Network


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

Hotspot


Dive into the research topics where Mark B. Carter is active.

Publication


Featured researches published by Mark B. Carter.


Nature Materials | 2011

The targeted delivery of multicomponent cargos to cancer cells by nanoporous particle-supported lipid bilayers

Carlee E. Ashley; Eric C. Carnes; Genevieve K Phillips; David Padilla; Paul N. Durfee; Page A. Brown; Tracey N. Hanna; Juewen Liu; Brandy Phillips; Mark B. Carter; Nick J. Carroll; Xingmao Jiang; Darren R. Dunphy; Cheryl L. Willman; Dimiter N. Petsev; Deborah G. Evans; Atul N. Parikh; Bryce Chackerian; Walker Wharton; David S. Peabody; C. Jeffrey Brinker

Encapsulation of drugs within nanocarriers that selectively target malignant cells promises to mitigate side effects of conventional chemotherapy and to enable delivery of the unique drug combinations needed for personalized medicine. To realize this potential, however, targeted nanocarriers must simultaneously overcome multiple challenges, including specificity, stability, and a high capacity for disparate cargos. Here we report porous nanoparticle-supported lipid bilayers (protocells) that synergistically combine properties of liposomes and nanoporous particles. Protocells modified with a targeting peptide that binds to human hepatocellular carcinoma (HCC) exhibit a 10,000-fold greater affinity for HCC than for hepatocytes, endothelial cells, and immune cells. Furthermore, protocells can be loaded with combinations of therapeutic (drugs, siRNA, and toxins) and diagnostic (quantum dots) agents and modified to promote endosomal escape and nuclear accumulation of selected cargos. The enormous capacity of the high-surface-area nanoporous core combined with the enhanced targeting efficacy enabled by the fluid supported lipid bilayer allow a single protocell loaded with a drug cocktail to kill a drug-resistant HCC cell, representing a 106-fold improvement over comparable liposomes.


ACS Nano | 2011

Cell-Specific Delivery of Diverse Cargos by Bacteriophage MS2 Virus-Like Particles

Carlee E. Ashley; Eric C. Carnes; Genevieve K Phillips; Paul N. Durfee; Buley; Christopher A. Lino; David Padilla; Brandy Phillips; Mark B. Carter; Cheryl L. Willman; Brinker Cj; Caldeira Jdo C; Bryce Chackerian; Walker Wharton; David S. Peabody

Virus-like particles (VLPs) of bacteriophage MS2 possess numerous features that make them well-suited for use in targeted delivery of therapeutic and imaging agents. MS2 VLPs can be rapidly produced in large quantities using in vivo or in vitro synthesis techniques. Their capsids can be modified in precise locations via genetic insertion or chemical conjugation, facilitating the multivalent display of targeting ligands. MS2 VLPs also self-assemble in the presence of nucleic acids to specifically encapsidate siRNA and RNA-modified cargos. Here we report the use of MS2 VLPs to selectively deliver nanoparticles, chemotherapeutic drugs, siRNA cocktails, and protein toxins to human hepatocellular carcinoma (HCC). MS2 VLPs modified with a peptide (SP94) that binds HCC exhibit a 10(4)-fold higher avidity for HCC than for hepatocytes, endothelial cells, monocytes, or lymphocytes and can deliver high concentrations of encapsidated cargo to the cytosol of HCC cells. SP94-targeted VLPs loaded with doxorubicin, cisplatin, and 5-fluorouracil selectively kill the HCC cell line, Hep3B, at drug concentrations <1 nM, while SP94-targeted VLPs that encapsidate a siRNA cocktail, which silences expression of cyclin family members, induce growth arrest and apoptosis of Hep3B at siRNA concentrations <150 pM. Impressively, MS2 VLPs, when loaded with ricin toxin A-chain (RTA) and modified to codisplay the SP94 targeting peptide and a histidine-rich fusogenic peptide (H5WYG) that promotes endosomal escape, kill virtually the entire population of Hep3B cells at an RTA concentration of 100 fM without affecting the viability of control cells. Our results demonstrate that MS2 VLPs, because of their tolerance of multivalent peptide display and their ability to specifically encapsidate a variety of chemically disparate cargos, induce selective cytotoxicity of cancer in vitro and represent a significant improvement in the characteristics of VLP-based delivery systems.


Journal of Biological Chemistry | 2013

Characterization of a Cdc42 Protein Inhibitor and Its Use as a Molecular Probe

Lin Hong; S. Ray Kenney; Genevieve K Phillips; Denise S. Simpson; Chad E. Schroeder; Julica Nöth; Elsa Romero; Scarlett Swanson; Anna Waller; J. Jacob Strouse; Mark B. Carter; Alexandre Chigaev; Oleg Ursu; Tudor I. Oprea; Brian Hjelle; Jennifer E. Golden; Jeffrey Aubé; Laurie G. Hudson; Tione Buranda; Larry A. Sklar; Angela Wandinger-Ness

Background: By integrating extracellular signals with actin cytoskeletal changes, Cdc42 plays important roles in cell physiology and has been implicated in human diseases. Results: A small molecule was found to selectively inhibit Cdc42 in biochemical and cellular assays. Conclusion: The identified compound is a highly Cdc42-selective inhibitor. Significance: The described first-in-class Cdc42 GTPase-selective inhibitor will have applications in drug discovery and fundamental research. Cdc42 plays important roles in cytoskeleton organization, cell cycle progression, signal transduction, and vesicle trafficking. Overactive Cdc42 has been implicated in the pathology of cancers, immune diseases, and neuronal disorders. Therefore, Cdc42 inhibitors would be useful in probing molecular pathways and could have therapeutic potential. Previous inhibitors have lacked selectivity and trended toward toxicity. We report here the characterization of a Cdc42-selective guanine nucleotide binding lead inhibitor that was identified by high throughput screening. A second active analog was identified via structure-activity relationship studies. The compounds demonstrated excellent selectivity with no inhibition toward Rho and Rac in the same GTPase family. Biochemical characterization showed that the compounds act as noncompetitive allosteric inhibitors. When tested in cellular assays, the lead compound inhibited Cdc42-related filopodia formation and cell migration. The lead compound was also used to clarify the involvement of Cdc42 in the Sin Nombre virus internalization and the signaling pathway of integrin VLA-4. Together, these data present the characterization of a novel Cdc42-selective allosteric inhibitor and a related analog, the use of which will facilitate drug development targeting Cdc42-related diseases and molecular pathway studies that involve GTPases.


Molecular Biology of the Cell | 2011

The proteomics of quiescent and nonquiescent cell differentiation in yeast stationary-phase cultures

George S. Davidson; Ray M. Joe; Sushmita Roy; Osorio Meirelles; Chris Allen; Melissa R. Wilson; Phillip H. Tapia; Elaine Manzanilla; Anne E. Dodson; Swagata Chakraborty; Mark B. Carter; Susan Young; Bruce S. Edwards; Larry A. Sklar; Margaret Werner-Washburne

As yeast cultures enter stationary phase in rich, glucose-based medium, differentiation of two major subpopulations of cells, termed quiescent and nonquiescent, is observed. Differences in mRNA abundance between exponentially growing and stationary-phase cultures and quiescent and nonquiescent cells are known, but little was known about protein abundance in these cells. To measure protein abundance in exponential and stationary-phase cultures, the yeast GFP-fusion library (4159 strains) was examined during exponential and stationary phases, using high-throughput flow cytometry (HyperCyt). Approximately 5% of proteins in the library showed twofold or greater changes in median fluorescence intensity (abundance) between the two conditions. We examined 38 strains exhibiting two distinct fluorescence-intensity peaks in stationary phase and determined that the two fluorescence peaks distinguished quiescent and nonquiescent cells, the two major subpopulations of cells in stationary-phase cultures. GFP-fusion proteins in this group were more abundant in quiescent cells, and half were involved in mitochondrial function, consistent with the sixfold increase in respiration observed in quiescent cells and the relative absence of Cit1p:GFP in nonquiescent cells. Finally, examination of quiescent cell-specific GFP-fusion proteins revealed symmetry in protein accumulation in dividing quiescent and nonquiescent cells after glucose exhaustion, leading to a new model for the differentiation of these cells.


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.


Cytometry Part A | 2012

Cluster cytometry for high-capacity bioanalysis

Bruce S. Edwards; Jingshu S. Zhu; Jun Chen; Mark B. Carter; David M. Thal; John J. G. Tesmer; Steven W. Graves; Larry A. Sklar

Flow cytometry specializes in high‐content measurements of cells and particles in suspension. Having long excelled in analytical throughput of single cells and particles, only recently with the advent of HyperCyt sampling technology, flow cytometrys multiexperiment throughput has begun to approach the point of practicality for efficiently analyzing hundreds‐of‐thousands of samples, the realm of high‐throughput screening (HTS). To extend performance and automation compatibility, we built a HyperCyt‐linked Cluster Cytometer platform, a network of flow cytometers for analyzing samples displayed in high‐density, 1,536‐well plate format. To assess the performance, we used cell‐ and microsphere‐based HTS assays that had been well characterized in the previous studies. Experiments addressed important technical issues: challenges of small wells (assay volumes 10 μL or less, reagent mixing, cell and particle suspension), detecting and correcting for differences in performance of individual flow cytometers, and the ability to reanalyze a plate in the event of problems encountered during the primary analysis. Boosting sample throughput an additional fourfold, this platform is uniquely positioned to synergize with expanding suspension array and cell barcoding technologies in which as many as 100 experiments are performed in a single well or sample. As high‐performance flow cytometers shrink in cost and size, cluster cytometry promises to become a practical, productive approach for HTS, and other large‐scale investigations of biological complexity.


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.


Nature Protocols | 2011

Simultaneous in vitro Molecular Screening of Protein - Peptide Interactions by Flow Cytometry Using Six Bcl-2 Family Proteins as Examples

Peter C. Simons; Susan M. Young; Mark B. Carter; Anna Waller; Dayong Zhai; John C. Reed; Bruce S. Edwards; Larry A. Sklar

The B-cell lymphoma-2 (Bcl-2) family contains six antiapoptotic members, each with a hydrophobic pocket in which Bcl-2 homology region 3 (BH3) helices bind. This binding quenches apoptotic signals from activated BH3 family members. Many tumor cells either have increased expression of one of these six proteins or become overexpressed under treatment. Six fusion proteins made up of glutathione-S-transferase and each of the Bcl-2 members are bound individually to six glutathione bead sets, each set being easily distinguished by its different intensity of red fluorescence. The coated bead sets are washed, combined and incubated with green fluorescent Bim-BH3 peptide and a small molecule in 10-μl wells for 1 h. The green fluorescence signal for each bead set is resolved, and selective inhibitors are expected to reduce the signal for individual bead sets. Each 384-well plate is analyzed in 12 min, measuring 200 of 2,000 beads (∼10%) of each type per well.


ACS Chemical Biology | 2012

Identification of a Small Molecule Yeast TORC1 Inhibitor with a Multiplex Screen Based on Flow Cytometry

Jun Chen; Susan M. Young; Chris Allen; Andrew Seeber; Marie-Pierre Péli-Gulli; Nicolas Panchaud; Anna Waller; Oleg Ursu; Tuanli Yao; Jennifer E. Golden; J. Jacob Strouse; Mark B. Carter; Huining Kang; Cristian G. Bologa; Terry D. Foutz; Bruce S. Edwards; Blake R. Peterson; Jeffrey Aubé; Margaret Werner-Washburne; Robbie Loewith; Claudio De Virgilio; Larry A. Sklar

TOR (target of rapamycin) is a serine/threonine kinase, evolutionarily conserved from yeast to human, which functions as a fundamental controller of cell growth. The moderate clinical benefit of rapamycin in mTOR-based therapy of many cancers favors the development of new TOR inhibitors. Here we report a high-throughput flow cytometry multiplexed screen using five GFP-tagged yeast clones that represent the readouts of four branches of the TORC1 signaling pathway in budding yeast. Each GFP-tagged clone was differentially color-coded, and the GFP signal of each clone was measured simultaneously by flow cytometry, which allows rapid prioritization of compounds that likely act through direct modulation of TORC1 or proximal signaling components. A total of 255 compounds were confirmed in dose-response analysis to alter GFP expression in one or more clones. To validate the concept of the high-throughput screen, we have characterized CID 3528206, a small molecule most likely to act on TORC1 as it alters GFP expression in all five GFP clones in a manner analogous to that of rapamycin. We have shown that CID 3528206 inhibited yeast cell growth and that CID 3528206 inhibited TORC1 activity both in vitro and in vivo with EC(50)s of 150 nM and 3.9 μM, respectively. The results of microarray analysis and yeast GFP collection screen further support the notion that CID 3528206 and rapamycin modulate similar cellular pathways. Together, these results indicate that the HTS has identified a potentially useful small molecule for further development of TOR inhibitors.


PLOS ONE | 2015

A Pan-GTPase Inhibitor as a Molecular Probe.

Lin Hong; Yuna Guo; Soumik BasuRay; Jacob O. Agola; Elsa Romero; Denise S. Simpson; Chad E. Schroeder; Peter C. Simons; Anna Waller; Matthew Garcia; Mark B. Carter; Oleg Ursu; Kristine Gouveia; Jennifer E. Golden; Jeffrey Aubé; Angela Wandinger-Ness; Larry A. Sklar

Overactive GTPases have often been linked to human diseases. The available inhibitors are limited and have not progressed far in clinical trials. We report here a first-in-class small molecule pan-GTPase inhibitor discovered from a high throughput screening campaign. The compound CID1067700 inhibits multiple GTPases in biochemical, cellular protein and protein interaction, as well as cellular functional assays. In the biochemical and protein interaction assays, representative GTPases from Rho, Ras, and Rab, the three most generic subfamilies of the GTPases, were probed, while in the functional assays, physiological processes regulated by each of the three subfamilies of the GTPases were examined. The chemical functionalities essential for the activity of the compound were identified through structural derivatization. The compound is validated as a useful molecular probe upon which GTPase-targeting inhibitors with drug potentials might be developed.

Collaboration


Dive into the Mark B. Carter'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

Tudor I. Oprea

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar

Susan M. Young

University of New Mexico

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge