Network


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

Hotspot


Dive into the research topics where Andrew J. Paterson is active.

Publication


Featured researches published by Andrew J. Paterson.


Journal of Neurochemistry | 2002

Modulation of Intracellular Cyclic AMP Levels by Different Human Dopamine D4 Receptor Variants

Vida Asghari; Suparna Sanyal; Saskia Buchwaldt; Andrew J. Paterson; Vera Jovanovic; Hubert H. M. Van Tol

Abstract: To investigate whether polymorphic forms of the human dopamine D4 receptor have different functional characteristics, we have stably expressed cDNAs of the D4.2, D4.4, and D4.7 isoforms in several cell lines. Chinese hamster ovary CHO‐K1 cell lines expressing D4 receptor variants displayed pharmacological profiles that were in close agreement with previous data from transiently expressed D4 receptors in COS‐7 cells. Dopamine stimulation of the D4 receptors resulted in a concentration‐dependent inhibition of the forskolin‐stimulated cyclic AMP (cAMP) levels. The potency of dopamine to inhibit cAMP formation was about twofold reduced for D4.7 (EC50 of ∼37 nM) compared with the D4.2 and D4.4 variants (EC50 of ∼16 nM). Antagonists block the dopamine‐mediated inhibition of cAMP formation with a rank order of potency of emonapride > haloperidol = clozapine ≫ raclopride. There was no obvious correlation between the efficacy of inhibition of forskolin‐stimulated cAMP levels and the D4 subtypes. Dopamine could completely reverse prostaglandin E2‐stimulated cAMP levels for all three D4 receptor variants. Deletion of the repeat sequence does not affect functional activity of the receptor. The data presented indicate that the polymorphic repeat sequence causes only small changes in the ability of the D4 receptor to block cAMP production in CHO cells.


Cell | 2003

O-GlcNAc Modification Is an Endogenous Inhibitor of the Proteasome

Fengxue Zhang; Kaihong Su; Xiaoyong Yang; Damon B. Bowe; Andrew J. Paterson; Jeffrey E. Kudlow

The ubiquitin proteasome system classically selects its substrates for degradation by tagging them with ubiquitin. Here, we describe another means of controlling proteasome function in a global manner. The 26S proteasome can be inhibited by modification with the enzyme, O-GlcNAc transferase (OGT). This reversible modification of the proteasome inhibits the proteolysis of the transcription factor Sp1 and a hydrophobic peptide through inhibition of the ATPase activity of 26S proteasomes. The Rpt2 ATPase in the mammalian proteasome 19S cap is modified by O-GlcNAc in vitro and in vivo and as its modification increases, proteasome function decreases. This mechanism may couple proteasomes to the general metabolic state of the cell. The O-GlcNAc modification of proteasomes may allow the organism to respond to its metabolic needs by controlling the availability of amino acids and regulatory proteins.


Proceedings of the National Academy of Sciences of the United States of America | 2001

O-linkage of N-acetylglucosamine to Sp1 activation domain inhibits its transcriptional capability

Xiaoyong Yang; Kaihong Su; Mark D. Roos; Qing Chang; Andrew J. Paterson; Jeffrey E. Kudlow

The posttranslational modification of eukaryotic intracellular proteins by O-linked N-acetylglucosamine (O-GlcNAc) monosaccharides is essential for cell viability, yet its precise functional roles are largely unknown. O-GlcNAc transferase utilizes UDP-GlcNAc, the end product of hexosamine biosynthesis, to catalyze this modification. The availability of UDP-GlcNAc correlates with glycosylation levels of intracellular proteins as well as with transcriptional levels of some genes. Meanwhile, transcription factors and RNA polymerase II can be modified by O-GlcNAc. A linkage between transcription factor O-GlcNAcylation and transcriptional regulation therefore has been postulated. Here, we show that O-GlcNAcylation of a chimeric transcriptional activator containing the second activation domain of Sp1 decreases its transcriptional activity both in an in vitro transcription system and in living cells, which is in concert with our observation that O-GlcNAcylation of Sp1 activation domain blocks its in vitro and in vivo interactions with other Sp1 molecules and TATA-binding protein-associated factor II 110. Furthermore, overexpression of O-GlcNAc transferase specifically inhibits transcriptional activation by native Sp1 in cells. Thus, our studies provide direct evidence that O-GlcNAcylation of transcription factors is involved in transcriptional regulation.


Oncogene | 2005

Induction of KLF4 in basal keratinocytes blocks the proliferation-differentiation switch and initiates squamous epithelial dysplasia

K. Wade Foster; Zhaoli Liu; Nail Cd; Xingnan Li; Thomas J. Fitzgerald; Sarah K. Bailey; Andra R. Frost; Iúri Drumond Louro; Tim M Townes; Andrew J. Paterson; Jeffrey E. Kudlow; Susan M. Lobo-Ruppert; J. Michael Ruppert

KLF4/GKLF normally functions in differentiating epithelial cells, but also acts as a transforming oncogene in vitro. To examine the role of this zinc finger protein in skin, we expressed the wild-type human allele from inducible and constitutive promoters. When induced in basal keratinocytes, KLF4 rapidly abolished the distinctive properties of basal and parabasal epithelial cells. KLF4 caused a transitory apoptotic response and the skin progressed through phases of hyperplasia and dysplasia. By 6 weeks, lesions exhibited nuclear KLF4 and other morphologic and molecular similarities to squamous cell carcinoma in situ. p53 determined the patch size sufficient to establish lesions, as induction in a mosaic pattern produced skin lesions only when p53 was deficient. Compared with p53 wild-type animals, p53 hemizygous animals had early onset of lesions and a pronounced fibrovascular response that included outgrowth of subcutaneous sarcoma. A KLF4-estrogen receptor fusion protein showed tamoxifen-dependent nuclear localization and conditional transformation in vitro. The results suggest that KLF4 can function in the nucleus to induce squamous epithelial dysplasia, and indicate roles for p53 and epithelial–mesenchymal signaling in these early neoplastic lesions.


Journal of Biological Chemistry | 2004

Characterization of the histone acetyltransferase (HAT) domain of a bifunctional protein with activable O-GlcNAcase and HAT activities.

Clifford A. Toleman; Andrew J. Paterson; Thomas R. Whisenhunt; Jeffrey E. Kudlow

Histones and transcription factors are regulated by a number of post-translational modifications that in turn regulate the transcriptional activity of genes. These modifications occur in large, multisubunit complexes. We have reported previously that mSin3A can recruit O-GlcNAc transferase (OGT) along with histone deacetylase into such a corepressor complex. This physical association allows OGT to act cooperatively with histone deacetylation in gene repression by catalyzing the O-GlcNAc modification on specific transcription factors to inhibit their activity. For rapid, reversible gene regulation, the enzymes responsible for the converse reactions must be present. Here, we report that O-GlcNAcase, which is responsible for the removal of O-GlcNAc additions on nuclear and cytosolic proteins, possesses intrinsic histone acetyltransferase (HAT) activity in vitro. Free as well as reconstituted nucleosomal histones are substrates of this bifunctional enzyme. This protein, now termed NCOAT (nuclear cytoplasmic O-GlcNAcase and acetyltransferase) has a typical HAT domain that has both active and inactive states. This finding demonstrates that NCOAT may be regulated to reduce the state of glycosylation of transcriptional activators while increasing the acetylation of histones to allow for the concerted activation of eukaryotic gene transcription.


Journal of Biological Chemistry | 2007

Proteasome function is regulated by cyclic AMP-dependent protein kinase through phosphorylation of RPT6

Fengxue Zhang; Yong Hu; Ping Huang; Clifford A. Toleman; Andrew J. Paterson; Jeffrey E. Kudlow

Dysregulation of the proteasome has been documented in a variety of human diseases such as Alzheimer, muscle atrophy, cataracts etc. Proteolytic activity of 26 S proteasome is ATP- and ubiquitin-dependent. O-GlcNAcylation of Rpt2, one of the AAA ATPases in the 19 S regulatory cap, shuts off the proteasome through the inhibition of ATPase activity. Thus, through control of the flux of glucose into O-GlcNAc, the function of the proteasome is coupled to glucose metabolism. In the present study we found another metabolic control of the proteasome via cAMP-dependent protein kinase (PKA). Contrary to O-Glc-NAcylation, PKA activated proteasomes both in vitro and in vivo in association with the phosphorylation at Ser120 of another AAA ATPase subunit, Rpt6. Mutation of Ser120 to Ala blocked proteasome function. The stimulatory effect of PKA and the phosphorylation of Rpt6 were reversible by protein phosphatase 1γ. Thus, hormones using the PKA system can also regulate proteasomes often in concert with glucose metabolism. This finding might lead to novel strategies for the treatment of proteasome-related diseases.


Molecular and Cellular Biology | 2006

Gene Expression Analysis Exposes Mitochondrial Abnormalities in a Mouse Model of Rett Syndrome

Skirmantas Kriaucionis; Andrew J. Paterson; John C. Curtis; Jacqueline Guy; Nikki MacLeod; Adrian Bird

ABSTRACT Rett syndrome (RTT) is a severe neurological disorder caused by mutations in the X-linked MECP2 gene, which encodes a methyl-CpG binding transcriptional repressor. Using the Mecp2-null mouse (an animal model for RTT) and differential display, we found that mice with neurological symptoms overexpress the nuclear gene for ubiquinol-cytochrome c reductase core protein 1 (Uqcrc1). Chromatin immunoprecipitation demonstrated that MeCP2 interacts with the Uqcrc1 promoter. Uqcrc1 encodes a subunit of mitochondrial respiratory complex III, and isolated mitochondria from the Mecp2-null brain showed elevated respiration rates associated with respiratory complex III and an overall reduction in coupling. A causal link between Uqcrc1 gene overexpression and enhanced complex III activity was established in neuroblastoma cells. Our findings raise the possibility that mitochondrial dysfunction contributes to pathology of the Mecp2-null mouse and may contribute to the long-known resemblance between Rett syndrome and certain mitochondrial disorders.


American Journal of Physiology-heart and Circulatory Physiology | 2009

Protein O-GlcNAcylation: a new signaling paradigm for the cardiovascular system

Boglárka Laczy; Bradford G. Hill; Kai Wang; Andrew J. Paterson; C. Roger White; Dongqi Xing; Yiu-Fai Chen; Victor M. Darley-Usmar; Suzanne Oparil; John C. Chatham

The posttranslational modification of serine and threonine residues of nuclear and cytoplasmic proteins by the O-linked attachment of the monosaccharide beta-N-acetylglucosamine (O-GlcNAc) is a highly dynamic and ubiquitous protein modification. Protein O-GlcNAcylation is rapidly emerging as a key regulator of critical biological processes including nuclear transport, translation and transcription, signal transduction, cytoskeletal reorganization, proteasomal degradation, and apoptosis. Increased levels of O-GlcNAc have been implicated as a pathogenic contributor to glucose toxicity and insulin resistance, which are both major hallmarks of diabetes mellitus and diabetes-related cardiovascular complications. Conversely, there is a growing body of data demonstrating that the acute activation of O-GlcNAc levels is an endogenous stress response designed to enhance cell survival. Reports on the effect of altered O-GlcNAc levels on the heart and cardiovascular system have been growing rapidly over the past few years and have implicated a role for O-GlcNAc in contributing to the adverse effects of diabetes on cardiovascular function as well as mediating the response to ischemic injury. Here, we summarize our present understanding of protein O-GlcNAcylation and its effect on the regulation of cardiovascular function. We examine the pathways regulating protein O-GlcNAcylation and discuss, in more detail, our understanding of the role of O-GlcNAc in both mediating the adverse effects of diabetes as well as its role in mediating cellular protective mechanisms in the cardiovascular system. In addition, we also explore the parallels between O-GlcNAc signaling and redox signaling, as an alternative paradigm for understanding the role of O-GlcNAcylation in regulating cell function.


Journal of Biological Chemistry | 1999

An N-terminal region of Sp1 targets its proteasome-dependent degradation in vitro

Kaihong Su; Mark D. Roos; Xiaoyong Yang; Inn-Oc Han; Andrew J. Paterson; Jeffrey E. Kudlow

The transcription factor Sp1 is important for the expression of many cellular genes. Previously, it was shown that reduced O-glycosylation of Sp1 is associated with increased proteasome susceptibility. Sp1 undergoes proteasome-dependent degradation in cells stressed with glucose deprivation and adenylate cyclase activation, and this process is blocked in cells treated with glucosamine. In this study, using a reconstituted in vitro system, we identified the principal structural determinant in Sp1 that targets Sp1 for proteasome-dependent degradation. We found by using deletion analysis that the N-terminal 54 amino acids of Sp1 is required for Sp1 degradation. This element can act as an independent processing signal by directing degradation of an unrelated protein. Recognition of this Sp1 element by the proteasome-dependent system is saturable, and ubiquitination of this element is not required for recognition. Time course experiments revealed that Sp1 degradation is a two-step process. First, a discrete endoproteolytic cleavage occurs downstream of the target region immediately C-terminal to Leu56. The Sp1 sequence C-terminal to the cleavage site is subsequently degraded, whereas the N-terminal peptide remains intact. The identification of this Sp1 degradation-targeting signal will facilitate the identification of the critical proteins involved in the control of Sp1 proteasome-dependent degradation and the role of OGlcNAc in this process.


Journal of Neurochemistry | 2004

Accumulation of protein O-GlcNAc modification inhibits proteasomes in the brain and coincides with neuronal apoptosis in brain areas with high O-GlcNAc metabolism

Kan Liu; Andrew J. Paterson; Fengxue Zhang; Joanne McAndrew; Ken Ichiro Fukuchi; J. Michael Wyss; Ling Peng; Yong Hu; Jeffrey E. Kudlow

All tissues contain the enzymes that modify and remove O‐GlcNAc dynamically from nucleocytoplasmic proteins. These enzymes have been shown to play a role in the control of transcription, vesicular trafficking and, more recently, proteasome function. Modification by O‐GlcNAc of the 19S cap of the proteasome inhibits proteasomal function. Transcripts of both O‐GlcNAc transferase and O‐GlcNAcase are very abundant in the brain, with the highest concentrations in hippocampal neurons and Purkinje cells. When the on‐rate of modification is favored over the off‐rate by intraventricular administration of a drug, streptozocin, these areas of the brain display the most rapid accumulation of O‐GlcNAc. Cerebral proteasome function is reduced and ubiquitin and p53 accumulate in these brain regions, with the subsequent activation of a p53‐dependent transgene and the endogenous Mdm2 gene. Later, some hippocampal cells, but not Purkinje cells, undergo apoptosis. These observations suggest that the O‐GlcNAc system may participate in neurodegeneration, particularly in the hippocampus.

Collaboration


Dive into the Andrew J. Paterson's collaboration.

Top Co-Authors

Avatar

Jeffrey E. Kudlow

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Clifford A. Toleman

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Edward Chin

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Kaihong Su

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ping Huang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Yong Hu

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Damon B. Bowe

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Fengxue Zhang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

John C. Chatham

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge