Network


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

Hotspot


Dive into the research topics where Maria S. Michener is active.

Publication


Featured researches published by Maria S. Michener.


The Journal of Neuroscience | 2010

Acute gamma-secretase inhibition of nonhuman primate CNS shifts amyloid precursor protein (APP) metabolism from amyloid-beta production to alternative APP fragments without amyloid-beta rebound

Jacquelynn J. Cook; Kristin R. Wildsmith; David Gilberto; Marie A. Holahan; Gene G. Kinney; Parker Mathers; Maria S. Michener; Eric A. Price; Mark S. Shearman; Adam J. Simon; Jennifer X. Wang; Guoxin Wu; Kevin E. Yarasheski; Randall J. Bateman

The accumulation of amyloid β (Aβ) in Alzheimers disease is caused by an imbalance of production and clearance, which leads to increased soluble Aβ species and extracellular plaque formation in the brain. Multiple Aβ-lowering therapies are currently in development: an important goal is to characterize the molecular mechanisms of action and effects on physiological processing of Aβ, as well as other amyloid precursor protein (APP) metabolites, in models which approximate human Aβ physiology. To this end, we report the translation of the human in vivo stable-isotope-labeling kinetics (SILK) method to a rhesus monkey cisterna magna ported (CMP) nonhuman primate model, and use the model to test the mechanisms of action of a γ-secretase inhibitor (GSI). A major concern of inhibiting the enzymes which produce Aβ (β- and γ-secretase) is that precursors of Aβ may accumulate and cause a rapid increase in Aβ production when enzyme inhibition discontinues. In this study, the GSI MK-0752 was administered to conscious CMP rhesus monkeys in conjunction with in vivo stable-isotope-labeling, and dose-dependently reduced newly generated CNS Aβ. In contrast to systemic Aβ metabolism, CNS Aβ production was not increased after the GSI was cleared. These results indicate that most of the CNS APP was metabolized to products other than Aβ, including C-terminal truncated forms of Aβ: 1-14, 1-15 and 1-16; this demonstrates an alternative degradation pathway for CNS amyloid precursor protein during γ-secretase inhibition.


Journal of Pharmacology and Experimental Therapeutics | 2010

Pharmacological Properties of MK-3207, a Potent and Orally Active Calcitonin Gene-Related Peptide Receptor Antagonist

Christopher A. Salvatore; Eric L. Moore; Amy Calamari; Jacquelynn J. Cook; Maria S. Michener; Stacey O'Malley; Patricia Miller; Cyrille Sur; David L. Williams; Zhizhen Zeng; Andrew Danziger; Joseph J. Lynch; Christopher P. Regan; John F. Fay; Yui S. Tang; Chi-Chung Li; Nicole T. Pudvah; Rebecca B. White; Ian M. Bell; Steven N. Gallicchio; Samuel Graham; Harold G. Selnick; Joseph P. Vacca; Stefanie A. Kane

Calcitonin gene-related peptide (CGRP) has long been hypothesized to play a key role in migraine pathophysiology, and the advent of small-molecule antagonists has clearly demonstrated a clinical link between blocking the CGRP receptor and migraine efficacy. 2-[(8R)-8-(3,5-Difluorophenyl)-10-oxo-6,9-diazaspiro[4.5]dec-9-yl]-N-[(2R)-2′-oxo-1,1′,2′,3-tetrahydrospiro[indene-2,3′-pyrrolo[2,3-b]pyridin]-5-yl]acetamide (MK-3207) represents the third CGRP receptor antagonist to display clinical efficacy in migraine trials. Here, we report the pharmacological characterization of MK-3207, a potent and orally bioavailable CGRP receptor antagonist. In vitro, MK-3207 is a potent antagonist of the human and rhesus monkey CGRP receptors (Ki = 0.024 nM). In common with other CGRP receptor antagonists, MK-3207 displays lower affinity for CGRP receptors from other species, including canine and rodent. As a consequence of species selectivity, the in vivo potency was assessed in a rhesus monkey pharmacodynamic assay measuring capsaicin-induced changes in forearm dermal blood flow via laser Doppler imaging. MK-3207 produced a concentration-dependent inhibition of dermal vasodilation, with plasma concentrations of 0.8 and 7 nM required to block 50 and 90% of the blood flow increase, respectively. The tritiated analog [3H]MK-3207 was used to study the binding characteristics on the human CGRP receptor. [3H]MK-3207 displayed reversible and saturable binding (KD = 0.06 nM), and the off-rate was determined to be 0.012 min−1, with a t1/2 value of 59 min. In vitro autoradiography studies on rhesus monkey brain slices identified the highest level of binding in the cerebellum, brainstem, and meninges. Finally, as an index of central nervous system penetrability, the in vivo cerebrospinal fluid/plasma ratio was determined to be 2 to 3% in cisterna magna-ported rhesus monkeys.


Journal of Neuroscience Research | 2012

Characterization of plasma β-secretase (BACE1) activity and soluble amyloid precursor proteins as potential biomarkers for Alzheimer's disease

Guoxin Wu; Sethu Sankaranarayanan; Jacky Wong; Katherine Tugusheva; Maria S. Michener; Xiao-Ping Shi; Jacquelynn J. Cook; Adam J. Simon; Mary J. Savage

Reduction in cerebrospinal fluid (CSF) amyloid β42 (Aβ42) and elevation in total tau and phospho‐thr181 tau consistently differentiate between Alzheimers disease (AD) and age‐matched control subjects. In contrast, CSF β‐site APP‐cleaving enzyme activity (BACE1) and soluble amyloid precursor proteins α and β (sAPPα and sAPPβ) are without consistent patterns in AD subjects. Plasma sampling is much easier, with fewer side effects, and is readily applied in primary care centers, so we have developed and validated novel plasma BACE activity, sAPPβ, and sAPPα assays and investigated their ability to distinguish AD from age‐matched controls. Plasma BACE activity assay was sensitive and specific, with signal being immunodepleted with a specific BACE1 antibody and inhibited with a BACE1‐specific inhibitor. Plasma sAPPβ and sAPPα assays were specific, with signal diluting linearly, immunodepleted with specific antibodies, and at background levels in APP knockout mice. In rhesus monkeys, BACE1 but not γ‐secretase inhibitor led to significant lowering of plasma sAPPβ with concurrent elevation of plasma sAPPα. AD subjects showed a significant increase in plasma BACE1 activity, sAPPβ, sAPPα, and Aβ42 (P < 0.001) compared with age‐matched controls. In conclusion, plasma BACE activity and sAPP endpoints provide novel investigative biomarkers for AD diagnosis and potential pharmacodynamic biomarkers for secretase inhibitor studies.


The Journal of Neuroscience | 2014

CNS Amyloid-β, Soluble APP-α and -β Kinetics during BACE Inhibition

Justyna Dobrowolska; Maria S. Michener; Guoxin Wu; Bruce W. Patterson; Robert Chott; Vitaliy Ovod; Yuriy Pyatkivskyy; Kristin R. Wildsmith; Tom Kasten; Parker Mathers; Mandy Dancho; Christina Lennox; Brad Smith; David Gilberto; Debra McLoughlin; Daniel J. Holder; Andrew W. Stamford; Kevin E. Yarasheski; Matthew E. Kennedy; Mary J. Savage; Randall J. Bateman

BACE, a β-secretase, is an attractive potential disease-modifying therapeutic strategy for Alzheimers disease (AD) as it results directly in the decrease of amyloid precursor protein (APP) processing through the β-secretase pathway and a lowering of CNS amyloid-β (Aβ) levels. The interaction of the β-secretase and α-secretase pathway-mediated processing of APP in the rhesus monkey (nonhuman primate; NHP) CNS is not understood. We hypothesized that CNS inhibition of BACE would result in decreased newly generated Aβ and soluble APPβ (sAPPβ), with increased newly generated sAPPα. A stable isotope labeling kinetics experiment in NHPs was performed with a 13C6-leucine infusion protocol to evaluate effects of BACE inhibition on CNS APP processing by measuring the kinetics of sAPPα, sAPPβ, and Aβ in CSF. Each NHP received a low, medium, or high dose of MBI-5 (BACE inhibitor) or vehicle in a four-way crossover design. CSF sAPPα, sAPPβ, and Aβ were measured by ELISA and newly incorporated label following immunoprecipitation and liquid chromatography-mass spectrometry. Concentrations, kinetics, and amount of newly generated APP fragments were calculated. sAPPβ and sAPPα kinetics were similar, but both significantly slower than Aβ. BACE inhibition resulted in decreased labeled sAPPβ and Aβ in CSF, without observable changes in labeled CSF sAPPα. ELISA concentrations of sAPPβ and Aβ both decreased and sAPPα increased. sAPPα increased by ELISA, with no difference by labeled sAPPα kinetics indicating increases in product may be due to APP shunting from the β-secretase to the α-secretase pathway. These results provide a quantitative understanding of pharmacodynamic effects of BACE inhibition on NHP CNS, which can inform about target development.


Bioorganic & Medicinal Chemistry Letters | 2010

Rapid P1 SAR of brain penetrant tertiary carbinamine derived BACE inhibitors

Hong Zhu; Mary B. Young; Philippe G. Nantermet; Samuel L. Graham; Dennis Colussi; Ming-Tain Lai; Beth Pietrak; Eric A. Price; Sethu Sankaranarayanan; Xiao-Ping Shi; Katherine Tugusheva; Marie A. Holahan; Maria S. Michener; Jacquelynn J. Cook; Adam J. Simon; Daria J. Hazuda; Joseph P. Vacca; Hemaka A. Rajapakse

This Letter describes the one pot synthesis of tertiary carbinamine 3 and related analogs of brain penetrant BACE-1 inhibitors via the alkylation of the Schiff base intermediate 2. The methodology developed for this study provided a convenient and rapid means to explore the P1 region of these types of inhibitors, where the P1 group is installed in the final step using a one-pot two-step protocol. Further SAR studies led to the identification of 10 which is twofold more potent in vitro as compared to the lead compound. This inhibitor was characterized in a cisterna magna ported rhesus monkey model, where significant lowering of CSF Abeta40 was observed.


Biochemical Pharmacology | 2009

Effect of P-glycoprotein-mediated efflux on cerebrospinal fluid concentrations in rhesus monkeys.

Cuyue Tang; Yuhsin Kuo; Nicole T. Pudvah; Joan D. Ellis; Maria S. Michener; Melissa S. Egbertson; Samuel L. Graham; Jacquelynn J. Cook; Jerome H. Hochman; Thomayant Prueksaritanont

Brain penetration of drugs which are subject to P-glycoprotein (Pgp)-mediated efflux is attenuated, as manifested by the fact that the cerebrospinal fluid concentration (C(CSF)), a good surrogate of the unbound brain concentration (C(ub)), is lower than the unbound plasma concentration (C(up)) for Pgp substrates. In rodents, the attenuation magnitude of brain penetration by Pgp-mediated efflux has been estimated by correlating the ratio of CSF to plasma exposures (C(CSF)/C(p)) with the unbound fraction in plasma (f(u)) upon the incorporation of the in vivo or in vitro Pgp-mediated efflux ratios (ERs). In the present work, we investigated the impact of Pgp-mediated efflux on C(CSF) in monkeys. Following intravenous administration to cisterna magna ported rhesus monkeys, the CSF and plasma concentrations were determined for 25 compounds from three discovery programs. We also evaluated their f(u) in rhesus plasma and ER in human and African green monkey MDR-transfected LLC-PK1 cells. These compounds varied significantly in the f(u) (0.025-0.73), and 24 out of 25 are considered Pgp substrates based on their appreciable directional transport (ER>2). The C(CSF)/C(p) was significantly lower than the corresponding f(u) (>or=3-fold) for 16 compounds regardless of a significant correlation (R(2)=0.59, p=4 x 10(-5)) when the C(CSF)/C(p) was plotted against the f(u). When the f(u) was normalized to the ER (f(u)/ER) the correlation was improved (R(2)=0.75, p=8 x 10(-8)). More importantly, only one compound showed the C(CSF)/C(p) that exceeded 3-fold of the normalized f(u). The results suggest that the impact of Pgp-mediated efflux in monkeys, similar to the case in rodents, is reasonably reflected by the gradient between the free concentrations in plasma and in CSF. Therefore, f(u) and Pgp ER may serve as useful measurements in estimating in vivo C(CSF)/C(p) ratios in monkeys, and potentially in humans.


ChemMedChem | 2009

Evolution of tertiary carbinamine BACE-1 inhibitors: Aβ reduction in rhesus CSF upon oral dosing

Philippe G. Nantermet; Hemaka A. Rajapakse; Mathew Stanton; Shaun R. Stauffer; James C. Barrow; Allison R. Gregro; Keith P. Moore; Melissa A. Steinbeiser; John Swestock; Harold G. Selnick; Samuel L. Graham; Georgia B. McGaughey; Dennis Colussi; Ming Tain Lai; Sethu Sankaranarayanan; Adam J. Simon; Sanjeev Munshi; Jacquelynn J. Cook; Marie A. Holahan; Maria S. Michener; Joseph P. Vacca

Evolution of Tertiary Carbinamine BACE-1 Inhibitors: Ab Reduction in Rhesus CSF upon Oral Dosing Philippe G. Nantermet,* Hemaka A. Rajapakse, Mathew G. Stanton, Shaun R. Stauffer, James C. Barrow, Allison R. Gregro, Keith P. Moore, Melissa A. Steinbeiser, John Swestock, Harold G. Selnick, Samuel L. Graham, Georgia B. McGaughey, Dennis Colussi, Ming-Tain Lai, Sethu Sankaranarayanan, Adam J. Simon, Sanjeev Munshi, Jacquelynn J. Cook, Marie A. Holahan, Maria S. Michener, and Joseph P. Vacca


European Journal of Pharmacology | 2015

Platelet transfusion reverses bleeding evoked by triple anti-platelet therapy including vorapaxar, a novel platelet thrombin receptor antagonist

Tian-Quan Cai; L. Alexandra Wickham; Gary R. Sitko; Maria S. Michener; Richard Raubertas; Larry Handt; Madhu Chintala; Dietmar Seiffert; Michael J. Forrest

Vorapaxar is a novel protease-activated receptor-1 (PAR-1) antagonist recently approved for the reduction of thrombotic cardiovascular events in patients with a history of myocardial infarction or with peripheral arterial disease. Patients who received vorapaxar in addition to standard of care antiplatelet therapy had an increased incidence of major bleeding events compared with placebo. To assess whether platelet transfusion can restore hemostasis in primates on triple antiplatelet therapy, template bleeding times were assessed concurrently in the buccal mucosa, finger pad, and distolateral tail of anesthetized cynomolgus macaques to evaluate bleeding with vorapaxar as either monotherapy or in combination with aspirin or aspirin and clopidogrel. Aspirin (5mg/kg, IV) or vorapaxar (1mg/kg, PO) alone had no significant effect on bleeding times in the three vascular beds examined. A modest (<2-fold) increase in bleeding time was achieved in the three beds with the dual combination of aspirin and vorapaxar. Major increases in bleeding time were achieved in the three beds with the triple combination of aspirin (5mg/kg, IV), vorapaxar (1mg/kg, PO), and clopidogrel (1mg/kg, PO). Transfusion of fresh human platelet rich plasma, but not platelet poor plasma, reversed the increase in bleeding time in the triple therapy group. Transfusion of human platelets may be a viable approach in situations requiring a rapid reversal of platelet function in individuals treated with triple anti-platelet therapy that includes vorapaxar.


Journal of Medicinal Chemistry | 2016

Identification and in Vivo Evaluation of Liver X Receptor β-Selective Agonists for the Potential Treatment of Alzheimer’s Disease

Shawn J. Stachel; Celina Zerbinatti; Michael T. Rudd; Mali Cosden; Sokreine Suon; Kausik K. Nanda; Keith Wessner; Jillian DiMuzio; Jill Maxwell; Zhenhua Wu; Jason M. Uslaner; Maria S. Michener; Peter Szczerba; Edward J. Brnardic; Vanessa Rada; Yuntae Kim; Robert S. Meissner; Peter Wuelfing; Yang Yuan; Jeanine Ballard; Marie A. Holahan; Daniel Klein; Jun Lu; Xavier Fradera; Gopal Parthasarathy; Victor N. Uebele; Zhongguo Chen; Yingjie Li; Jian Li; Andrew John Cooke

Herein, we describe the development of a functionally selective liver X receptor β (LXRβ) agonist series optimized for Emax selectivity, solubility, and physical properties to allow efficacy and safety studies in vivo. Compound 9 showed central pharmacodynamic effects in rodent models, evidenced by statistically significant increases in apolipoprotein E (apoE) and ATP-binding cassette transporter levels in the brain, along with a greatly improved peripheral lipid safety profile when compared to those of full dual agonists. These findings were replicated by subchronic dosing studies in non-human primates, where cerebrospinal fluid levels of apoE and amyloid-β peptides were increased concomitantly with an improved peripheral lipid profile relative to that of nonselective compounds. These results suggest that optimization of LXR agonists for Emax selectivity may have the potential to circumvent the adverse lipid-related effects of hepatic LXR activity.


Bioorganic & Medicinal Chemistry Letters | 2015

Discovery of novel triazolobenzazepinones as γ-secretase modulators with central Aβ42 lowering in rodents and rhesus monkeys.

Christian Fischer; Susan L. Zultanski; Hua Zhou; Joey L. Methot; Sanjiv Shah; Ikuo Hayashi; Bethany Hughes; Christopher M. Moxham; Nathan Bays; Nadya Smotrov; Armetta D. Hill; Bo-Sheng Pan; Zhenhua Wu; Lily Y. Moy; Flobert Tanga; Candia M. Kenific; Jonathan C. Cruz; Deborah Walker; Melanie Bouthillette; George Nikov; Sujal V. Deshmukh; Valentina V. Jeliazkova-Mecheva; Damaris Diaz; Maria S. Michener; Jacquelynn J. Cook; Benito Munoz; Mark S. Shearman

Synthesis and SAR studies of novel triazolobenzazepinones as gamma secretase modulators (GSMs) are presented in this communication. Starting from our azepinone leads, optimization studies toward improving central lowering of Aβ42 led to the discovery of novel benzo-fused azepinones. Several benzazepinones were profiled in vivo and found to lower brain Aβ42 levels in Sprague Dawley rats and transgenic APP-YAC mice in a dose-dependent manner after a single oral dose. Compound 34 was further progressed into a pilot study in our cisterna-magna-ported rhesus monkey model, where we observed robust lowering of CSF Aβ42 levels.

Collaboration


Dive into the Maria S. Michener's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mary J. Savage

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Guoxin Wu

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Adam J. Simon

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Marie A. Holahan

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Sethu Sankaranarayanan

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Brad Smith

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Christina Lennox

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

David Gilberto

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge