Network


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

Hotspot


Dive into the research topics where Virginia M. Salas is active.

Publication


Featured researches published by Virginia M. Salas.


Analytical Biochemistry | 2013

Fluorescent substrates for flow cytometric evaluation of efflux inhibition in ABCB1, ABCC1, and ABCG2 transporters

J. Jacob Strouse; Irena Ivnitski-Steele; Anna Waller; Susan M. Young; Dominique Perez; Annette M. Evangelisti; Oleg Ursu; Cristian G. Bologa; Mark B. Carter; Virginia M. Salas; George P. Tegos; Richard S. Larson; Tudor I. Oprea; Bruce S. Edwards; Larry A. Sklar

ATP binding cassette (ABC) transmembrane efflux pumps such as P-glycoprotein (ABCB1), multidrug resistance protein 1 (ABCC1), and breast cancer resistance protein (ABCG2) play an important role in anticancer drug resistance. A large number of structurally and functionally diverse compounds act as substrates or modulators of these pumps. In vitro assessment of the affinity of drug candidates for multidrug resistance proteins is central to predict in vivo pharmacokinetics and drug-drug interactions. The objective of this study was to identify and characterize new substrates for these transporters. As part of a collaborative project with Life Technologies, 102 fluorescent probes were investigated in a flow cytometric screen of ABC transporters. The primary screen compared substrate efflux activity in parental cell lines with their corresponding highly expressing resistant counterparts. The fluorescent compound library included a range of excitation/emission profiles and required dual laser excitation as well as multiple fluorescence detection channels. A total of 31 substrates with active efflux in one or more pumps and practical fluorescence response ranges were identified and tested for interaction with eight known inhibitors. This screening approach provides an efficient tool for identification and characterization of new fluorescent substrates for ABCB1, ABCC1, and ABCG2.


Journal of Biomolecular Screening | 2013

A Selective ATP-Binding Cassette Subfamily G Member 2 Efflux Inhibitor Revealed via High-Throughput Flow Cytometry

J. Jacob Strouse; Irena Ivnitski-Steele; Hadya M. Khawaja; Dominique Perez; Jerec Ricci; Tuanli Yao; Warren S. Weiner; Chad E. Schroeder; Denise S. Simpson; Brooks E. Maki; Kelin Li; Jennifer E. Golden; Terry D. Foutz; Anna Waller; Annette M. Evangelisti; Susan M. Young; Stephanie E. Chavez; Matthew Garcia; Oleg Ursu; Cristian G. Bologa; Mark B. Carter; Virginia M. Salas; Kristine Gouveia; George P. Tegos; Tudor I. Oprea; Bruce S. Edwards; Jeffrey Aubé; Richard S. Larson; Larry A. Sklar

Chemotherapeutics tumor resistance is a principal reason for treatment failure, and clinical and experimental data indicate that multidrug transporters such as ATP-binding cassette (ABC) B1 and ABCG2 play a leading role by preventing cytotoxic intracellular drug concentrations. Functional efflux inhibition of existing chemotherapeutics by these pumps continues to present a promising approach for treatment. A contributing factor to the failure of existing inhibitors in clinical applications is limited understanding of specific substrate/inhibitor/pump interactions. We have identified selective efflux inhibitors by profiling multiple ABC transporters against a library of small molecules to find molecular probes to further explore such interactions. In our primary screening protocol using JC-1 as a dual-pump fluorescent reporter substrate, we identified a piperazine-substituted pyrazolo[1,5-a]pyrimidine substructure with promise for selective efflux inhibition. As a result of a focused structure-activity relationship (SAR)–driven chemistry effort, we describe compound 1 (CID44640177), an efflux inhibitor with selectivity toward ABCG2 over ABCB1. Compound 1 is also shown to potentiate the activity of mitoxantrone in vitro as well as preliminarily in vivo in an ABCG2-overexpressing tumor model. At least two analogues significantly reduce tumor size in combination with the chemotherapeutic topotecan. To our knowledge, low nanomolar chemoreversal activity coupled with direct evidence of efflux inhibition for ABCG2 is unprecedented.


Current protocols in immunology | 2007

High‐Throughput Cytotoxicity Screening by Propidium Iodide Staining

Bruce S. Edwards; Irena Ivnitski-Steele; Susan M. Young; Virginia M. Salas; Larry A. Sklar

This unit describes a system for the automated high‐throughput analysis of cell cytotoxicity in 96‐well and 384‐well microplates. Discrete cell cultures are analyzed at rates of 40/min (∼2.5 min/96 wells, ∼10 min/384 wells) and cytotoxicity is quantified on the basis of a combination of propidium iodide (PI) fluorescence analysis and cell counting performed by the flow cytometer. Only 2 µl is aspirated from a culture for analysis so that assays can be performed in small volumes to minimize reagent cost and usage.


Advances in Clinical Chemistry | 2008

Advances in Multiple Analyte Profiling

Virginia M. Salas; Bruce S. Edwards; Larry A. Sklar

The advent of multiparameter technology has been driven by the need to understand the complexity in biological systems. It has spawned two main branches, one in the arena of high-content measurements, primarily in microscopy and flow cytometry where it has become commonplace to analyze multiple fluorescence signatures arising from multiple excitation sources and multiple emission wavelengths. Microscopy is augmented by topographical content that identifies the source location of the signature. The other branch involves multiplex technology. Here, the intent is to measure multiple analytes simultaneously. A key feature of multiplexing is an address system for the individual analytes. In planar arrays the address system is spatial, in which affinity reactions occur at defined locations. In suspension arrays, the address is encoded as a fluorescent signature in the particle assigned to a specific reaction or analyte. Several hybrid systems have also been developed for multiplexing. In the commercial regime, the most widespread applications of multiplexing are currently in the areas of genome and biomarker analysis. Planar chips with fixed arrays are now available to probe the entire genome at the level of message expression and large segments of the genome at the level of single nucleotide polymorphism (SNP). In contrast, suspension arrays provide the potential for probing segments of the genome in a customized way, using capture tags that locate specific oligonucleotide sequences to specific array elements.


In Vitro Cellular & Developmental Biology – Animal | 1996

Purification of primary human hepatocytes using ricin a chain

David E. Johnston; Daniel Marcadis; George B. Corcoran; Michele DeBetta; Virginia M. Salas; Hector Ramos

Dear Editor: In a previous publication, we documented that primary rat hepatocyte cultures contain significant numbers of Kupffer and endothelial cells (3). This contamination was found despite the use of various published methods for the purification of primary hepatocytes. We developed a method for purifying rat hepatocyte monolayers based on the selective uptake of the active A subunit of ricin by Kupffer and endothelial cells (3,5). Ricin inhibits protein synthesis by catalytically hydrolyzing a critical adenine from the 18S ribosomal RNA and thus destroying ribosomes (2). We now report that primary human hepatocytes contain significant numbers of contaminating Kupffer and endothelial cells and that ricin A chain can be used to purify human hepatocytes. A wedge of human liver tissue was obtained from the normal margin of a liver lobe resected for a giant cavernous hemangioma, a benign tumor. Normal liver from the resection margin was used with the patients consent, under a protocol approved by the Human Research Review Committee of the University of New Mexico. Two-step collagenase perfusion was performed through two cut veins visible in the edge of the liver, as previously described (3). The collagenase digest was passed through a sterile 80-~tm mesh and partially purified by sedimentation three times at 50 • g for 2 min. Hepatocyte viability was 92% by Trypan blue exclusion. One million viable hepatocytes in 1.5 ml of medium were plated on each collagen-coated 35mm plastic dish. The medium consisted of Williams medium E with 5% fetal bovine serum (Sigma Chemical Co., St. Louis, MO) and a mixture of hormones as previously described (4).


ACS Chemical Biology | 2018

Activation of Rho Family GTPases by Small Molecules

Charuta C. Palsuledesai; Zurab Surviladze; Anna Waller; T. Fabiola Miscioscia; Yuna Guo; Yang Wu; Jake Strouse; Elsa Romero; Virginia M. Salas; Ramona Curpan; Susan Young; Mark B. Carter; Terry D. Foutz; Zhanna Galochkina; Harold Ames; Mark K. Haynes; Bruce S. Edwards; Orazio Nicolotti; Li Luo; Oleg Ursu; Cristian G. Bologa; Tudor I. Oprea; Angela Wandinger-Ness; Larry A. Sklar

Ras and Ras-related small GTPases are key regulators of diverse cellular functions that impact cell growth, survival, motility, morphogenesis, and differentiation. They are important targets for studies of disease mechanisms as well as drug discovery. Here, we report the characterization of small molecule agonists of one or more of six Rho, Rab, and Ras family GTPases that were first identified through flow cytometry-based, multiplexed high-throughput screening of 200000 compounds. The activators were categorized into three distinct chemical families that are represented by three lead compounds having the highest activity. Virtual screening predicted additional compounds with potential GTPase activating properties. Secondary dose–response assays performed on compounds identified through these screens confirmed agonist activity of 43 compounds. While the lead and second most active small molecules acted as pan activators of multiple GTPase subfamilies, others showed partial selectivity for Ras and Rab proteins. The compounds did not stimulate nucleotide exchange by guanine nucleotide exchange factors and did not protect against GAP-stimulated GTP hydrolysis. The activating properties were caused by a reversible stabilization of the GTP-bound state and prolonged effector protein interactions. Notably, these compounds were active both in vitro and in cell-based assays, and small molecule-mediated changes in Rho GTPase activities were directly coupled to measurable changes in cytoskeletal rearrangements that dictate cell morphology.


Toxicology and Applied Pharmacology | 1998

Apoptosis in Daudi Human B Cells in Response to Benzo[a]pyrene and Benzo[a]pyrene-7,8-dihydrodiol☆

Virginia M. Salas; Scott W. Burchiel


Carcinogenesis | 2000

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) inhibits growth factor withdrawal-induced apoptosis in the human mammary epithelial cell line, MCF-10A

John W. Davis; Karla Melendez; Virginia M. Salas; Fredine T. Lauer; Scott W. Burchiel


Hepatology | 1997

Calcium-dependent DNA damage and adenosine 3',5'-cyclic monophosphate-independent glycogen phosphorylase activation in an in vitro model of acetaminophen-induced liver injury.

Virginia M. Salas; George B. Corcoran


Archive | 2010

A Potent and Selective Inhibitor of Cdc42 GTPase

Zurab Surviladze; Anna Waller; J. Jacob Strouse; Cristian G. Bologa; Oleg Ursu; Virginia M. Salas; Genevieve K Phillips; John F Parkinson; Elsa Romero; Angela Wandinger-Ness; Larry A. Sklar; Chad E. Schroeder; Denise S. Simpson; Julica Nöth; Jenna Wang; Jennifer E. Golden; Jeffrey Aubé

Collaboration


Dive into the Virginia M. Salas's collaboration.

Top Co-Authors

Avatar

Larry A. Sklar

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Anna Waller

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar

Oleg Ursu

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar

Bruce S. Edwards

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tudor I. Oprea

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark B. Carter

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar

Susan M. Young

University of New Mexico

View shared research outputs
Researchain Logo
Decentralizing Knowledge