Network


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

Hotspot


Dive into the research topics where Dominique Perez is active.

Publication


Featured researches published by Dominique Perez.


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.


Analytical Biochemistry | 2010

Recognition of decay accelerating factor and αvβ3 by inactivated hantaviruses: Toward the development of high-throughput screening flow cytometry assays

Tione Buranda; Yang Wu; Dominique Perez; Stephen D. Jett; Virginie Bondu-Hawkins; Chunyan Ye; Bruce S. Edwards; Pamela R. Hall; Richard S. Larson; Gabriel P. Lopez; Larry A. Sklar; Brian Hjelle

Hantaviruses cause two severe diseases in humans: hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS). The lack of vaccines or specific drugs to prevent or treat HFRS and HCPS and the requirement for conducting experiments in a biosafety level 3 laboratory (BSL-3) limit the ability to probe the mechanism of infection and disease pathogenesis. In this study, we developed a generalizable spectroscopic assay to quantify saturable fluorophore sites solubilized in envelope membranes of Sin Nombre virus (SNV) particles. We then used flow cytometry and live cell confocal fluorescence microscopy imaging to show that ultraviolet (UV)-killed SNV particles bind to the cognate receptors of live virions, namely, decay accelerating factor (DAF/CD55) expressed on Tanoue B cells and alpha(v)beta(3) integrins expressed on Vero E6 cells. SNV binding to DAF is multivalent and of high affinity (K(d) approximately 26pM). Self-exchange competition binding assays between fluorescently labeled SNV and unlabeled SNV are used to evaluate an infectious unit-to-particle ratio of approximately 1:14,000. We configured the assay for measuring the binding of fluorescently labeled SNV to Tanoue B suspension cells using a high-throughput flow cytometer. In this way, we established a proof-of-principle high-throughput screening (HTS) assay for binding inhibition. This is a first step toward developing HTS format assays for small molecule inhibitors of viral-cell interactions as well as dissecting the mechanism of infection in a BSL-2 environment.


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.


PLOS ONE | 2012

A Novel Flow Cytometric HTS Assay Reveals Functional Modulators of ATP Binding Cassette Transporter ABCB6

Kishore Polireddy; Mohiuddin Md. Taimur Khan; Hemantkumar Chavan; Susan Young; Xiaochao Ma; Anna Waller; Matthew Garcia; Dominique Perez; Stephanie E. Chavez; J. Jacob Strouse; Mark K. Haynes; Cristian G. Bologa; Tudor I. Oprea; George P. Tegos; Larry A. Sklar; Partha Krishnamurthy

ABCB6 is a member of the adenosine triphosphate (ATP)-binding cassette family of transporter proteins that is increasingly recognized as a relevant physiological and therapeutic target. Evaluation of modulators of ABCB6 activity would pave the way toward a more complete understanding of the significance of this transport process in tumor cell growth, proliferation and therapy-related drug resistance. In addition, this effort would improve our understanding of the function of ABCB6 in normal physiology with respect to heme biosynthesis, and cellular adaptation to metabolic demand and stress responses. To search for modulators of ABCB6, we developed a novel cell-based approach that, in combination with flow cytometric high-throughput screening (HTS), can be used to identify functional modulators of ABCB6. Accumulation of protoporphyrin, a fluorescent molecule, in wild-type ABCB6 expressing K562 cells, forms the basis of the HTS assay. Screening the Prestwick Chemical Library employing the HTS assay identified four compounds, benzethonium chloride, verteporfin, tomatine hydrochloride and piperlongumine, that reduced ABCB6 mediated cellular porphyrin levels. Validation of the identified compounds employing the hemin-agarose affinity chromatography and mitochondrial transport assays demonstrated that three out of the four compounds were capable of inhibiting ABCB6 mediated hemin transport into isolated mitochondria. However, only verteporfin and tomatine hydrochloride inhibited ABCB6’s ability to compete with hemin as an ABCB6 substrate. This assay is therefore sensitive, robust, and suitable for automation in a high-throughput environment as demonstrated by our identification of selective functional modulators of ABCB6. Application of this assay to other libraries of synthetic compounds and natural products is expected to identify novel modulators of ABCB6 activity.


Oncotarget | 2016

Cyclic AMP efflux inhibitors as potential therapeutic agents for leukemia

Dominique Perez; Yelena Smagley; Matthew Garcia; Mark B. Carter; Annette M. Evangelisti; Ksenia Matlawska-Wasowska; Stuart S. Winter; Larry A. Sklar; Alexandre Chigaev

Apoptotic evasion is a hallmark of cancer. We propose that some cancers may evade cell death by regulating 3′-5′-cyclic adenosine monophosphate (cAMP), which is associated with pro-apoptotic signaling. We hypothesize that leukemic cells possess mechanisms that efflux cAMP from the cytoplasm, thus protecting them from apoptosis. Accordingly, cAMP efflux inhibition should result in: cAMP accumulation, activation of cAMP-dependent downstream signaling, viability loss, and apoptosis. We developed a novel assay to assess cAMP efflux and performed screens to identify inhibitors. In an acute myeloid leukemia (AML) model, several identified compounds reduced cAMP efflux, appropriately modulated pathways that are responsive to cAMP elevation (cAMP-responsive element-binding protein phosphorylation, and deactivation of Very Late Antigen-4 integrin), and induced mitochondrial depolarization and caspase activation. Blocking adenylyl cyclase activity was sufficient to reduce effects of the most potent compounds. These compounds also decreased cAMP efflux and viability of B-lineage acute lymphoblastic leukemia (B-ALL) cell lines and primary patient samples, but not of normal primary peripheral blood mononuclear cells. Our data suggest that cAMP efflux is a functional feature that could be therapeutically targeted in leukemia. Furthermore, because some of the identified drugs are currently used for treating other illnesses, this work creates an opportunity for repurposing.


SLAS DISCOVERY: Advancing Life Sciences R&D | 2018

High-Throughput Flow Cytometry Identifies Small-Molecule Inhibitors for Drug Repurposing in T-ALL:

Dominique Perez; Christian K. Nickl; Anna Waller; Cristina Delgado-Martin; Travis A. Woods; Nitesh Sharma; Michelle L. Hermiston; Mignon L. Loh; Stephen P. Hunger; Stuart S. Winter; Alexandre Chigaev; Bruce S. Edwards; Larry A. Sklar; Ksenia Matlawska-Wasowska

Kinase inhibitors have dramatically increased patient survival in a multitude of cancers, including hematological malignancies. However, kinase inhibitors have not yet been integrated into current clinical trials for patients with T-cell-lineage acute lymphoblastic leukemia (T-ALL). In this study, we used a high-throughput flow cytometry (HTFC) approach to test a collection of small-molecule inhibitors, including 26 FDA-approved tyrosine kinase inhibitors in a panel of T-ALL cell lines and patient-derived xenografts. Because hypoxia is known to cause resistance to chemotherapy, we developed a synthetic niche that mimics the low oxygen levels found in leukemic bone marrow to evaluate the effects of hypoxia on the tested inhibitors. Drug sensitivity screening was performed using the Agilent BioCel automated liquid handling system integrated with the HyperCyt HT flow cytometry platform, and the uptake of propidium iodide was used as an indication of cell viability. The HTFC dose–response testing identified several compounds that were efficacious in both normal and hypoxic conditions. This study shows that some clinically approved kinase inhibitors target T-ALL in the hypoxic niche of the bone marrow.


SLAS DISCOVERY: Advancing Life Sciences R&D | 2018

A High-Throughput Flow Cytometry Screen Identifies Molecules That Inhibit Hantavirus Cell Entry:

Tione Buranda; Catherine Gineste; Yang Wu; Virginie Bondu; Dominique Perez; Kaylin R. Lake; Bruce S. Edwards; Larry A. Sklar

Hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS), which infects more than 200,000 people worldwide. Sin Nombre virus (SNV) and Andes virus (ANDV) cause the most severe form of HCPS, with case fatality ratios of 30%–40%. There are no specific therapies or vaccines for SNV. Using high-throughput flow cytometry, we screened the Prestwick Chemical Library for small-molecule inhibitors of the binding interaction between UV-inactivated and fluorescently labeled SNVR18 particles, and decay-accelerating factor (DAF) expressed on Tanoue B cells. Eight confirmed hit compounds from the primary screen were investigated further in secondary screens that included infection inhibition, cytotoxicity, and probe interference. Antimycin emerged as a bona fide hit compound that inhibited cellular infection of the major HCPS (SNV)- and HCPS (Hantaan)-causing viruses. Confirming our assay’s ability to detect active compounds, orthogonal testing of the hit compound showed that antimycin binds directly to the virus particle and blocks recapitulation of physiologic integrin activation caused by SNV binding to the integrin PSI domain.


SLAS DISCOVERY: Advancing Life Sciences R&D | 2018

High-Throughput Flow Cytometry Drug Combination Discovery with Novel Synergy Analysis Software, SynScreen

Dominique Perez; Bruce S. Edwards; Larry A. Sklar; Alexandre Chigaev

Classical therapeutic regimens are subject to toxicity, low efficacy, and/or the development of drug resistance. Thus, the discovery of synergistic drug combinations would permit treatment with lower, tolerable dosages of each agent and restored sensitivity. We describe the development and use of the SynScreen software application, which allows for visual and mathematical determinations of compound concentrations that produce super-additive effects. This software uses nonlinear regression fits of dose responses to determine synergism by the Bliss independence and Loewe additivity analysis models. We demonstrate the utility of SynScreen with data analysis from in vitro high-throughput flow cytometry (HTFC) combination screens with repurposed drugs and multiplexed synergy analysis of multiple biologic parameters in parallel. The applicability of SynScreen was confirmed by testing open-source data sets used in published drug combination literature. A key benefit of SynScreen for high-throughput drug combination screening is that observed measurements are graphically depicted in comparison with a three-dimensional surface that represents the theoretical responses at which Bliss additivity would occur. These images and summary tables for the calculated drug interactions are automatically exported. This allows for substantial data sets to be visually assessed, expediting the quick identification of efficacious drug combinations and thereby facilitating the design of confirmatory studies and clinical trials.


Journal of Physical Chemistry B | 2010

Real-Time Partitioning of Octadecyl Rhodamine B into Bead-Supported Lipid Bilayer Membranes Revealing Quantitative Differences in Saturable Binding Sites in DOPC and 1:1:1 DOPC/SM/Cholesterol Membranes

Tione Buranda; Yang Wu; Dominique Perez; Alexandre Chigaev; Larry A. Sklar


Archive | 2013

Profiling a Selective Probe for RTG Branch of Yeast TORC1 Signaling Pathway

Jun Chen; Susan M. Young; Christopher D.C. Allen; Anna Waller; Oleg Ursu; J. Jacob Strouse; Tuanli Yao; Jennifer E. Golden; Blake R. Peterson; Terry D. Foutz; Stephanie E. Chavez; Dominique Perez; Annette M. Evangelisti; Mathew J. Garcia; Cristian G. Bologa; Mark B. Carter; Virginia M. Salas; Tudor I. Oprea; Bruce S. Edwards; Nicolas Panchaud; Claudio De Virgilio; Andrew Seeber; Robbie Loewith; Elaine Manzanilla; Margaret Werner-Washburne; Jeffrey Aubé; Larry A. Sklar

Collaboration


Dive into the Dominique Perez's collaboration.

Top Co-Authors

Avatar

Larry A. Sklar

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Waller

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar
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

Mark B. Carter

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Oleg Ursu

University of New Mexico

View shared research outputs
Researchain Logo
Decentralizing Knowledge