Network


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

Hotspot


Dive into the research topics where Richard L. Kraus is active.

Publication


Featured researches published by Richard L. Kraus.


Journal of Neurochemistry | 2005

Antioxidant properties of minocycline: neuroprotection in an oxidative stress assay and direct radical‐scavenging activity

Richard L. Kraus; Rodger Pasieczny; Karen Lariosa-Willingham; Mary Turner; Alice Jiang; John W. Trauger

Minocycline is neuroprotective in animal models of a number of acute CNS injuries and neurodegenerative diseases. While anti‐inflammatory and anti‐apoptotic effects of minocycline have been characterized, the molecular basis for the neuroprotective effects of minocycline remains unclear. We report here that minocycline and a number of antioxidant compounds protect mixed neuronal cultures in an oxidative stress assay. To evaluate the role of minocyclines direct antioxidant properties in neuroprotection, we determined potencies for minocycline, other tetracycline antibiotics, and reference antioxidant compounds using a panel of in vitro radical scavenging assays. Data from in vitro rat brain homogenate lipid peroxidation and 2,2‐diphenyl‐1‐picrylhydrazyl (DPPH) radical scavenging assays show that minocycline, in contrast to tetracycline, is an effective antioxidant with radical scavenging potency similar to vitamin E. Our findings suggest that the direct antioxidant activity of minocycline may contribute to its neuroprotective effects in some cell‐based assays and animal models of neuronal injury.


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 | 2015

Voltage-Gated Sodium Channels: Structure, Function, Pharmacology, and Clinical Indications

Manuel de Lera Ruiz; Richard L. Kraus

The tremendous therapeutic potential of voltage-gated sodium channels (Na(v)s) has been the subject of many studies in the past and is of intense interest today. Na(v)1.7 channels in particular have received much attention recently because of strong genetic validation of their involvement in nociception. Here we summarize the current status of research in the Na(v) field and present the most relevant recent developments with respect to the molecular structure, general physiology, and pharmacology of distinct Na(v) channel subtypes. We discuss Na(v) channel ligands such as small molecules, toxins isolated from animal venoms, and the recently identified Na(v)1.7-selective antibody. Furthermore, we review eight characterized ligand binding sites on the Na(v) channel α subunit. Finally, we examine possible therapeutic applications of Na(v) ligands and provide an update on current clinical studies.


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 Pharmacology and Experimental Therapeutics | 2010

In Vitro Characterization of T-Type Calcium Channel Antagonist TTA-A2 and In Vivo Effects on Arousal in Mice

Richard L. Kraus; Yuxing Li; Yun Gregan; Anthony L. Gotter; Victor N. Uebele; Steven V. Fox; Scott M. Doran; James C. Barrow; Zhi Qiang Yang; Thomas S. Reger; Kenneth S. Koblan; John J. Renger

T-type calcium channels have been implicated in many behaviorally important neurophysiological processes, and altered channel activity has been linked to the pathophysiology of neurological disorders such as insomnia, epilepsy, Parkinsons disease, depression, schizophrenia, and pain. We have previously identified a number of potent and selective T-type channel antagonists (Barrow et al., 2007; Shipe et al., 2008; Yang et al., 2008). Here we describe the properties of the antagonist TTA-A2 [2-(4-cyclopropylphenyl)-N-((1R)-1-{5-[(2,2,2-trifluoroethyl)oxo]-pyridin-2-yl}ethyl)acetamide], assessed in patch-clamp experiments. TTA-A2 blocks T-type channels (Cav3.1, 3.2, 3.3) voltage dependently and with high potency (IC50 ∼100 nM). Stimulation at 3 Hz revealed additional use dependence of inhibition. A hyperpolarized shift of the channel availability curve and delayed channel recovery from inactivation suggest that the compound preferentially interacts with and stabilizes inactivated channels. The compound showed a ∼300-fold selectivity for Cav3 channels over high-voltage activated calcium channels. Inhibitory effects on native T-type currents were confirmed in brain slice recordings from the dorsal lateral geniculate nucleus and the subthalamic nucleus. Furthermore, we demonstrate that in vivo T-type channel inhibition by TTA-A2 suppresses active wake and promotes slow-wave sleep in wild-type mice but not in mice lacking both Cav3.1 and Cav3.3, suggesting the selective effect of TTA-A2 on recurrent thalamocortical network activity. The discovery of the potent and selective T-type channel antagonist TTA-A2 has enabled us to study the in vivo effects of pharmacological T-channel inhibition on arousal in mice, and it will help to explore the validity of these channels as potential drug targets for sleep-related and other neurological diseases.


Molecular Pharmacology | 2007

Inhibition of sodium channel gating by trapping the domain II voltage sensor with protoxin II.

Stanislav Sokolov; Richard L. Kraus; Todd Scheuer; William A. Catterall

ProTx-II, an inhibitory cysteine knot toxin from the tarantula Thrixopelma pruriens, inhibits voltage-gated sodium channels. Using the cut-open oocyte preparation for electrophysiological recording, we show here that ProTx-II impedes movement of the gating charges of the sodium channel voltage sensors and reduces maximum activation of sodium conductance. At a concentration of 1 μM, the toxin inhibits 65.3 ± 4.1% of the sodium conductance and 24.6 ± 6.8% of the gating current of brain Nav1.2a channels, with a specific effect on rapidly moving gating charge. Strong positive prepulses can reverse the inhibitory effect of ProTx-II, indicating voltage-dependent dissociation of the toxin. Voltage-dependent reversal of the ProTx-II effect is more rapid for cardiac Nav1.5 channels, suggesting subtype-specific action of this toxin. Voltage-dependent binding and block of gating current are hallmarks of gating modifier toxins, which act by binding to the extracellular end of the S4 voltage sensors of ion channels. The mutation L833C in the S3-S4 linker in domain II reduces affinity for ProTx-II, and mutation of the outermost two gating-charge-carrying arginine residues in the IIS4 voltage sensor to glutamine abolishes voltage-dependent reversal of toxin action and toxin block of gating current. Our results support a voltage-sensor-trapping model for ProTx-II action in which the bound toxin impedes the normal outward gating movement of the IIS4 transmembrane segment, traps the domain II voltage sensor module in its resting state, and thereby inhibits channel activation.


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.


ChemMedChem | 2009

Discovery of a potent, CNS-penetrant orexin receptor antagonist based on an n,n-disubstituted-1,4-diazepane scaffold that promotes sleep in rats.

David B. Whitman; Christopher D. Cox; Michael J. Breslin; Karen M. Brashear; John D. Schreier; Michael J. Bogusky; Rodney A. Bednar; Wei Lemaire; Joseph G. Bruno; George D. Hartman; Duane R. Reiss; C. Meacham Harrell; Richard L. Kraus; Yuxing Li; Susan L. Garson; Scott M. Doran; Thomayant Prueksaritanont; Chunze Li; Christopher J. Winrow; Kenneth S. Koblan; John J. Renger; Paul J. Coleman

Silent Night: Antagonism of the orexin (or hypocretin) system has recently been identified as a novel mechanism for the treatment of insomnia. Herein, we describe discovery of a dual (OX1R/OX2R) orexin receptor antagonist featuring a 1,4‐diazepane central constraint that blocks orexin signaling in vivo. In telemetry‐implanted rats, oral administration of this antagonist produced a decrease in wakefulness, while increasing REM and non‐REM sleep.


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.


ChemMedChem | 2012

Discovery of a Pharmacologically Active Antagonist of the Two-Pore-Domain Potassium Channel K2P9.1 (TASK-3)

Craig A. Coburn; Yunfu Luo; Mingxiang Cui; Jiabing Wang; Richard Soll; Jingchao Dong; Bin Hu; Michael A. Lyon; Vincent P. Santarelli; Richard L. Kraus; Yun Gregan; Yi. Wang; Steven V. Fox; Jacquelyn Binns; Scott M. Doran; Duane R. Reiss; Pamela L. Tannenbaum; Anthony L. Gotter; Peter T. Meinke; John J. Renger

TWIK‐related acid‐sensitive K+ (K2P9.1, TASK‐3) ion channels have the capacity to regulate the activity of neuronal pathways by influencing the resting membrane potential of neurons on which they are expressed. The central nervous system (CNS) expression of these channels suggests potential roles in neurologic disorders, and it is believed that the development of TASK‐3 antagonists could lead to the therapeutic treatment of a number of neurological conditions. While a therapeutic potential for TASK‐3 channel modulation exists, there are only a few documented examples of potent and selective small‐molecule channel blockers. Herein, we describe the discovery and lead optimization efforts for a novel series of TASK‐3 channel antagonists based on a 5,6,7,8‐tetrahydropyrido[4,3‐d]pyrimidine high‐throughput screening lead from which a subseries of potent and selective inhibitors were identified. One compound was profiled in detail with respect to its physical properties and demonstrated pharmacological target engagement as indicated by its ability to modulate sleep architecture in rodent electroencephalogram (EEG) telemetry models.

Collaboration


Dive into the Richard L. Kraus's collaboration.

Top Co-Authors

Avatar

Susan L. Garson

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Steven V. Fox

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge