; Cam Patterson; George A. Stouffer

BackgroundStrong epidemiologic evidence correlates tobacco use with a variety of serious adverse health effects, but the biological mechanisms that produce these effects remain elusive.ResultsWe analyzed gene transcription data to identify expression spectra related to tobacco use in circulating leukocytes of 67 Caucasian male subjects. Levels of cotinine, a nicotine metabolite, were used as a surrogate marker for tobacco exposure. Significance Analysis of Microarray and Gene Set Analysis identified 109 genes in 16 gene sets whose transcription levels were differentially regulated by nicotine exposure. We subsequently analyzed this gene set by hyperclustering, a technique that allows the data to be clustered by both expression ratio and gene annotation (e.g. Gene Ontologies).ConclusionOur results demonstrate that tobacco use affects transcription of groups of genes that are involved in proliferation and apoptosis in circulating leukocytes. These transcriptional effects include a repertoire of transcriptional changes likely to increase the incidence of neoplasia through an altered expression of genes associated with transcription and signaling, interferon responses and repression of apoptotic pathways.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

NADPH Oxidase–Generated Reactive Oxygen Species Are Required for Stromal Cell–Derived Factor-1α–Stimulated Angiogenesis

Xinchun Pi; Liang Xie; Andrea L. Portbury; Sarayu Kumar; Pamela Lockyer; Xi Li; Cam Patterson

Objective— Reactive oxygen species (ROS) act as signaling molecules during angiogenesis; however, the mechanisms used for such signaling events remain unclear. Stromal cell–derived factor-1&agr; (SDF-1&agr;) is one of the most potent angiogenic chemokines. Here, we examined the role of ROS in the regulation of SDF-1&agr;–dependent angiogenesis. Approach and Results— Bovine aortic endothelial cells were treated with SDF-1&agr;, and intracellular ROS generation was monitored. SDF-1&agr; treatment induced bovine aortic endothelial cell migration and ROS generation, with the majority of ROS generated by bovine aortic endothelial cells at the leading edge of the migratory cells. Antioxidants and nicotinamide adenine dinucleotide phosphate oxidase (NOX) inhibitors blocked SDF-1&agr;–induced endothelial migration. Furthermore, knockdown of either NOX5 or p22phox (a requisite subunit for NOX1/2/4 activation) significantly impaired endothelial motility and tube formation, suggesting that multiple NOXs regulate SDF-1&agr;–dependent angiogenesis. Our previous study demonstrated that c-Jun N-terminal kinase 3 activity is essential for SDF-1&agr;–dependent angiogenesis. Here, we identified that NOX5 is the dominant NOX required for SDF-1&agr;–induced c-Jun N-terminal kinase 3 activation and that NOX5 and MAP kinase phosphatase 7 (MKP7; the c-Jun N-terminal kinase 3 phosphatase) associate with one another but decrease this interaction on SDF-1&agr; treatment. Furthermore, MKP7 activity was inhibited by SDF-1&agr;, and this inhibition was relieved by NOX5 knockdown, indicating that NOX5 promotes c-Jun N-terminal kinase 3 activation by blocking MKP7 activity. Conclusions— We conclude that NOX is required for SDF-1&agr; signaling and that intracellular redox balance is critical for SDF-1&agr;–induced endothelial migration and angiogenesis.

Collaboration


Dive into the Andrea L. Portbury's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Monte S. Willis

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Pamela Lockyer

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jonathan C. Schisler

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Xinchun Pi

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yaxu Wu

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Michael Bode

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Weeranun Dechyapirom

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Eleanor Hilliard

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Holly McDonough

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge