Network


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

Hotspot


Dive into the research topics where Suzanne G. Mays is active.

Publication


Featured researches published by Suzanne G. Mays.


Molecular Cancer Therapeutics | 2011

Enigmol: A Novel Sphingolipid Analogue with Anticancer Activity against Cancer Cell Lines and In vivo Models for Intestinal and Prostate Cancer

Holly Symolon; Anatoliy S. Bushnev; Qiong Peng; Harsha Ramaraju; Suzanne G. Mays; Jeremy C. Allegood; Sarah T. Pruett; M. Cameron Sullards; Dirck L. Dillehay; Dennis C. Liotta; Alfred H. Merrill

Sphingoid bases are cytotoxic for many cancer cell lines and are thought to contribute to suppression of intestinal tumorigenesis in vivo by ingested sphingolipids. This study explored the behavior of a sphingoid base analogue, (2S,3S,5S)-2-amino-3,5-dihydroxyoctadecane (Enigmol), that cannot be phosphorylated by sphingosine kinases and is slowly N-acylated and therefore is more persistent than natural sphingoid bases. Enigmol had potential anticancer activity in a National Cancer Institute (NCI-60) cell line screen and was confirmed to be more cytotoxic and persistent than naturally occurring sphingoid bases using HT29 cells, a colon cancer cell line. Although the molecular targets of sphingoid bases are not well delineated, Enigmol shared one of the mechanisms that has been found for naturally occurring sphingoid bases: normalization of the aberrant accumulation of β-catenin in the nucleus and cytoplasm of colon cancer cells due to defect(s) in the adenomatous polyposis coli (APC)/β-catenin regulatory system. Enigmol also had antitumor efficacy when administered orally to Min mice, a mouse model with a truncated APC gene product (C57Bl/6JMin/+ mice), decreasing the number of intestinal tumors by half at 0.025% of the diet (w/w), with no evidence of host toxicity until higher dosages. Enigmol was also tested against the prostate cancer cell lines DU145 and PC-3 in nude mouse xenografts and suppressed tumor growth in both. Thus, Enigmol represents a novel category of sphingoid base analogue that is orally bioavailable and has the potential to be effective against multiple types of cancer. Mol Cancer Ther; 10(4); 648–57. ©2011 AACR.


ACS Medicinal Chemistry Letters | 2011

Novel Synthesis and Biological Evaluation of Enigmols as Therapeutic Agents for Treating Prostate Cancer

Ethel C. Garnier-Amblard; Suzanne G. Mays; Richard F. Arrendale; Mark T. Baillie; Anatoliy S. Bushnev; Deborah G. Culver; Taylor J. Evers; Jason J. Holt; Randy B. Howard; Lanny S. Liebeskind; David S. Menaldino; Michael G. Natchus; John A. Petros; Harsha Ramaraju; G. Prabhakar Reddy; Dennis C. Liotta

Enigmol is a synthetic, orally active 1-deoxysphingoid base analogue that has demonstrated promising activity against prostate cancer. In these studies, the pharmacologic roles of stereochemistry and N-methylation in the structure of enigmols were examined. A novel enantioselective synthesis of all four possible 2S-diastereoisomers of enigmol (2-aminooctadecane-3,5-diols) from l-alanine is reported, which features a Liebeskind-Srogl cross-coupling reaction between l-alanine thiol ester and (E)-pentadec-1-enylboronic acid as the key step. In vitro biological evaluation of the four enigmol diastereoisomers and 2S,3S,5S-N-methylenigmol against two prostate cancer cell lines (PC-3 and LNCaP) indicates that all but one diastereomer demonstrate potent oncolytic activity. In nude mouse xenograft models of human prostate cancer, enigmol was equally effective as standard prostate cancer therapies (androgen deprivation or docetaxel), and two of the enigmol diastereomers, 2S,3S,5R-enigmol and 2S,3R,5S-enigmol, also caused statistically significant inhibition of tumor growth. A pharmacokinetic profile of enigmol and N-methylenigmol is also presented.


ACS Medicinal Chemistry Letters | 2016

Discovery of a Fluorinated Enigmol Analog with Enhanced in Vivo Pharmacokinetic and Anti-Tumor Properties

Eric Miller; Suzanne G. Mays; Mark T. Baillie; Randy B. Howard; Deborah G. Culver; Manohar Saindane; Sarah T. Pruett; Jason J. Holt; David S. Menaldino; Taylor J. Evers; G. Prabhakar Reddy; Richard F. Arrendale; Michael G. Natchus; John A. Petros; Dennis C. Liotta

The orally bioavailable 1-deoxy-sphingosine analog, Enigmol, has demonstrated anticancer activity in numerous in vivo settings. However, as no Enigmol analog with enhanced potency in vitro has been identified, a new strategy to improve efficacy in vivo by increasing tumor uptake was adopted. Herein, synthesis and biological evaluation of two novel fluorinated Enigmol analogs, CF3-Enigmol and CF2-Enigmol, are reported. Each analog was equipotent to Enigmol in vitro, but achieved higher plasma and tissue levels than Enigmol in vivo. Although plasma and tissue exposures were anticipated to trend with fluorine content, CF2-Enigmol absorbed into tissue at strikingly higher concentrations than CF3-Enigmol. Using mouse xenograft models of prostate cancer, we also show that CF3-Enigmol underperformed Enigmol-mediated inhibition of tumor growth and elicited systemic toxicity. By contrast, CF2-Enigmol was not systemically toxic and demonstrated significantly enhanced antitumor activity as compared to Enigmol.


Journal of Biological Chemistry | 2016

Crystal structures of the nuclear receptor, liver receptor homolog 1, bound to synthetic agonists

Suzanne G. Mays; C. Denise Okafor; Richard J. Whitby; Devrishi Goswami; Jozef Stec; Autumn R. Flynn; Michael C. Dugan; Nathan T. Jui; Patrick R. Griffin; Eric A. Ortlund

Liver receptor homolog 1 (NR5A2, LRH-1) is an orphan nuclear hormone receptor that regulates diverse biological processes, including metabolism, proliferation, and the resolution of endoplasmic reticulum stress. Although preclinical and cellular studies demonstrate that LRH-1 has great potential as a therapeutic target for metabolic diseases and cancer, development of LRH-1 modulators has been difficult. Recently, systematic modifications to one of the few known chemical scaffolds capable of activating LRH-1 failed to improve efficacy substantially. Moreover, mechanisms through which LRH-1 is activated by synthetic ligands are entirely unknown. Here, we use x-ray crystallography and other structural methods to explore conformational changes and receptor-ligand interactions associated with LRH-1 activation by a set of related agonists. Unlike phospholipid LRH-1 ligands, these agonists bind deep in the pocket and do not interact with residues near the mouth nor do they expand the pocket like phospholipids. Unexpectedly, two closely related agonists with similar efficacies (GSK8470 and RJW100) exhibit completely different binding modes. The dramatic repositioning is influenced by a differential ability to establish stable face-to-face π-π-stacking with the LRH-1 residue His-390, as well as by a novel polar interaction mediated by the RJW100 hydroxyl group. The differing binding modes result in distinct mechanisms of action for the two agonists. Finally, we identify a network of conserved water molecules near the ligand-binding site that are important for activation by both agonists. This work reveals a previously unappreciated complexity associated with LRH-1 agonist development and offers insights into rational design strategies.


Molecular Pharmacology | 2017

Structure and Dynamics of the Liver Receptor Homolog 1-PGC1 alpha Complex.

Suzanne G. Mays; C. Denise Okafor; Micheal L. Tuntland; Richard J. Whitby; Venkatasubramanian Dharmarajan; Jozef Stec; Patrick R. Griffin; Eric A. Ortlund

Peroxisome proliferator-activated gamma coactivator 1-α (PGC1α) regulates energy metabolism by directly interacting with transcription factors to modulate gene expression. Among the PGC1α binding partners is liver receptor homolog 1 (LRH-1; NR5A2), an orphan nuclear hormone receptor that controls lipid and glucose homeostasis. Although PGC1α is known to bind and activate LRH-1, mechanisms through which PGC1α changes LRH-1 conformation to drive transcription are unknown. Here, we used biochemical and structural methods to interrogate the LRH-1–PGC1α complex. Purified, full-length LRH-1, as well as isolated ligand binding domain, bound to PGC1α with higher affinity than to the coactivator, nuclear receptor coactivator-2 (Tif2), in coregulator peptide recruitment assays. We present the first crystal structure of the LRH-1–PGC1α complex, which depicts several hydrophobic contacts and a strong charge clamp at the interface between these partners. In molecular dynamics simulations, PGC1α induced correlated atomic motion throughout the entire LRH-1 activation function surface, which was dependent on charge-clamp formation. In contrast, Tif2 induced weaker signaling at the activation function surface than PGC1α but promoted allosteric signaling from the helix 6/β-sheet region of LRH-1 to the activation function surface. These studies are the first to probe mechanisms underlying the LRH-1–PGC1α interaction and may illuminate strategies for selective therapeutic targeting of PGC1α-dependent LRH-1 signaling pathways.


ACS Medicinal Chemistry Letters | 2018

Development of Hybrid Phospholipid Mimics as Effective Agonists for Liver Receptor Homologue-1

Autumn R. Flynn; Suzanne G. Mays; Eric A. Ortlund; Nathan T. Jui

The orphan nuclear receptor Liver Receptor Homologue-1 (LRH-1) is an emerging drug target for metabolic disorders. The most effective known LRH-1 modulators are phospholipids or synthetic hexahydropentalene compounds. While both classes have micromolar efficacy, they target different portions of the ligand binding pocket and activate LRH-1 through different mechanisms. Guided by crystallographic data, we combined aspects of both ligand classes into a single scaffold, resulting in the most potent and efficacious LRH-1 agonists to date.


Acta Crystallographica Section A | 2017

Targeting the nuclear receptor LRH-1 with synthetic agonists

Suzanne G. Mays; C. Denise Okafor; Richard J. Whitby; Devrishi Goswami; Jozef Stec; Autumn R. Flynn; Michael C. Dugan; Nathan T. Jui; Patrick R. Griffin; Eric A. Ortlund

Liver receptor homolog 1 (NR5A2, LRH-1) is an orphan nuclear hormone receptor that regulates diverse biological processes, including lipid and glucose metabolism, proliferation, and the resolution of endoplasmic reticulum stress. Preclinical studies have demonstrated a great therapeutic potential of targeting LRH-1 for treatment of metabolic diseases, such as diabetes; however, development of LRH-1 modulators has been challenging. In a recent study, systematic modifications to one of the few known chemical scaffolds capable of activating LRH-1 failed to improve efficacy substantially. Moreover, mechanisms through which LRH-1 is activated by synthetic ligands are entirely unknown. Here, we use x-ray crystallography, molecular dynamics simulations, and cellular activation assays to explore conformational changes and receptor-ligand interactions associated with LRH-1 activation by a set of related agonists. Unlike phospholipid (PL) LRH-1 ligands, these agonists bind deep in the pocket and do not interact with residues near the mouth, nor do they expand the pocket like PLs. Unexpectedly, two closely related agonists with similar efficacies (GSK8470 and RJW100) exhibit completely different binding modes. The dramatic repositioning is influenced by a differential ability to establish stable, face-to-face π-π-stacking with LRH1 residue H390, as well as by a novel polar interaction mediated by the RJW100 hydroxyl group. The differing binding modes result in distinct mechanisms of action for the two agonists. This work reveals a previously unappreciated complexity associated with LRH-1 agonist development and offers insights into rational design strategies.


The Journal of Urology | 2013

330 FLUORINATION OF ENIGMOL IMPROVES TISSUE UPTAKE AND AFFECTS IN VIVOPROSTATE CANCER EFFICACY

Suzanne G. Mays; Mark T. Baillie; Eric Miller; Anatoliy S. Bushnev; Sarah T. Pruett; Deborah G. Culver; Taylor J. Evers; Jingjing Gao; G. Prakabahr Reddy; Michael G. Natchus; Richard F. Arrendale; Randy B. Howard; Dennis C. Liotta; John A. Petros


The FASEB Journal | 2015

Role of Macroautophagy in Anti-Cancer Effects of the Novel Sphingolipid, Enigmol

Suzanne G. Mays; Anatoliy S. Bushnev; Mark T. Baillie; Hong Yi; Dennis C. Liotta; John A. Petros


The Journal of Urology | 2010

1438 THE CANCER DRUG ENIGMOL ALTERS SPHINGOLIPID METABOLISM AND REDUCES MACROPHAGE PRESENCE IN PROSTATE TUMORS

Suzanne G. Mays; Harsha Ramaraju; Anatoliy Buushnev; Adeboye O. Osunkoya; Elayne Wang; Dennis C. Liotta; Alfred H. Merrill; John A. Petros

Collaboration


Dive into the Suzanne G. Mays's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harsha Ramaraju

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alfred H. Merrill

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge