Network


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

Hotspot


Dive into the research topics where Kieran F. Geoghegan is active.

Publication


Featured researches published by Kieran F. Geoghegan.


Journal of Clinical Investigation | 1996

Cloning, expression, and type II collagenolytic activity of matrix metalloproteinase-13 from human osteoarthritic cartilage.

Peter G. Mitchell; Holly Magna; Lisa M. Reeves; Lori L. Lopresti-Morrow; Sue A. Yocum; Philip J. Rosner; Kieran F. Geoghegan; John E. Hambor

Proteolysis of triple-helical collagen is an important step in the progression toward irreversible tissue damage in osteoarthritis. Earlier work on the expression of enzymes in cartilage suggested that collagenase-1 (MMP-1) contributes to the process. Degenerate reverse transcription polymerase chain reaction experiments, Northern blot analysis, and direct immunodetection have now provided evidence that collagenase-3 (MMP-13), an enzyme recently cloned from human breast carcinoma, is expressed by chondrocytes in human osteoarthritic cartilage. Variable levels of MMP-13 and MMP-1 in cartilage was significantly induced at both the message and protein levels by interleukin-1 alpha. Recombinant MMP-13 cleaved type II collagen to give characteristic 3/4 and 1/4 fragments; however, MMP-13 turned over type II collagen at least 10 times faster than MMP-1. Experiments with intact type II collagen as well as a synthetic peptide suggested that MMP-13 cleaved type II collagen at the same bond as MMP-1, but this was then followed by a secondary cleavage that removed three amino acids from the 1/4 fragment amino terminus. The expression of MMP-13 in osteoarthritic cartilage and its activity against type II collagen suggest that the enzyme plays a significant role in cartilage collagen degradation, and must consequently form part of a complex target for proposed therapeutic interventions based on collagenase inhibition.


Nature Structural & Molecular Biology | 2007

Structural and biophysical studies of PCSK9 and its mutants linked to familial hypercholesterolemia.

David Cunningham; Dennis E. Danley; Kieran F. Geoghegan; Matthew C. Griffor; Julie Hawkins; Timothy A. Subashi; Alison H. Varghese; Mark Ammirati; Jeffrey S. Culp; Lise R. Hoth; Mahmoud N. Mansour; Katherine M McGrath; Andrew P. Seddon; Shirish Shenolikar; Kim Jonelle Stutzman-Engwall; Laurie C. Warren; Donghui Xia; Xiayang Qiu

Proprotein convertase subtilisin kexin type 9 (PCSK9) lowers the abundance of surface low-density lipoprotein (LDL) receptor through an undefined mechanism. The structure of human PCSK9 shows the subtilisin-like catalytic site blocked by the prodomain in a noncovalent complex and inaccessible to exogenous ligands, and that the C-terminal domain has a novel fold. Biosensor studies show that PCSK9 binds the extracellular domain of LDL receptor with Kd = 170 nM at the neutral pH of plasma, but with a Kd as low as 1 nM at the acidic pH of endosomes. The D374Y gain-of-function mutant, associated with hypercholesterolemia and early-onset cardiovascular disease, binds the receptor 25 times more tightly than wild-type PCSK9 at neutral pH and remains exclusively in a high-affinity complex at the acidic pH. PCSK9 may diminish LDL receptors by a mechanism that requires direct binding but not necessarily receptor proteolysis.


Nature Structural & Molecular Biology | 2007

Crystal structure of cholesteryl ester transfer protein reveals a long tunnel and four bound lipid molecules

Xiayang Qiu; Anil Mistry; Mark Ammirati; Boris A. Chrunyk; Ronald W. Clark; Yang Cong; Jeffrey S. Culp; Dennis E. Danley; Thomas B. Freeman; Kieran F. Geoghegan; Matthew C. Griffor; Steven J. Hawrylik; Cheryl Myers Hayward; Preston Hensley; Lise R. Hoth; George A. Karam; Maruja E. Lira; David B. Lloyd; Katherine M McGrath; Kim Jonelle Stutzman-Engwall; Ann Subashi; Timothy A. Subashi; John F. Thompson; Ing-Kae Wang; Honglei Zhao; Andrew P. Seddon

Cholesteryl ester transfer protein (CETP) shuttles various lipids between lipoproteins, resulting in the net transfer of cholesteryl esters from atheroprotective, high-density lipoproteins (HDL) to atherogenic, lower-density species. Inhibition of CETP raises HDL cholesterol and may potentially be used to treat cardiovascular disease. Here we describe the structure of CETP at 2.2-Å resolution, revealing a 60-Å-long tunnel filled with two hydrophobic cholesteryl esters and plugged by an amphiphilic phosphatidylcholine at each end. The two tunnel openings are large enough to allow lipid access, which is aided by a flexible helix and possibly also by a mobile flap. The curvature of the concave surface of CETP matches the radius of curvature of HDL particles, and potential conformational changes may occur to accommodate larger lipoprotein particles. Point mutations blocking the middle of the tunnel abolish lipid-transfer activities, suggesting that neutral lipids pass through this continuous tunnel.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Kinetic analysis of estrogen receptor/ligand interactions

Rebecca L. Rich; Lise R. Hoth; Kieran F. Geoghegan; Thomas A. Brown; Peter K. LeMotte; Samuel P. Simons; Preston Hensley; David G. Myszka

Surface plasmon resonance biosensor technology was used to directly measure the binding interactions of small molecules to the ligand-binding domain of human estrogen receptor. In a screening mode, specific ligands of the receptor were easily discerned from nonligands. In a high-resolution mode, the association and dissociation phase binding responses were shown to be reproducible and could be fit globally to a simple interaction model to extract reaction rate constants. On average, antagonist ligands (such as tamoxifen and nafoxidine) were observed to bind to the receptor with association rates that were 500-fold slower than agonists (such as estriol and β-estradiol). This finding is consistent with these antagonists binding to an altered conformation of the receptor. The biosensor assay also could identify subtle differences in how the same ligand interacted with two different isoforms of the receptor (α and β). The biosensors ability to determine kinetic rate constants for small molecule/protein interactions provides unique opportunities to understand the mechanisms associated with complex formation as well as new information to drive the optimization of drug candidates.


Journal of Pharmacology and Experimental Therapeutics | 2006

Concentration-Dependent Modulation of Amyloid-β in Vivo and in Vitro Using the γ-Secretase Inhibitor, LY-450139

Thomas A. Lanz; Michael J. Karmilowicz; Kathleen M. Wood; Nikolay Pozdnyakov; Ping Du; Mary A. Piotrowski; Tracy M. Brown; Charles E. Nolan; Karl E.G. Richter; James E. Finley; Qing Fei; Charles F. Ebbinghaus; Yuhpyng L. Chen; Douglas K. Spracklin; Barbara Tate; Kieran F. Geoghegan; Lit-Fui Lau; David D. Auperin; Joel B. Schachter

LY-450139 is a γ-secretase inhibitor shown to have efficacy in multiple cellular and animal models. Paradoxically, robust elevations of plasma amyloid-β (Aβ) have been reported in dogs and humans after administration of subefficacious doses. The present study sought to further evaluate Aβ responses to LY-450139 in the guinea pig, a nontransgenic model that has an Aβ sequence identical to that of human. Male guinea pigs were treated with LY-450139 (0.2–60 mg/kg), and brain, cerebrospinal fluid, and plasma Aβ levels were characterized at 1, 3, 6, 9, and 14 h postdose. Low doses significantly elevated plasma Aβ levels at early time points, with return to baseline within hours. Higher doses inhibited Aβ levels in all compartments at early time points, but elevated plasma Aβ levels at later time points. To determine whether this phenomenon occurs under steady-state drug exposure, guinea pigs were implanted with subcutaneous minipumps delivering LY-450139 (0.3–30 mg/kg/day) for 5 days. Plasma Aβ was significantly inhibited at 10–30 mg/kg/day, but significantly elevated at 1 mg/kg/day. To further understand the mechanism of Aβ elevation by LY-450139, H4 cells overexpressing the Swedish mutant of amyloid-precursor protein and a mouse embryonic stem cell-derived neuronal cell line were studied. In both cellular models, elevated levels of secreted Aβ were observed at subefficacious concentrations, whereas dose-responsive inhibition was observed at higher concentrations. These results suggest that LY-450139 modulates the γ-secretase complex, eliciting Aβ lowering at high concentrations but Aβ elevation at low concentrations.


Journal of Pharmacology and Experimental Therapeutics | 2008

Pharmacologic Inhibition of Site 1 Protease Activity Inhibits Sterol Regulatory Element-Binding Protein Processing and Reduces Lipogenic Enzyme Gene Expression and Lipid Synthesis in Cultured Cells and Experimental Animals

Julie Hawkins; Michael D. Robbins; Laurie C. Warren; Donghui Xia; Stephen F. Petras; James J. Valentine; Alison H. Varghese; Ing-Kae Wang; Timothy A. Subashi; Lorraine D. Shelly; Bruce A. Hay; Katherine T. Landschulz; Kieran F. Geoghegan; H. James Harwood

Sterol regulatory element-binding proteins (SREBPs) are major transcriptional regulators of cholesterol, fatty acid, and glucose metabolism. Genetic disruption of SREBP activity reduces plasma and liver levels of cholesterol and triglycerides and insulin-stimulated lipogenesis, suggesting that SREBP is a viable target for pharmacological intervention. The proprotein convertase SREBP site 1 protease (S1P) is an important posttranscriptional regulator of SREBP activation. This report demonstrates that 10 μM PF-429242 (Bioorg Med Chem Lett 17:4411–4414, 2007), a recently described reversible, competitive aminopyrrolidineamide inhibitor of S1P, inhibits endogenous SREBP processing in Chinese hamster ovary cells. The same compound also down-regulates the signal from an SRE-luciferase reporter gene in human embryonic kidney 293 cells and the expression of endogenous SREBP target genes in cultured HepG2 cells. In HepG2 cells, PF-429242 inhibited cholesterol synthesis, with an IC50 of 0.5 μM. In mice treated with PF-429242 for 24 h, the expression of hepatic SREBP target genes was suppressed, and the hepatic rates of cholesterol and fatty acid synthesis were reduced. Taken together, these data establish that small-molecule S1P inhibitors are capable of reducing cholesterol and fatty acid synthesis in vivo and, therefore, represent a potential new class of therapeutic agents for dyslipidemia and for a variety of cardiometabolic risk factors associated with diabetes, obesity, and the metabolic syndrome.


Journal of Biological Chemistry | 1997

Mutation of a Protease-sensitive Region in Firefly Luciferase Alters Light Emission Properties

John F. Thompson; Kieran F. Geoghegan; David B. Lloyd; Anthony J. Lanzetti; Rachelle A. Magyar; Shannon M. Anderson; Bruce R. Branchini

Luciferase (EC 1.13.12.7) from the North American firefly, Photinus pyralis, is widely used as a reporter enzyme in cell biology. One of its distinctive properties is a pronounced susceptibility to proteolytic degradation that causes luciferase to have a very short intracellular half-life. To define the structural basis for this behavior and possibly facilitate the design of more stable forms of luciferase, limited proteolysis studies were undertaken using trypsin and chymotrypsin to identify regions of the protein whose accessible and flexible character rendered them especially sensitive to cleavage. Regions of amino acid sequence 206–220 and 329–341 were found to be sensitive, and because the region around 206–220 had high homology with other luciferases, CoA ligases, and peptidyl synthetases, this region was selected for mutagenesis experiments intended to determine which of its amino acids were essential for activity. Surprisingly, many highly conserved residues including Ser198, Ser201, Thr202, and Gly203 could be mutated with little effect on the luminescent activity of P. pyralisluciferase. One mutation, however, S198T, caused several alterations in enzymatic properties including shifting the pH optimum from 8.1 to 8.7, lowering the K m for Mg-ATP by a factor of 4 and increasing the half-time for light emission decay by a factor of up to 150. While the S198T luciferase was less active than wild type, activity could be restored by the introduction of the additional L194F and N197Y mutations. In addition to indicating the involvement of this region in ATP binding, these results provide a new form of the enzyme that affords a more versatile reporter system.


Journal of Pharmacology and Experimental Therapeutics | 2012

Phosphodiesterase 9A regulates central cGMP and modulates responses to cholinergic and monoaminergic perturbation in vivo.

Robin J. Kleiman; Douglas S. Chapin; Curt Christoffersen; Jody Freeman; Kari R. Fonseca; Kieran F. Geoghegan; Sarah Grimwood; Victor Guanowsky; Mihály Hajós; John F. Harms; Christopher John Helal; William E. Hoffmann; Geralyn P. Kocan; Mark J. Majchrzak; Dina McGinnis; Stafford McLean; Frank S. Menniti; Fredrick R. Nelson; Robin Roof; Anne W. Schmidt; Patricia A. Seymour; Diane Stephenson; Francis David Tingley; Michelle Vanase-Frawley; Patrick Robert Verhoest; Christopher J. Schmidt

Cyclic nucleotides are critical regulators of synaptic plasticity and participate in requisite signaling cascades implicated across multiple neurotransmitter systems. Phosphodiesterase 9A (PDE9A) is a high-affinity, cGMP-specific enzyme widely expressed in the rodent central nervous system. In the current study, we observed neuronal staining with antibodies raised against PDE9A protein in human cortex, cerebellum, and subiculum. We have also developed several potent, selective, and brain-penetrant PDE9A inhibitors and used them to probe the function of PDE9A in vivo. Administration of these compounds to animals led to dose-dependent accumulation of cGMP in brain tissue and cerebrospinal fluid, producing a range of biological effects that implied functional significance for PDE9A-regulated cGMP in dopaminergic, cholinergic, and serotonergic neurotransmission and were consistent with the widespread distribution of PDE9A. In vivo effects of PDE9A inhibition included reversal of the respective disruptions of working memory by ketamine, episodic and spatial memory by scopolamine, and auditory gating by amphetamine, as well as potentiation of risperidone-induced improvements in sensorimotor gating and reversal of the stereotypic scratching response to the hallucinogenic 5-hydroxytryptamine 2A agonist mescaline. The results suggested a role for PDE9A in the regulation of monoaminergic circuitry associated with sensory processing and memory. Thus, PDE9A activity regulates neuronal cGMP signaling downstream of multiple neurotransmitter systems, and inhibition of PDE9A may provide therapeutic benefits in psychiatric and neurodegenerative diseases promoted by the dysfunction of these diverse neurotransmitter systems.


FEBS Letters | 1990

Fluorescence-based continuous assay for the aspartyl protease of human immunodeficiency virus-1

Kieran F. Geoghegan; Robin W. Spencer; Dennis E. Danley; Leonard G. Contillo; Glenn C. Andrews

5‐Dimethylaminonaphthalene‐1‐sulfonyl‐Ser‐Gln‐Asn‐Tyr‐Pro‐Ile‐Val‐Trp (Dns‐SQNYPIVW) is a fluorescent substrate for the aspartyl protease of human immunodeficiency virus‐1. In intact substrate, fluorescence of Trp (λex 290 nm, λem 360 nm) was 60% quenched by energy transfer to the dansyl group. Protease‐catalyzed cleavage at the Tyr‐Pro bond abolished the energy transfer, and the consequent increase in Trp fluorescence was used to follow the enzymatic reaction. At substrate concentrations <60 μM, initial reaction velocity increased as a linear function of substrate concentration, with k cat/K M= 9700 M−1s−1. Limited solubility and internal fluorescence quenching precluded a determination of K M for Dns‐SQNYPIVW, but this was clearly > 100 μM.


Journal of Neurochemistry | 2014

Phosphoproteomic evaluation of pharmacological inhibition of leucine‐rich repeat kinase 2 reveals significant off‐target effects of LRRK‐2‐IN‐1

Gregory C. Luerman; Chuong Nguyen; Harry Samaroo; Paula Loos; Hualin Xi; Andres Hurtado-Lorenzo; Elie Needle; G. Stephen Noell; Paul Galatsis; John Dunlop; Kieran F. Geoghegan; Warren D. Hirst

Genetic mutations in leucine‐rich repeat kinase 2 (LRRK2) have been linked to autosomal dominant Parkinsons disease. The most prevalent mutation, G2019S, results in enhanced LRRK2 kinase activity that potentially contributes to the etiology of Parkinsons disease. Consequently, disease progression is potentially mediated by poorly characterized phosphorylation‐dependent LRRK2 substrate pathways. To address this gap in knowledge, we transduced SH‐SY5Y neuroblastoma cells with LRRK2 G2019S via adenovirus, then determined quantitative changes in the phosphoproteome upon LRRK2 kinase inhibition (LRRK2‐IN‐1 treatment) using stable isotope labeling of amino acids in culture combined with phosphopeptide enrichment and LC‐MS/MS analysis. We identified 776 phosphorylation sites that were increased or decreased at least 50% in response to LRRK2‐IN‐1 treatment, including sites on proteins previously known to associate with LRRK2. Bioinformatic analysis of those phosphoproteins suggested a potential role for LRRK2 kinase activity in regulating pro‐inflammatory responses and neurite morphology, among other pathways. In follow‐up experiments, LRRK2‐IN‐1 inhibited lipopolysaccharide‐induced tumor necrosis factor alpha (TNFα) and C‐X‐C motif chemokine 10 (CXCL10) levels in astrocytes and also enhanced multiple neurite characteristics in primary neuronal cultures. However, LRRK2‐IN‐1 had almost identical effects in primary glial and neuronal cultures from LRRK2 knockout mice. These data suggest LRRK2‐IN‐1 may inhibit pathways of perceived LRRK2 pathophysiological function independently of LRRK2 highlighting the need to use multiple pharmacological tools and genetic approaches in studies determining LRRK2 function.

Collaboration


Dive into the Kieran F. Geoghegan's collaboration.

Top Co-Authors

Avatar

Lise R. Hoth

Australian National University

View shared research outputs
Top Co-Authors

Avatar

Lise R. Hoth

Australian National University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge