Network


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

Hotspot


Dive into the research topics where Sara McKellip is active.

Publication


Featured researches published by Sara McKellip.


PLOS Pathogens | 2014

Discovery of a novel compound with anti-venezuelan equine encephalitis virus activity that targets the nonstructural protein 2.

Dong-Hoon Chung; Colleen B. Jonsson; Nichole A. Tower; Yong Kyu Chu; Ergin Sahin; Jennifer E. Golden; James W. Noah; Chad E. Schroeder; Julie Sotsky; Melinda Sosa; Daniel E. Cramer; Sara McKellip; Lynn Rasmussen; E. Lucile White; Connie S. Schmaljohn; Justin G. Julander; Jeffrey M. Smith; Claire Marie Filone; John H. Connor; Yasuteru Sakurai; Robert A. Davey

Alphaviruses present serious health threats as emerging and re-emerging viruses. Venezuelan equine encephalitis virus (VEEV), a New World alphavirus, can cause encephalitis in humans and horses, but there are no therapeutics for treatment. To date, compounds reported as anti-VEEV or anti-alphavirus inhibitors have shown moderate activity. To discover new classes of anti-VEEV inhibitors with novel viral targets, we used a high-throughput screen based on the measurement of cell protection from live VEEV TC-83-induced cytopathic effect to screen a 340,000 compound library. Of those, we identified five novel anti-VEEV compounds and chose a quinazolinone compound, CID15997213 (IC50 = 0.84 µM), for further characterization. The antiviral effect of CID15997213 was alphavirus-specific, inhibiting VEEV and Western equine encephalitis virus, but not Eastern equine encephalitis virus. In vitro assays confirmed inhibition of viral RNA, protein, and progeny synthesis. No antiviral activity was detected against a select group of RNA viruses. We found mutations conferring the resistance to the compound in the N-terminal domain of nsP2 and confirmed the target residues using a reverse genetic approach. Time of addition studies showed that the compound inhibits the middle stage of replication when viral genome replication is most active. In mice, the compound showed complete protection from lethal VEEV disease at 50 mg/kg/day. Collectively, these results reveal a potent anti-VEEV compound that uniquely targets the viral nsP2 N-terminal domain. While the function of nsP2 has yet to be characterized, our studies suggest that the protein might play a critical role in viral replication, and further, may represent an innovative opportunity to develop therapeutic interventions for alphavirus infection.


Journal of Biomolecular Screening | 2012

High-throughput screening identifies a bisphenol inhibitor of SV40 large T antigen ATPase activity.

Sandlin P. Seguin; Carrie W. Evans; Miranda Nebane-Akah; Sara McKellip; Subramaniam Ananthan; Nichole A. Tower; Melinda Sosa; Lynn Rasmussen; E. Lucile White; Brooks E. Maki; Daljit S. Matharu; Jennifer E. Golden; Jeffrey Aubé; Jeffrey L. Brodsky; James W. Noah

The authors conducted a high-throughput screening campaign for inhibitors of SV40 large T antigen ATPase activity to identify candidate antivirals that target the replication of polyomaviruses. The primary assay was adapted to 1536-well microplates and used to screen the National Institutes of Health Molecular Libraries Probe Centers Network library of 306 015 compounds. The primary screen had an Z value of ~0.68, signal/background = 3, and a high (5%) DMSO tolerance. Two counterscreens and two secondary assays were used to prioritize hits by EC50, cytotoxicity, target specificity, and off-target effects. Hits that inhibited ATPase activity by >44% in the primary screen were tested in dose–response efficacy and eukaryotic cytotoxicity assays. After evaluation of hit cytotoxicity, drug likeness, promiscuity, and target specificity, three compounds were chosen for chemical optimization. Chemical optimization identified a class of bisphenols as the most effective biochemical inhibitors. Bisphenol A inhibited SV40 large T antigen ATPase activity with an IC50 of 41 µM in the primary assay and 6.2 µM in a cytoprotection assay. This compound class is suitable as probes for biochemical investigation of large T antigen ATPase activity, but because of their cytotoxicity, further optimization is necessary for their use in studying polyomavirus replication in vivo.


Journal of Biomolecular Screening | 2012

Primary and Secondary Drug Screening Assays for Friedreich Ataxia

M. Grazia Cotticelli; Lynn Rasmussen; Nicole L. Kushner; Sara McKellip; Melinda Sosa; Anna Manouvakhova; Shuang Feng; E. Lucile White; Joseph A. Maddry; Jill Heemskerk; Robert J. Oldt; Lea F. Surrey; Rachel Ochs; Robert B. Wilson

Friedreich ataxia (FRDA) is an autosomal recessive neuro- and cardiodegenerative disorder for which there are no proven effective treatments. FRDA is caused by decreased expression and/or function of the protein frataxin. Frataxin chaperones iron in the mitochondrial matrix for the assembly of iron–sulfur clusters (ISCs), which are prosthetic groups critical for the function of the Krebs cycle and the mitochondrial electron transport chain (ETC). Decreased expression of frataxin or the yeast frataxin orthologue, Yfh1p, is associated with decreased ISC assembly, mitochondrial iron accumulation, and increased oxidative stress, all of which contribute to mitochondrial dysfunction. Using yeast depleted of Yfh1p, a high-throughput screening (HTS) assay was developed in which mitochondrial function was monitored by reduction of the tetrazolium dye WST-1 in a growth medium with a respiration-only carbon source. Of 101 200 compounds screened, 302 were identified that effectively rescue mitochondrial function. To confirm activities in mammalian cells and begin understanding mechanisms of action, secondary screening assays were developed using murine C2C12 cells and yeast mutants lacking specific complexes of the ETC, respectively. The compounds identified in this study have potential relevance for other neurodegenerative disorders associated with mitochondrial dysfunction, such as Parkinson disease.


Journal of Neuroscience Research | 2011

Identification of novel small molecule activators of nuclear factor-κb with neuroprotective action via high-throughput screening

Marina Manuvakhova; Guyla G. Johnson; Misti C. White; Subramaniam Ananthan; Melinda Sosa; Clinton Maddox; Sara McKellip; Lynn Rasmussen; Krister Wennerberg; Judith V. Hobrath; E. Lucile White; Joseph A. Maddry; Maurizio Grimaldi

Neuronal noncytokine‐dependent p50/p65 nuclear factor‐κB (the primary NF‐κB complex in the brain) activation has been shown to exert neuroprotective actions. Thus neuronal activation of NF‐κB could represent a viable neuroprotective target. We have developed a cell‐based assay able to detect NF‐κB expression enhancement, and through its use we have identified small molecules able to up‐regulate NF‐κB expression and hence trigger its activation in neurons. We have successfully screened approximately 300,000 compounds and identified 1,647 active compounds. Cluster analysis of the structures within the hit population yielded 14 enriched chemical scaffolds. One high‐potency and chemically attractive representative of each of these 14 scaffolds and four singleton structures were selected for follow‐up. The experiments described here highlighted that seven compounds caused noncanonical long‐lasting NF‐κB activation in primary astrocytes. Molecular NF‐κB docking experiments indicate that compounds could be modulating NF‐κB‐induced NF‐κB expression via enhancement of NF‐κB binding to its own promoter. Prototype compounds increased p65 expression in neurons and caused its nuclear translocation without affecting the inhibitor of NF‐κB (I‐κB). One of the prototypical compounds caused a large reduction of glutamate‐induced neuronal death. In conclusion, we have provided evidence that we can use small molecules to activate p65 NF‐κB expression in neurons in a cytokine receptor‐independent manner, which results in both long‐lasting p65 NF‐κB translocation/activation and decreased glutamate neurotoxicity.


Journal of Medicinal Chemistry | 2014

Development of (E)-2-((1,4-dimethylpiperazin-2-ylidene)amino)-5-nitro-N-phenylbenzamide, ML336: Novel 2-amidinophenylbenzamides as potent inhibitors of venezuelan equine encephalitis virus.

Chad E. Schroeder; Tuanli Yao; Julie Sotsky; Robert A. Smith; Sudeshna Roy; Yong Kyu Chu; Haixun Guo; Nichole A. Tower; James W. Noah; Sara McKellip; Melinda Sosa; Lynn Rasmussen; Layton H. Smith; E. Lucile White; Jeffrey Aubé; Colleen B. Jonsson; Dong-Hoon Chung; Jennifer E. Golden

Venezuelan equine encephalitis virus (VEEV) is an emerging pathogenic alphavirus that can cause significant disease in humans. Given the absence of therapeutic options available and the significance of VEEV as a weaponized agent, an optimization effort was initiated around a quinazolinone screening hit 1 with promising cellular antiviral activity (EC50 = 0.8 μM), limited cytotoxic liability (CC50 > 50 μM), and modest in vitro efficacy in reducing viral progeny (63-fold at 5 μM). Scaffold optimization revealed a novel rearrangement affording amidines, specifically compound 45, which was found to potently inhibit several VEEV strains in the low nanomolar range without cytotoxicity (EC50 = 0.02–0.04 μM, CC50 > 50 μM) while limiting in vitro viral replication (EC90 = 0.17 μM). Brain exposure was observed in mice with 45. Significant protection was observed in VEEV-infected mice at 5 mg kg–1 day–1 and viral replication appeared to be inhibited through interference of viral nonstructural proteins.


Molecules | 2010

HTS-Driven Discovery of New Chemotypes with West Nile Virus Inhibitory Activity

Dong-Hoon Chung; Colleen B. Jonsson; Clinton Maddox; Sara McKellip; Blak E.P. Moore; Marintha Heil; E. Lucile White; Subramaniam Ananthan; Qianjun Li; Shuang Feng; Lynn Rasmussen

West Nile virus (WNV) is a positive sense, single-stranded RNA virus that can cause illness in humans when transmitted via mosquito vectors. Unfortunately, no antivirals or vaccines are currently available, and therefore efficient and safe antivirals are urgently needed. We developed a high throughput screen to discover small molecule probes that inhibit virus infection of Vero E6 cells. A primary screen of a 13,001 compound library at a 10 μM final concentration was conducted using the 384-well format. Z′ values ranged from 0.54–0.83 with a median of 0.74. Average S/B was 17 and S/N for each plate ranged from 10.8 to 23.9. Twenty-six compounds showed a dose response in the HT screen and were further evaluated in a time of addition assay and in a titer reduction assay. Seven compounds showed potential as small molecule probes directed at WNV. The hit rate from the primary screen was 0.185% (24 compounds out of 13,001 compounds) and from the secondary screens was 0.053% (7 out of 13,001 compounds) respectively.


ChemBioChem | 2010

A High‐Throughput Screen for Chemical Inhibitors of Exocytic Transport in Yeast

Lisha Zhang; N. Miranda Nebane; Krister Wennerberg; Yujie Li; Valerie Neubauer; Judith V. Hobrath; Sara McKellip; Lynn Rasmussen; Nice Shindo; Melinda Sosa; Joseph A. Maddry; Subramaniam Ananthan; Gary A. Piazza; E. Lucile White; Edina Harsay

Most of the components of the membrane and protein traffic machinery were discovered by perturbing their functions, either with bioactive compounds or by mutations. However, the mechanisms responsible for exocytic transport vesicle formation at the Golgi and endosomes are still largely unknown. Both the exocytic traffic routes and the signaling pathways that regulate these routes are highly complex and robust, so that defects can be overcome by alternate pathways or mechanisms. A classical yeast genetic screen designed to account for the robustness of the exocytic pathway identified a novel conserved gene, AVL9, which functions in late exocytic transport. We now describe a chemical‐genetic version of the mutant screen, in which we performed a high‐throughput phenotypic screen of a large compound library and identified novel small‐molecule secretory inhibitors. To maximize the number and diversity of our hits, the screen was performed in a pdr5Δ snq2Δ mutant background, which lacks two transporters responsible for pleiotropic drug resistance. However, we found that deletion of both transporters reduced the fitness of our screen strain, whereas the pdr5Δ mutation had a relatively small effect on growth and was also the more important transporter mutation for conferring sensitivity to our hits. In this and similar chemical‐genetic yeast screens, using just a single pump mutation might be sufficient for increasing hit diversity while minimizing the physiological effects of transporter mutations.


Journal of Laboratory Automation | 2013

High-throughput RNA interference screening: tricks of the trade.

N. Miranda Nebane; Tatjana Coric; Kanupriya Whig; Sara McKellip; LaKeisha Woods; Melinda Sosa; Russell Sheppard; Lynn Rasmussen; Mary-Ann Bjornsti; E. Lucile White

The process of validating an assay for high-throughput screening (HTS) involves identifying sources of variability and developing procedures that minimize the variability at each step in the protocol. The goal is to produce a robust and reproducible assay with good metrics. In all good cell-based assays, this means coefficient of variation (CV) values of less than 10% and a signal window of fivefold or greater. HTS assays are usually evaluated using Z′ factor, which incorporates both standard deviation and signal window. A Z′ factor value of 0.5 or higher is acceptable for HTS. We used a standard HTS validation procedure in developing small interfering RNA (siRNA) screening technology at the HTS center at Southern Research. Initially, our assay performance was similar to published screens, with CV values greater than 10% and Z′ factor values of 0.51 ± 0.16 (average ± standard deviation). After optimizing the siRNA assay, we got CV values averaging 7.2% and a robust Z′ factor value of 0.78 ± 0.06 (average ± standard deviation). We present an overview of the problems encountered in developing this whole-genome siRNA screening program at Southern Research and how equipment optimization led to improved data quality.


Journal of Laboratory Automation | 2016

Acoustic Droplet Ejection Technology and Its Application in High-Throughput RNA Interference Screening

N. Miranda Nebane; Tatjana Coric; Sara McKellip; LaKeisha Woods; Melinda Sosa; Lynn Rasmussen; Mary-Ann Bjornsti; E. Lucile White

The development of acoustic droplet ejection (ADE) technology has resulted in many positive changes associated with the operations in a high-throughput screening (HTS) laboratory. Originally, this liquid transfer technology was used to simply transfer DMSO solutions of primarily compounds. With the introduction of Labcyte’s Echo 555, which has aqueous dispense capability, the application of this technology has been expanded beyond its original use. This includes the transfer of many biological reagents solubilized in aqueous buffers, including siRNAs. The Echo 555 is ideal for siRNA dispensing because it is accurate at low volumes and a step-down dilution is not necessary. The potential for liquid carryover and cross-contamination is eliminated, as no tips are needed. Herein, we describe the siRNA screening platform at Southern Research’s HTS Center using the ADE technology. With this technology, an siRNA library can be dispensed weeks or even months in advance of the assay itself. The protocol has been optimized to achieve assay parameters comparable to small-molecule screening parameters, and exceeding the norm reported for genomewide siRNA screens.


Cancer Research | 2016

Abstract LB-055: High-throughput screening efforts for the identification of selective and potent inhibitors of CD38 for the treatment of hematological cancers

Davide Botta; Tulin Dadali; Betty J. Mousseau; Fen Zhou; Michael Schultz; Esther Zumaquero; Anna Manouvakhova; Melinda Sosa; Sara McKellip; LaKeisha Woods; Nichole A. Tower; Larry J. Ross; Lynn Rasmussen; E. Lucille White; Indira Padmalayam; Wei Zhang; Maaike Everts; Corinne E. Augelli-Szafran; James R. Bostwick; Mark J. Suto; Frances E. Lund

By means of a phenomenon termed “the Warburg effect,” tumor cells shift their energy production by mitochondrial oxidative phosphorylation to aerobic glycolysis, resulting in the upregulation of glucose consumption and increased cellular oxidative and nitrosative stress. To compensate for such toxic levels of ROS/RNS, cancer cells rely heavily on their antioxidant defense mechanisms, which are largely controlled by the NAD(P)/NAD(P)H redox partners. We found that the modulation of NAD metabolism in vivo, specifically via the deletion of the NAD glycohydrolase CD38, resulted in increased production of intrinsic ROS and increased DNA damage following exposure to chemotherapeutics. Furthermore, in vitro experiments showed that CD38 knockdown in CD38-expressing tumor cells prevented the generation of stable transfectants, highlighting a role for CD38 in tumor cell survival. In light of these findings, we hypothesized that pharmacological inhibition of CD38 may be an effective therapy for the treatment of hematological cancers, in particular those which uniformly overexpress CD38, such as MM and chronic lymphocytic leukemia. Indeed, treatment of human MM cell lines LP-1 and KMS-12-PE with CD38 antagonists sensitized the cells to standard ROS-inducing chemotherapeutics. We conducted a high-throughput screening (HTS) campaign of over two hundred thousand unique and non-proprietary lead-like compounds using an optimized and miniaturized HTS based on a luminescent NAD quantitation platform. Five hundred active hits were analyzed for toxicity using a cell-based HTS assay purposely designed with CD38-negative HEK293 cells to avoid elimination of desirable compounds toxic to CD38-positive cells. Hits with non-specific properties, such as PAINS (Pan Assay Interference Compounds), were removed by computational filtering, and the remaining compounds were tested for inhibition of human CD38 activity in cells. The last phase of the compound progression pathway involved testing for non-selective inhibition of other NAD-consuming enzymes, namely Poly(ADP-ribose) polymerase-1, and Sirtuin-1, which led to the identification of two distinct chemical series that exhibit >10-fold selectivity for human CD38. Hit-to-lead chemistry is currently underway to synthesize key analogs by rational drug design. In summary, our data suggests that CD38 is an antioxidant protein selectively used to maintain a cellular redox balance, and proposes that targeting the enzymatic activity of CD38 may be a novel therapeutic strategy for chemosensitizing hematological cancers. Our HTS campaign efforts are paving the way for the discovery and development of potent and selective small-molecule inhibitors of CD38. Citation Format: Davide Botta, Tulin Dadali, Betty J. Mousseau, Fen Zhou, Michael D. Schultz, Esther Zumaquero, Anna Manouvakhova, Melinda I. Sosa, Sara N. McKellip, LaKeisha Woods, Nichole A. Tower, Larry J. Ross, Lynn Rasmussen, E. Lucille White, Indira Padmalayam, Wei Zhang, Maaike Everts, Corinne E. Augelli-Szafran, James R. Bostwick, Mark J. Suto, Frances E. Lund. High-throughput screening efforts for the identification of selective and potent inhibitors of CD38 for the treatment of hematological cancers. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr LB-055.

Collaboration


Dive into the Sara McKellip's collaboration.

Top Co-Authors

Avatar

Lynn Rasmussen

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar

E. Lucile White

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar

Melinda Sosa

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar

Nichole A. Tower

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar

Clinton Maddox

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James W. Noah

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donghoon Chung

Southern Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge