Network


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

Hotspot


Dive into the research topics where Cindy E. Nuss is active.

Publication


Featured researches published by Cindy E. Nuss.


Journal of Medicinal Chemistry | 2008

Design, Synthesis, and Evaluation of a Novel 4-Aminomethyl-4-fluoropiperidine as a T-Type Ca2+ Channel Antagonist

William D. Shipe; James C. Barrow; Zhi Qiang Yang; Craig W. Lindsley; F. Vivien Yang; Kelly Ann S. Schlegel; Youheng Shu; Kenneth E. Rittle; Mark G. Bock; George D. Hartman; Cuyue Tang; Jeanine Ballard; Yuhsin Kuo; Emily D. Adarayan; Thomayant Prueksaritanont; Matthew M. Zrada; Victor N. Uebele; Cindy E. Nuss; Thomas M. Connolly; Scott M. Doran; Steven V. Fox; Richard L. Kraus; Michael J. Marino; Valerie Kuzmick Graufelds; Hugo M. Vargas; Patricia B. Bunting; Martha Hasbun-Manning; Rose M. Evans; Kenneth S. Koblan; John J. Renger

The novel T-type antagonist ( S)- 5 has been prepared and evaluated in in vitro and in vivo assays for T-type calcium ion channel activity. Structural modification of the piperidine leads 1 and 2 afforded the fluorinated piperidine ( S)- 5, a potent and selective antagonist that displayed in vivo CNS efficacy without adverse cardiovascular effects.


Journal of Medicinal Chemistry | 2008

Discovery of 1,4-Substituted Piperidines as Potent and Selective Inhibitors of T-Type Calcium Channels

Zhi-Qiang Yang; James C. Barrow; William D. Shipe; Kelly-Ann S. Schlegel; Youheng Shu; F. Vivien Yang; Craig W. Lindsley; Kenneth E. Rittle; Mark G. Bock; George D. Hartman; Victor N. Uebele; Cindy E. Nuss; Steve V. Fox; Richard L. Kraus; Scott M. Doran; Thomas M. Connolly; Cuyue Tang; Jeanine Ballard; Yuhsin Kuo; Emily D. Adarayan; Thomayant Prueksaritanont; Matthew M. Zrada; Michael J. Marino; Valerie Kuzmick Graufelds; Anthony G. DiLella; Ian J. Reynolds; Hugo M. Vargas; Patricia B. Bunting; Richard Woltmann; Michael Magee

The discovery of a novel series of potent and selective T-type calcium channel antagonists is reported. Initial optimization of high-throughput screening leads afforded a 1,4-substituted piperidine amide 6 with good potency and limited selectivity over hERG and L-type channels and other off-target activities. Further SAR on reducing the basicity of the piperidine and introducing polarity led to the discovery of 3-axial fluoropiperidine 30 with a significantly improved selectivity profile. Compound 30 showed good oral bioavailability and brain penetration across species. In a rat genetic model of absence epilepsy, compound 30 demonstrated a robust reduction in the number and duration of seizures at 33 nM plasma concentration, with no cardiovascular effects at up to 5.6 microM. Compound 30 also showed good efficacy in rodent models of essential tremor and Parkinsons disease. Compound 30 thus demonstrates a wide margin between CNS and peripheral effects and is a useful tool for probing the effects of T-type calcium channel inhibition.


Journal of Clinical Investigation | 2009

Antagonism of T-type calcium channels inhibits high-fat diet–induced weight gain in mice

Victor N. Uebele; Anthony L. Gotter; Cindy E. Nuss; Richard L. Kraus; Scott M. Doran; Susan L. Garson; Duane R. Reiss; Yuxing Li; James C. Barrow; Thomas S. Reger; Zhi Qiang Yang; Jeanine Ballard; Cuyue Tang; Joseph M. Metzger; Sheng Ping Wang; Kenneth S. Koblan; John J. Renger

The epidemics of obesity and metabolic disorders have well-recognized health and economic burdens. Pharmacologic treatments for these diseases remain unsatisfactory with respect to both efficacy and side-effect profiles. Here, we have identified a potential central role for T-type calcium channels in regulating body weight maintenance and sleep. Previously, it was shown that mice lacking CaV3.1 T-type calcium channels have altered sleep/wake activity. We found that these mice were also resistant to high-fat diet-induced weight gain, without changes in food intake or sensitivity to high-fat diet-induced disruptions of diurnal rhythm. Administration of a potent and selective antagonist of T-type calcium channels, TTA-A2, to normal-weight animals prior to the inactive phase acutely increased sleep, decreased body core temperature, and prevented high-fat diet-induced weight gain. Administration of TTA-A2 to obese rodents reduced body weight and fat mass while concurrently increasing lean muscle mass. These effects likely result from better alignment of diurnal feeding patterns with daily changes in circadian physiology and potentially an increased metabolic rate during the active phase. Together, these studies reveal what we believe to be a previously unknown role for T-type calcium channels in the regulation of sleep and weight maintenance and suggest the potential for a novel therapeutic approach to treating obesity.


Cell Biochemistry and Biophysics | 2009

Positive allosteric interaction of structurally diverse T-type calcium channel antagonists.

Victor N. Uebele; Cindy E. Nuss; Steven V. Fox; Susan L. Garson; Razvan Cristescu; Scott M. Doran; Richard L. Kraus; Vincent P. Santarelli; Yuxing Li; Æ James C. Barrow; Zhi-Qiang Yang; Kelly-Ann S. Schlegel; Kenneth E. Rittle; Thomas S. Reger; Rodney A. Bednar; Wei Lemaire; Faith A. Mullen; Jeanine Ballard; Cuyue Tang; Ge Dai; Owen B. McManus; Kenneth S. Koblan; John J. Renger

Low-voltage-activated (T-type) calcium channels play a role in diverse physiological responses including neuronal burst firing, hormone secretion, and cell growth. To better understand the biological role and therapeutic potential of the target, a number of structurally diverse antagonists have been identified. Multiple drug interaction sites have been identified for L-type calcium channels, suggesting a similar possibility exists for the structurally related T-type channels. Here, we radiolabel a novel amide T-type calcium channel antagonist (TTA-A1) and show that several known antagonists, including mibefradil, flunarizine, and pimozide, displace binding in a concentration-dependent manner. Further, we identify a novel quinazolinone T-type antagonist (TTA-Q4) that enhanced amide radioligand binding, increased affinity in a saturable manner and slowed dissociation. Functional evaluation showed these compounds to be state-dependent antagonists which show a positive allosteric interaction. Consistent with slowing dissociation, the duration of efficacy was prolonged when compounds were co-administered to WAG/Rij rats, a genetic model of absence epilepsy. The development of a T-type calcium channel radioligand has been used to demonstrate structurally distinct TTAs interact at allosteric sites and to confirm the potential for synergistic inhibition of T-type calcium channels with structurally diverse antagonists.


Bioorganic & Medicinal Chemistry Letters | 2011

Pyridyl amides as potent inhibitors of T-type calcium channels

Thomas S. Reger; Zhi Qiang Yang; Kelly Ann S Schlegel; Youheng Shu; Christa Mattern; Rowena V. Cube; Kenneth E. Rittle; Georgia B. McGaughey; George D. Hartman; Cuyue Tang; Jeanine Ballard; Yuhsin Kuo; Thomayant Prueksaritanont; Cindy E. Nuss; Scott M. Doran; Steven V. Fox; Susan L. Garson; Yuxing Li; Richard L. Kraus; Victor N. Uebele; John J. Renger; James C. Barrow

A novel series of amide T-type calcium channel antagonists were prepared and evaluated using in vitro and in vivo assays. Optimization of the screening hit 3 led to identification of the potent and selective T-type antagonist 37 that displayed in vivo efficacy in rodent models of epilepsy and sleep.


Bioorganic & Medicinal Chemistry Letters | 2010

Discovery and expanded SAR of 4,4-disubstituted quinazolin-2-ones as potent T-type calcium channel antagonists.

Kelly Ann S. Schlegel; Zhi Qiang Yang; Thomas S. Reger; Youheng Shu; Rowena V. Cube; Kenneth E. Rittle; Phung Le Bondiskey; Mark G. Bock; George D. Hartman; Cuyue Tang; Jeanine Ballard; Yuhsin Kuo; Thomayant Prueksaritanont; Cindy E. Nuss; Scott M. Doran; Steven V. Fox; Susan L. Garson; Richard L. Kraus; Yuxing Li; Victor N. Uebele; John J. Renger; James C. Barrow

The discovery and synthesis of 4,4-disubstituted quinazolinones as T-type calcium channel antagonists is reported. Based on lead compounds 2 and 3, a focused SAR campaign driven by the optimization of potency, metabolic stability, and pharmacokinetic profile identified 45 as a potent T-type Ca(2+) channel antagonist with minimized PXR activation. In vivo, 45 suppressed seizure frequency in a rat model of absence epilepsy and showed significant alterations of sleep architecture after oral dosing to rats as measured by EEG.


ACS Medicinal Chemistry Letters | 2010

Discovery of 4,4-Disubstituted Quinazolin-2-ones as T-Type Calcium Channel Antagonists

James C. Barrow; Kenneth E. Rittle; Thomas S. Reger; Zhi Qiang Yang; Phung Le Bondiskey; Georgia B. McGaughey; Mark G. Bock; George D. Hartman; Cuyue Tang; Jeanine Ballard; Yuhsin Kuo; Thomayant Prueksaritanont; Cindy E. Nuss; Scott M. Doran; Steven V. Fox; Susan L. Garson; Richard L. Kraus; Yuxing Li; Michael J. Marino; Valerie Kuzmick Graufelds; Victor N. Uebele; John J. Renger

A novel series of quinazolinone T-type calcium channel antagonists have been prepared and evaluated using in vitro and in vivo assays. Optimization of the screening hit 3 by modifications of the 3- and 4-positions of the quinazolinone ring afforded potent and selective antagonists that displayed in vivo central nervous system efficacy in epilepsy and tremor models, as well as significant effects on rat active wake as measured by electrocorticogram.


Molecular Pharmacology | 2013

Cross modulation and molecular interaction at the Cav3.3 protein between the endogenous lipids and the T-type calcium channel antagonist TTA-A2.

Magali Cazade; Cindy E. Nuss; Isabelle Bidaud; John J. Renger; Victor N. Uebele; Philippe Lory; Jean Chemin

T-type calcium channels (T/Cav3-channels) are implicated in various physiologic and pathophysiologic processes such as epilepsy, sleep disorders, hypertension, and cancer. T-channels are the target of endogenous signaling lipids including the endocannabinoid anandamide, the ω3-fatty acids, and the lipoamino-acids. However, the precise molecular mechanism by which these molecules inhibit T-current is unknown. In this study, we provided a detailed electrophysiologic and pharmacologic analysis indicating that the effects of the major N-acyl derivatives on the Cav3.3 current share many similarities with those of TTA-A2 [(R)-2-(4-cyclopropylphenyl)-N-(1-(5-(2,2,2-trifluoroethoxy)pyridin-2-yl)ethyl)acetamide], a synthetic T-channel inhibitor. Using radioactive binding assays with the TTA-A2 derivative [3H]TTA-A1 [(R)-2-(4-(tert-butyl)phenyl)-N-(1-(5-methoxypyridin-2-yl)ethyl)acetamide], we demonstrated that polyunsaturated lipids, which inhibit the Cav3.3 current, as NAGly (N-arachidonoyl glycine), NASer (N-arachidonoyl-l-serine), anandamide, NADA (N-arachidonoyl dopamine), NATau (N-arachidonoyl taurine), and NA-5HT (N-arachidonoyl serotonin), all displaced [3H]TTA-A1 binding to membranes prepared from cells expressing Cav3.3, with Ki in a micromolar or submicromolar range. In contrast, lipids with a saturated alkyl chain, as N-arachidoyl glycine and N-arachidoyl ethanolamine, which did not inhibit the Cav3.3 current, had no effect on [3H]TTA-A1 binding. Accordingly, bio-active lipids occluded TTA-A2 effect on Cav3.3 current. In addition, TTA-Q4 [(S)-4-(6-chloro-4-cyclopropyl-3-(2,2-difluoroethyl)-2-oxo-1,2,3,4-tetrahydroquinazolin-4-yl)benzonitrile], a positive allosteric modulator of [3H]TTA-A1 binding and TTA-A2 functional inhibition, acted in a synergistic manner to increase lipid-induced inhibition of the Cav3.3 current. Overall, our results demonstrate a common molecular mechanism for the synthetic T-channel inhibitors and the endogenous lipids, and indicate that TTA-A2 and TTA-Q4 could be important pharmacologic tools to dissect the involvement of T-current in the physiologic effects of endogenous lipids.


The Journal of Steroid Biochemistry and Molecular Biology | 2004

Role of voltage-gated calcium channels in potassium-stimulated aldosterone secretion from rat adrenal zona glomerulosa cells.

Victor N. Uebele; Cindy E. Nuss; John J. Renger; Thomas M. Connolly

The mineralocorticoid aldosterone plays an important role in the regulation of plasma electrolyte homeostasis. Exposure of acutely isolated rat adrenal zona glomerulosa cells to elevated K(+) activates voltage-gated calcium channels and initiates a calcium-dependent increase in aldosterone synthesis. We developed a novel 96-well format aldosterone secretion assay to rapidly evaluate the effect of known T- and L-type calcium channel antagonists on K(+)-stimulated aldosterone secretion and better define the role of voltage-gated calcium channels in this process. Reported T-type antagonists, mibefradil and Ni(2+), and selected L-type antagonist dihydropyridines, inhibited K(+)-stimulated aldosterone synthesis. Dihydropyridine-mediated inhibition occurred at concentrations which had no effect on rat alpha1H T-type Ca(2+) currents. In contrast, below 10 microM, the L-type antagonists verapamil and diltiazem showed only minimal inhibitory effects. To examine the selectivity of the calcium channel antagonist-mediated inhibition, we established an aldosterone secretion assay in which 8Br-cAMP stimulates aldosterone secretion independent of extracellular calcium. Mibefradil remained inhibitory in this assay, while the dihydropyridines had only limited effects. Taken together, these data demonstrate a role for the L-type calcium channel in K(+)-stimulated aldosterone secretion. Further, they confirm the need for selective T-type calcium channel antagonists to better address the role of T-type channels in K(+)-stimulated aldosterone secretion.


Neuroreport | 2009

T-type calcium channels regulate cortical plasticity in-vivo

Victor N. Uebele; Cindy E. Nuss; Vincent P. Santarelli; Susan L. Garson; James C. Barrow; Shaun R. Stauffer; Kenneth S. Koblan; John J. Renger; Sara J. Aton; Julie Seibt; Michelle Dumoulin; Sushil K. Jha; Tammi Coleman; Marcos G. Frank

T-type voltage-dependent calcium channels may play an important role in synaptic plasticity, but lack of specific antagonists has hampered investigation into this possible function. We investigated the role of the T-type channel in a canonical model of in-vivo cortical plasticity triggered by monocular deprivation. We identified a compound (TTA-I1) with subnanomolar potency in standard voltage clamp assays and high selectivity for the T-type channel. When infused intracortically, TTA-I1 reduced cortical plasticity triggered by monocular deprivation while preserving normal visual response properties. These results show that the T-type calcium channel plays a central role in cortical plasticity.

Collaboration


Dive into the Cindy E. Nuss's collaboration.

Researchain Logo
Decentralizing Knowledge