Network


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

Hotspot


Dive into the research topics where Suzan Aschmies is active.

Publication


Featured researches published by Suzan Aschmies.


The Journal of Neuroscience | 2005

Acute γ-Secretase Inhibition Improves Contextual Fear Conditioning in the Tg2576 Mouse Model of Alzheimer's Disease

Thomas A. Comery; Robert Martone; Suzan Aschmies; Kevin Atchison; George Diamantidis; Xiaohai Gong; Hua Zhou; Anthony F. Kreft; Menelas N. Pangalos; June Sonnenberg-Reines; J. Steven Jacobsen; Karen L. Marquis

Transgenic mice (Tg2576) overexpressing the Swedish mutation of the human amyloid precursor protein display biochemical, pathological, and behavioral markers consistent with many aspects of Alzheimers disease, including impaired hippocampal function. Impaired, hippocampal-dependent, contextual fear conditioning (CFC) is observed in mice as young as 20 weeks of age. This impairment can be attenuated after treatment before training with the phosphodiesterase-4 inhibitor rolipram (0.1 mg/kg, i.p.). A rolipram-associated improvement is also observed in the littermate controls, suggesting that the effect of rolipram is independent of β-amyloid. Acute treatment before training (but not after training or before testing) with the γ-secretase inhibitor (GSI) N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine-t-butylester (DAPT), at a dose that reduces brain concentrations of β-amyloid (100 mg/kg), attenuates the impairment in 20- to 65-week-old Tg2576 mice. Importantly, DAPT had no effect on performance of control littermates. These data are supportive of a role of β-amyloid in the impairment of CFC in Tg2576 mice. Furthermore, they suggest that acute treatment with GSI may provide improved cognitive functioning as well as disease-modifying effects in Alzheimers disease.


The Journal of Neuroscience | 2008

Impact of Apolipoprotein E (ApoE) Polymorphism on Brain ApoE Levels

Riddell Dr; Hua Zhou; Kevin Atchison; Warwick Hk; Atkinson Pj; Jefferson J; Xu L; Suzan Aschmies; Kirksey Y; Hu Y; Wagner E; Parratt A; Xu J; Li Z; Zaleska Mm; Jacobsen Js; Menelas N. Pangalos; Peter Reinhart

Inheritance of the apoE4 allele (ε4) increases the risk of developing Alzheimers disease; however, the mechanisms underlying this association remain elusive. Recent data suggest that inheritance of ε4 may lead to reduced apoE protein levels in the CNS. We therefore examined apoE protein levels in the brains, CSF and plasma of ε2/2, ε3/3, and ε4/4 targeted replacement mice. These apoE mice showed a genotype-dependent decrease in apoE levels; ε2/2 >ε3/3 >ε4/4. Next, we sought to examine the relative contributions of apoE4 and apoE3 in the ε3/4 mouse brains. ApoE4 represented 30–40% of the total apoE. Moreover, the absolute amount of apoE3 per allele was similar between ε3/3 and ε3/4 mice, implying that the reduced levels of total apoE in ε3/4 mice can be explained by the reduction in apoE4 levels. In culture medium from ε3/4 human astrocytoma or ε3/3, ε4/4 and ε3/4 primary astrocytes, apoE4 levels were consistently lower than apoE3. Secreted cholesterol levels were also lower from ε4/4 astrocytes. Pulse-chase experiments showed an enhanced degradation and reduced half-life of newly synthesized apoE4 compared with apoE3. Together, these data suggest that astrocytes preferentially degrade apoE4, leading to reduced apoE4 secretion and ultimately to reduced brain apoE levels. Moreover, the genotype-dependent decrease in CNS apoE levels, mirror the relative risk of developing AD, and suggest that low levels of total apoE exhibited by ε4 carriers may directly contribute to the disease progression, perhaps by reducing the capacity of apoE to promote synaptic repair and/or Aβ clearance.


Molecular and Cellular Neuroscience | 2007

The LXR agonist TO901317 selectively lowers hippocampal Aβ42 and improves memory in the Tg2576 mouse model of Alzheimer's disease

David Riddell; Hua Zhou; Thomas A. Comery; Evguenia Kouranova; C. Frederick Lo; Helen K. Warwick; Robert H. Ring; Suzan Aschmies; Jane Xu; Katie Kubek; Warren D. Hirst; Catherine Gonzales; Yi Chen; Erin Murphy; Sarah K. Leonard; Dmytro Vasylyev; Aram Oganesian; Robert Martone; Menelas N. Pangalos; Peter Reinhart; J. Steve Jacobsen

Recent studies show that intracellular cholesterol levels can modulate the processing of amyloid precursor protein to Abeta peptide. Moreover, cholesterol-rich apoE-containing lipoproteins may also promote Abeta clearance. Agonists of the liver X receptor (LXR) transcriptionally induce genes involved in intracellular lipid efflux and transport, including apoE. Thus, LXR agonists have the potential to both inhibit APP processing and promote Abeta clearance. Here we show that LXR agonist, TO901317, increased hippocampal ABCA1 and apoE and decreased Abeta42 levels in APP transgenic mice. TO901317 had no significant effects on levels of Abeta40, full length APP, or the APP processing products. Next, we examined the effects of TO901317 in the contextual fear conditioning paradigm; TO901317 completely reversed the contextual memory deficit in these mice. These data demonstrate that LXR agonists do not directly inhibit APP processing but rather facilitate the clearance of Abeta42 and may represent a novel therapeutic approach to Alzheimers disease.


Journal of Pharmacology and Experimental Therapeutics | 2009

Begacestat (GSI-953): A Novel, Selective Thiophene Sulfonamide Inhibitor of Amyloid Precursor Protein γ-Secretase for the Treatment of Alzheimer's Disease

Robert Martone; Hua Zhou; Kevin Atchison; Thomas A. Comery; Jane Z. Xu; Xinyi Huang; Xioahai Gong; Mei Jin; Anthony F. Kreft; Boyd L. Harrison; Scott Christian Mayer; Suzan Aschmies; Cathleen Gonzales; Margaret M. Zaleska; David Riddell; Erik Wagner; Peimin Lu; Shaiu-Ching Sun; June Sonnenberg-Reines; Aram Oganesian; Karissa Adkins; Michael W. Leach; David W. Clarke; Donna M. Huryn; Magid Abou-Gharbia; Ronald L. Magolda; Glen S. Frick; Sangeeta Raje; S. Bradley Forlow; Carrie Balliet

The presenilin containing γ-secretase complex is responsible for the regulated intramembraneous proteolysis of the amyloid precursor protein (APP), the Notch receptor, and a multitude of other substrates. γ-Secretase catalyzes the final step in the generation of Aβ40 and Aβ42 peptides from APP. Amyloid β-peptides (Aβ peptides) aggregate to form neurotoxic oligomers, senile plaques, and congophilic angiopathy, some of the cardinal pathologies associated with Alzheimers disease. Although inhibition of this protease acting on APP may result in potentially therapeutic reductions of neurotoxic Aβ peptides, nonselective inhibition of the enzyme may cause severe adverse events as a result of impaired Notch receptor processing. Here, we report the preclinical pharmacological profile of GSI-953 (begacestat), a novel thiophene sulfonamide γ-secretase inhibitor (GSI) that selectively inhibits cleavage of APP over Notch. This GSI inhibits Aβ production with low nanomolar potency in cellular and cell-free assays of γ-secretase function, and displaces a tritiated analog of GSI-953 from enriched γ-secretase enzyme complexes with similar potency. Cellular assays of Notch cleavage reveal that this compound is approximately 16-fold selective for the inhibition of APP cleavage. In the human APP-overexpressing Tg2576 transgenic mouse, treatment with this orally active compound results in a robust reduction in brain, plasma, and cerebral spinal fluid Aβ levels, and a reversal of contextual fear-conditioning deficits that are correlated with Aβ load. In healthy human volunteers, oral administration of a single dose of GSI-953 produces dose-dependent changes in plasma Aβ levels, confirming pharmacodynamic activity of GSI-953 in humans.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Enhanced clearance of Aβ in brain by sustaining the plasmin proteolysis cascade

J. Steven Jacobsen; Thomas A. Comery; Robert Martone; Hassan Mahmoud Elokdah; David L. Crandall; Aram Oganesian; Suzan Aschmies; Cathleen Gonzales; Jane Xu; Hua Zhou; Kevin Atchison; Erik Wagner; Margaret M. Zaleska; Indranil Das; Robert Arias; David Riddell; Stephen J. Gardell; Magid Abou-Gharbia; Albert Jean Robichaud; Ronald L. Magolda; George P. Vlasuk; Thorir Bjornsson; Peter Reinhart; Menelas N. Pangalos

The amyloid hypothesis states that a variety of neurotoxic β-amyloid (Aβ) species contribute to the pathogenesis of Alzheimers disease. Accordingly, a key determinant of disease onset and progression is the appropriate balance between Aβ production and clearance. Enzymes responsible for the degradation of Aβ are not well understood, and, thus far, it has not been possible to enhance Aβ catabolism by pharmacological manipulation. We provide evidence that Aβ catabolism is increased after inhibition of plasminogen activator inhibitor-1 (PAI-1) and may constitute a viable therapeutic approach for lowering brain Aβ levels. PAI-1 inhibits the activity of tissue plasminogen activator (tPA), an enzyme that cleaves plasminogen to generate plasmin, a protease that degrades Aβ oligomers and monomers. Because tPA, plasminogen and PAI-1 are expressed in the brain, we tested the hypothesis that inhibitors of PAI-1 will enhance the proteolytic clearance of brain Aβ. Our data demonstrate that PAI-1 inhibitors augment the activity of tPA and plasmin in hippocampus, significantly lower plasma and brain Aβ levels, restore long-term potentiation deficits in hippocampal slices from transgenic Aβ-producing mice, and reverse cognitive deficits in these mice.


Journal of Medicinal Chemistry | 2008

Discovery of Begacestat, a Notch-1-Sparing γ-Secretase Inhibitor for the Treatment of Alzheimer's Disease

Scott Christian Mayer; Anthony F. Kreft; Boyd L. Harrison; Magid Abou-Gharbia; Madelene Antane; Suzan Aschmies; Kevin Atchison; Michael Chlenov; Derek Cecil Cole; Thomas A. Comery; George Diamantidis; John W. Ellingboe; Kristi Fan; Rocco John Galante; Cathleen Gonzales; Douglas M. Ho; Molly Hoke; Yun Hu; Donna M. Huryn; Uday Jain; Mei Jin; Kenneth Alfred Martin Kremer; Dennis M. Kubrak; Melissa Lin; Peimin Lu; Ron Magolda; Robert Martone; William M. Moore; Aram Oganesian; Menelas N. Pangalos

SAR on HTS hits 1 and 2 led to the potent, Notch-1-sparing GSI 9, which lowered brain Abeta in Tg2576 mice at 100 mg/kg po. Converting the metabolically labile methyl groups in 9 to trifluoromethyl groups afforded the more stable analogue 10, which had improved in vivo potency. Further side chain modification afforded the potent Notch-1-sparing GSI begacestat (5), which was selected for development for the treatment of Alzheimers disease.


Journal of Pharmacology and Experimental Therapeutics | 2008

Correlating Efficacy in Rodent Cognition Models with in Vivo 5-Hydroxytryptamine1A Receptor Occupancy by a Novel Antagonist, (R)-N-(2-Methyl-(4-indolyl-1-piperazinyl)ethyl)-N-(2-pyridinyl)-cyclohexane Carboxamide (WAY-101405)

Warren D. Hirst; Terrance H. Andree; Suzan Aschmies; Wayne E. Childers; Thomas A. Comery; Lee A. Dawson; Mark L. Day; Irene B. Feingold; Steven M. Grauer; Boyd L. Harrison; Zoe A. Hughes; John Kao; Michael G. Kelly; Heidi van der Lee; Sharon Rosenzweig-Lipson; Annmarie Saab; Deborah L. Smith; Kelly Sullivan; Stacey J. Sukoff Rizzo; Cesario O. Tio; Mei-Yi Zhang; Lee E. Schechter

5-Hydroxytryptamine (5-HT)1A receptors play an important role in multiple cognitive processes, and compelling evidence suggests that 5-HT1A antagonists can reverse cognitive impairment. We have examined the therapeutic potential of a potent (Ki = 1.1 nM), selective (>100-fold), orally bioavailable, silent 5-HT1A receptor antagonist (KB = 1.3 nM) (R)-N-(2-methyl-(4-indolyl-1-piperazinyl)-ethyl)-N-(2-pyridinyl)-cyclohexane carboxamide (WAY-101405). Oral administration of WAY-101405 was shown to be effective in multiple rodent models of learning and memory. In a novel object recognition paradigm, 1 mg/kg enhanced retention (memory) for previously learned information, and it was able to reverse the memory deficits induced by scopolamine. WAY-101405 (1 mg/kg) was also able to reverse scopolamine-induced deficits in a rat contextual fear conditioning model. In the Morris water maze, WAY-101405 (3 mg/kg) significantly improved learning in a paradigm of increasing task difficulty. In vivo microdialysis studies in the dorsal hippocampus of freely moving adult rats demonstrated that acute administration of WAY-101405 (10 mg/kg) increased extracellular acetylcholine levels. The selective radioligand [3H]WAY-100635, administered i.v., was used for in vivo receptor occupancy studies, where WAY-101405 occupied 5-HT1A receptors in the rat cortex, with an ED50 value of 0.1 mg/kg p.o. Taken together, these studies demonstrate that WAY-101405 is a potent and selective, brain penetrant, orally bioavailable 5-HT1A receptor “silent” antagonist that is effective in preclinical memory paradigms at doses where approximately 90% of the postsynaptic 5-HT1A receptors are occupied. These results further support the rationale for use of this compound class in the treatment of cognitive dysfunction associated with psychiatric and neurological conditions.


Journal of Medicinal Chemistry | 2010

Novel Alpha-7 Nicotinic Acetylcholine Receptor Agonists Containing a Urea Moiety: Identification and Characterization of the Potent, Selective, and Orally Efficacious Agonist 1-[6-(4-Fluorophenyl)pyridin-3-yl]-3-(4-piperidin-1-ylbutyl) Urea (SEN34625/WYE-103914)

Chiara Ghiron; Simon N. Haydar; Suzan Aschmies; Hendrick Bothmann; Cristiana Castaldo; Giuseppe Cocconcelli; Thomas A. Comery; Li Di; John Dunlop; Tim Lock; Angela Kramer; Dianne Kowal; Flora Jow; Steve Grauer; Boyd L. Harrison; Salvatore La Rosa; Laura Maccari; Karen L. Marquis; Iolanda Micco; Arianna Nencini; Joanna Quinn; Albert Jean Robichaud; Renza Roncarati; Carla Scali; Georg C. Terstappen; Elisa Turlizzi; Michela Valacchi; Maurizio Varrone; Riccardo Zanaletti; Ugo Zanelli

Alpha-7 nicotinic acetylcholine receptor (alpha7 nAChR) agonists are promising therapeutic candidates for the treatment of cognitive impairment. We report a series of novel, potent small molecule agonists (4-18) of the alpha7 nAChR deriving from our continuing efforts in the areas of Alzheimers disease and schizophrenia. One of the compounds of the series containing a urea moiety (16) was further shown to be a selective agonist of the alpha7 nAChR with excellent in vitro and in vivo profiles, brain penetration, and oral bioavailability and demonstrated in vivo efficacy in multiple behavioral cognition models. Structural modifications leading to the improved selectivity profile and the biological evaluation of this series of compounds are discussed.


Bioorganic & Medicinal Chemistry Letters | 2008

Discovery of a novel series of Notch-sparing γ-secretase inhibitors

Anthony F. Kreft; Boyd L. Harrison; Suzan Aschmies; Kevin Atchison; David S. Casebier; Derek Cecil Cole; George Diamantidis; John W. Ellingboe; Diane B. Hauze; Yun Hu; Donna M. Huryn; Mei Jin; Dennis M. Kubrak; Peimin Lu; Joseph T. Lundquist; Charles William Mann; Robert Martone; William M. Moore; Aram Oganesian; Alex Porte; Dave R. Riddell; June Sonnenberg-Reines; Joseph Raymond Stock; Shaiu-Ching Sun; Erik Wagner; Kevin R. Woller; Zheng Xu; Hua Zhou; J. Steven Jacobsen

Using a cell-based assay, we have identified a new series of Notch-sparing gamma-secretase inhibitors from HTS screening leads 2a and 2e. Lead optimization studies led to the discovery of analog 8e with improved gamma-secretase inhibitory potency and Notch-sparing selectivity.


Bioorganic & Medicinal Chemistry Letters | 2009

(S)-N-(5-Chlorothiophene-2-sulfonyl)-β,β-diethylalaninol a Notch-1-sparing γ-secretase inhibitor

Derek Cecil Cole; Joseph Raymond Stock; Anthony F. Kreft; Madelene Antane; Suzan Aschmies; Kevin Atchison; David S. Casebier; Thomas A. Comery; George Diamantidis; John W. Ellingboe; Boyd L. Harrison; Yun Hu; Mei Jin; Dennis M. Kubrak; Peimin Lu; Charles William Mann; Robert Martone; William Jay Moore; Aram Oganesian; David Riddell; June Sonnenberg-Reines; Shaiu-Ching Sun; Erik Wagner; Zheng Wang; Kevin R. Woller; Zheng Xu; Hua Zhou; J. Steven Jacobsen

Accumulation of beta-amyloid (Abeta), produced by the proteolytic cleavage of amyloid precursor protein (APP) by beta- and gamma-secretase, is widely believed to be associated with Alzheimers disease (AD). Research around the high-throughput screening hit (S)-4-chlorophenylsulfonyl isoleucinol led to the identification of the Notch-1-sparing (9.5-fold) gamma-secretase inhibitor (S)-N-(5-chlorothiophene-2-sulfonyl)-beta,beta-diethylalaninol 7.b.2 (Abeta(40/42) EC(50)=28 nM), which is efficacious in reduction of Abeta production in vivo.

Collaboration


Dive into the Suzan Aschmies's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hua Zhou

Princeton University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge