Network


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

Hotspot


Dive into the research topics where Sandra Nelson is active.

Publication


Featured researches published by Sandra Nelson.


Molecular Pharmacology | 2013

Hits of a High-Throughput Screen Identify the Hydrophobic Pocket of Autotaxin/Lysophospholipase D As an Inhibitory Surface

James I. Fells; Sue Chin Lee; Yuko Fujiwara; Derek D. Norman; Keng Gat Lim; Ryoko Tsukahara; Jianxiong Liu; Renukadevi Patil; Duane D. Miller; R. Jason Kirby; Sandra Nelson; William Seibel; Ruben Papoian; Daniel L. Baker; Robert Bittman; Gabor Tigyi

Autotaxin (ATX), a lysophospholipase D, plays an important role in cancer invasion, metastasis, tumor progression, tumorigenesis, neuropathic pain, fibrotic diseases, cholestatic pruritus, lymphocyte homing, and thrombotic diseases by producing the lipid mediator lysophosphatidic acid (LPA). A high-throughput screen of ATX inhibition using the lysophosphatidylcholine-like substrate fluorogenic substrate 3 (FS-3) and ∼10,000 compounds from the University of Cincinnati Drug Discovery Center identified several small-molecule inhibitors with IC50 vales ranging from nanomolar to low micromolar. The pharmacology of the three most potent compounds: 918013 (1; 2,4-dichloro-N-(3-fluorophenyl)-5-(4-morpholinylsulfonyl) benzamide), 931126 (2; 4-oxo-4-{2-[(5-phenoxy-1H-indol-2-yl)carbonyl]hydrazino}-N-(4-phenylbutan-2-yl)butanamide), and 966791 (3; N-(2,6-dimethylphenyl)-2-[N-(2-furylmethyl)(4-(1,2,3,4-tetraazolyl)phenyl)carbonylamino]-2-(4-hydroxy-3-methoxyphenyl) acetamide), were further characterized in enzyme, cellular, and whole animal models. Compounds 1 and 2 were competitive inhibitors of ATX-mediated hydrolysis of the lysophospholipase substrate FS-3. In contrast, compound 3 was a competitive inhibitor of both FS-3 and the phosphodiesterase substrate p-nitrophenyl thymidine 5′-monophosphate. Computational docking and mutagenesis suggested that compounds 1 and 2 target the hydrophobic pocket, thereby blocking access to the active site of ATX. The potencies of compounds 1–3 were comparable to each other in each of the assays. All of these compounds significantly reduced invasion of A2058 human melanoma cells in vitro and the colonization of lung metastases by B16-F10 murine melanoma cells in C57BL/6 mice. The compounds had no agonist or antagonist effects on select LPA or sphingosine 1-phosphate receptors, nor did they inhibit nucleotide pyrophosphatase/phosphodiesterase (NPP) enzymes NPP6 and NPP7. These results identify the molecular surface of the hydrophobic pocket of ATX as a target-binding site for inhibitors of enzymatic activity.


PLOS ONE | 2013

Inhibition of the Growth Factor MDK/Midkine by a Novel Small Molecule Compound to Treat Non-Small Cell Lung Cancer

Huifang Hao; Yutaka Maeda; Takuya Fukazawa; Tomoki Yamatsuji; Munenori Takaoka; Xiao Hong Bao; Junji Matsuoka; Tatsuo Okui; Tsuyoshi Shimo; Nagio Takigawa; Yasuko Tomono; Motowo Nakajima; Iris M. Fink-Baldauf; Sandra Nelson; William Seibel; Ruben Papoian; Jeffrey A. Whitsett; Yoshio Naomoto

Midkine (MDK) is a heparin-binding growth factor that is highly expressed in many malignant tumors, including lung cancers. MDK activates the PI3K pathway and induces anti-apoptotic activity, in turn enhancing the survival of tumors. Therefore, the inhibition of MDK is considered a potential strategy for cancer therapy. In the present study, we demonstrate a novel small molecule compound (iMDK) that targets MDK. iMDK inhibited the cell growth of MDK-positive H441 lung adenocarcinoma cells that harbor an oncogenic KRAS mutation and H520 squamous cell lung cancer cells, both of which are types of untreatable lung cancer. However, iMDK did not reduce the cell viability of MDK-negative A549 lung adenocarcinoma cells or normal human lung fibroblast (NHLF) cells indicating its specificity. iMDK suppressed the endogenous expression of MDK but not that of other growth factors such as PTN or VEGF. iMDK suppressed the growth of H441 cells by inhibiting the PI3K pathway and inducing apoptosis. Systemic administration of iMDK significantly inhibited tumor growth in a xenograft mouse model in vivo. Inhibition of MDK with iMDK provides a potential therapeutic approach for the treatment of lung cancers that are driven by MDK.


FEBS Journal | 2014

Targeting the hydrophobic pocket of autotaxin with virtual screening of inhibitors identifies a common aromatic sulfonamide structural motif.

James I. Fells; Sue Chin Lee; Derek D. Norman; Ryoko Tsukahara; Jason Kirby; Sandra Nelson; William Seibel; Ruben Papoian; Renukadevi Patil; Duane D. Miller; Truc Chi T. Pham; Daniel L. Baker; Robert Bittman; Gabor Tigyi

Modulation of autotaxin (ATX), the lysophospholipase D enzyme that produces lysophosphatidic acid, with small‐molecule inhibitors is a promising strategy for blocking the ATX–lysophosphatidic acid signaling axis. Although discovery campaigns have been successful in identifying ATX inhibitors, many of the reported inhibitors target the catalytic cleft of ATX. A recent study provided evidence for an additional inhibitory surface in the hydrophobic binding pocket of ATX, confirming prior studies that relied on enzyme kinetics and differential inhibition of substrates varying in size. Multiple hits from previous high‐throughput screening for ATX inhibitors were obtained with aromatic sulfonamide derivatives interacting with the hydrophobic pocket. Here, we describe the development of a ligand‐based strategy and its application in virtual screening, which yielded novel high‐potency inhibitors that target the hydrophobic pocket of ATX. Characterization of the structure–activity relationship of these new inhibitors forms the foundation of a new pharmacophore model of the hydrophobic pocket of ATX.


Journal of Biological Chemistry | 2015

Combined Rational Design and a High-Throughput Screening Platform for Identifying Chemical Inhibitors of a Ras Activating Enzyme

Chris R. Evelyn; Jacek Biesiada; Xin Duan; Hong Tang; Xun Shang; Ruben Papoian; William Seibel; Sandra Nelson; Jaroslaw Meller; Yi Zheng

Background: GEFs can serve as targets in Ras family GTPase activity signaling. Results: A virtual screening coupled with the experimental screening platform is established targeting a Ras GEF enzyme. Conclusion: The multiple tier screening platform is useful for lead discovery targeting GEFs of Ras-like small GTPases. Significance: This study provides a unique screening approach for inhibitors of Ras and Ras-like GTPase activities. The Ras family small GTPases regulate multiple cellular processes, including cell growth, survival, movement, and gene expression, and are intimately involved in cancer pathogenesis. Activation of these small GTPases is catalyzed by a special class of enzymes, termed guanine nucleotide exchange factors (GEFs). Herein, we developed a small molecule screening platform for identifying lead hits targeting a Ras GEF enzyme, SOS1. We employed an ensemble structure-based virtual screening approach in combination with a multiple tier high throughput experimental screen utilizing two complementary fluorescent guanine nucleotide exchange assays to identify small molecule inhibitors of GEF catalytic activity toward Ras. From a library of 350,000 compounds, we selected a set of 418 candidate compounds predicted to disrupt the GEF-Ras interaction, of which dual wavelength GDP dissociation and GTP-loading experimental screening identified two chemically distinct small molecule inhibitors. Subsequent biochemical validations indicate that they are capable of dose-dependently inhibiting GEF catalytic activity, binding to SOS1 with micromolar affinity, and disrupting GEF-Ras interaction. Mutagenesis studies in conjunction with structure-activity relationship studies mapped both compounds to different sites in the catalytic pocket, and both inhibited Ras signaling in cells. The unique screening platform established here for targeting Ras GEF enzymes could be broadly useful for identifying lead inhibitors for a variety of small GTPase-activating GEF reactions.


Journal of Biomolecular Screening | 2003

Development and use of a high-throughput bacterial DNA gyrase assay to identify mammalian topoisomerase II inhibitors with whole-cell anticancer activity.

Siddhartha Roychoudhury; Kelly M. Makin; Tracy L. Twinem; David T. Stanton; Sandra Nelson; Carl E. Catrenich

A high-throughput screen (HTS)was developed and used to identify inhibitors of bacterial DNA gyrase. Among the validated hits were 53 compounds that also inhibited mammalian topoisomerase II with IC50 values of <12.5 µg/mL for 51 of them. Using computational methods, these compounds were subjected to cluster analysis to categorize them according to their chemical and structural properties. Nine compounds from different clusters were tested for their whole-cell inhibitory activity against 3 cancer cell lines—NCI-H460 (lung), MCF7 (breast), and SF-268 (CNS)—at a concentration of 100 µM. Five compounds inhibited cell growth by >50% for all 3 cell lines tested. These compounds were tested further against a panel of 53 to 57 cell lines representing leukemia, melanoma, colon, CNS, ovarian, renal, prostate, breast, and non–small cell lung cancers. In this assay, PGE-7143417 was found to be the most potent compound, which inhibited the growth of all the cell lines by 50% at a concentration range of 0.31 to 2.58 µM, with an average of 1.21 µM. An additional 17 compounds were also tested separately against a panel of 10 cell lines representing melanoma, colon, lung, mammary, ovarian, prostate, and renal cancers. In this assay, 4 compounds—PGE-3782569, PGE-7411516, PGE-2908955, and PGE-3521917—were found to have activity with concentrations for 50% cell growth inhibition in the 0.59 to 3.33, 22.5 to 59.1, 7.1 to >100, and 24.7 to >100 µM range. (Journal of Biomolecular Screening 2003:157-163)


Journal of Biomolecular Screening | 2013

Development of a Small-Molecule Screening Method for Inhibitors of Cellular Response to Myostatin and Activin A

Jennifer N. Cash; Elizabeth B. Angerman; R. Jason Kirby; Lisa Merck; William Seibel; Matthew Wortman; Ruben Papoian; Sandra Nelson; Thomas B. Thompson

Myostatin, a member of the transforming growth factor (TGF)-β family of secreted ligands, is a strong negative regulator of muscle growth. As such, therapeutic inhibitors of myostatin are actively being investigated for their potential in the treatment of muscle-wasting diseases such as muscular dystrophy and sarcopenia. Here, we sought to develop a high-throughput screening (HTS) method for small-molecule inhibitors that target myostatin. We created a HEK293 stable cell line that expresses the (CAGA)12-luciferase reporter construct and robustly responds to signaling of certain classes of TGF-β family ligands. After optimization and miniaturization of the assay to a 384-well format, we successfully screened a library of compounds for inhibition of myostatin and the closely related activin A. Selection of some of the tested compounds was directed by in silico screening against myostatin, which led to an enrichment of target hits as compared with random selection. Altogether, we present an HTS method that will be useful for screening potential inhibitors of not only myostatin but also many other ligands of the TGF-β family.


Journal of Biomolecular Screening | 2013

High-Throughput Screening for Small-Molecule Inhibitors of Staphylococcus epidermidis RP62a Biofilms

Warunya Panmanee; Deborah Taylor; Chloe Ja Shea; Hong Tang; Sandra Nelson; William Seibel; Ruben Papoian; Ryan M. Kramer; Daniel J. Hassett; Thomas J. Lamkin

High-throughput screening (HTS) of 42 865 compounds was performed to identify compounds that inhibit formation of or kill Staphylococcus epidermidis RP62a biofilms. Three biological processes were assayed, including (1) growth of planktonic/biofilm bacteria, (2) assessment of metabolically active biofilm bacteria using a resazurin assay, and (3) assessment of biofilm biomass by crystal violet staining. After completing the three tiers (primary screening, hit confirmation, and dose-response curves), 352 compounds (representing ~0.8%) were selected as confirmed hit compounds from the HTS assay. The compounds were divided into groups based on their effectiveness on S. epidermidis biofilm properties. The majority of these affected both inhibition and killing of bacterial biofilm cultures. Only 16 of the confirmed hit compounds that have either an AC50 lower than 10 µM and/or Sconst ≥70 from those processed were selected for further study by confocal laser scanning microscopy (CLSM). The CLSM was used to evaluate the confirmed hit compounds on (1) inhibition of biofilm formation and (2) killing of preexisting S. epidermidis biofilms. Taken together, with further testing (e.g., disease-related conditions), such compounds may have applications as broad antimicrobial/antibiofilm use for prophylactic or therapeutic intervention to combat infections in surgical and intensive care clinics and battlefield settings.


Journal of Biomolecular Screening | 2012

Automated Analysis and Classification of Infected Macrophages Using Bright-Field Amplitude Contrast Data

Umesh Adiga; Debbie Taylor; Brian L. Bell; Larissa Ponomareva; Stephen Kanzlemar; Ryan Kramer; Roland Saldanha; Sandra Nelson; Thomas J. Lamkin

This article presents a methodology for acquisition and analysis of bright-field amplitude contrast image data in high-throughput screening (HTS) for the measurement of cell density, cell viability, and classification of individual cells into phenotypic classes. We present a robust image analysis pipeline, where the original data are subjected to image standardization, image enhancement, and segmentation by region growing. This work develops new imaging and analysis techniques for cell analysis in HTS and successfully addresses a particular need for direct measurement of cell density and other features without using dyes.


Molecular Cancer Therapeutics | 2011

Discovery of a Small Molecule Targeting IRA2 Deletion in Budding Yeast and Neurofibromin Loss in Malignant Peripheral Nerve Sheath Tumor Cells

Matthew D. Wood; Melissa Rawe; Gunnar Johansson; Shu Pang; Ryan S. Soderquist; Ami V. Patel; Sandra Nelson; William Seibel; Nancy Ratner; Yolanda Sanchez

Malignant peripheral nerve sheath tumor (MPNST) is a life-threatening complication of neurofibromatosis type 1 (NF1). NF1 is caused by mutation in the gene encoding neurofibromin, a negative regulator of Ras signaling. There are no effective pharmacologic therapies for MPNST. To identify new therapeutic approaches targeting this dangerous malignancy, we developed assays in NF1+/+ and NF1−/− MPNST cell lines and in budding yeast lacking the NF1 homologue IRA2 (ira2Δ). Here, we describe UC1, a small molecule that targets NF1−/− cell lines and ira2Δ budding yeast. By using yeast genetics, we identified NAB3 as a high-copy suppressor of UC1 sensitivity. NAB3 encodes an RNA binding protein that associates with the C-terminal domain of RNA Pol II and plays a role in the termination of nonpolyadenylated RNA transcripts. Strains with deletion of IRA2 are sensitive to genetic inactivation of NAB3, suggesting an interaction between Ras signaling and Nab3-dependent transcript termination. This work identifies a lead compound and a possible target pathway for NF1-associated MPNST, and shows a novel model system approach to identify and validate target pathways for cancer cells in which NF1 loss drives tumor formation. Mol Cancer Ther; 10(9); 1740–50. ©2011 AACR.


Drug Discovery Today: Technologies | 2008

Academic HTS: diverse portraits

Sandra Nelson

High-throughput screening (HTS) centers have been established at many universities over the past five years. The characteristics of academic HTS centers vary widely. They generally have smaller compound collections, less automation and less software than their industrial counterparts. Academic HTS centers have had success in finding biological probes, and many of their contributions are documented in the scientific literature.

Collaboration


Dive into the Sandra Nelson's collaboration.

Top Co-Authors

Avatar

William Seibel

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Ruben Papoian

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Derek D. Norman

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Gabor Tigyi

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Hong Tang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

James I. Fells

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Ryoko Tsukahara

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Sue Chin Lee

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Thomas J. Lamkin

Wright-Patterson Air Force Base

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge