Network


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

Hotspot


Dive into the research topics where Jeffrey D. Clogston is active.

Publication


Featured researches published by Jeffrey D. Clogston.


Nanomedicine: Nanotechnology, Biology and Medicine | 2009

Interaction of colloidal gold nanoparticles with human blood: effects on particle size and analysis of plasma protein binding profiles.

Marina A. Dobrovolskaia; Anil K. Patri; Jiwen Zheng; Jeffrey D. Clogston; Nader Ayub; Parag Aggarwal; Barry W. Neun; Jennifer B. Hall; Scott E. McNeil

Nanoparticle size and plasma binding profile contribute to a particles longevity in the bloodstream, which can have important consequences for therapeutic efficacy. In this study an approximate doubling in nanoparticle hydrodynamic size was observed upon in vitro incubation of 30- and 50-nm colloidal gold in human plasma. Plasma proteins that bind the surface of citrate-stabilized gold colloids have been identified. Effects of protein binding on the nanoparticle hydrodynamic size, elements of coagulation, and the complement system have been investigated. The difference in size measurements obtained from dynamic light scattering, electron microscopy, and scanning probe microscopy are also discussed.


Methods of Molecular Biology | 2011

Zeta Potential Measurement

Jeffrey D. Clogston; Anil K. Patri

This chapter describes a method for the measurement of the electrostatic potential at the electrical double layer surrounding a nanoparticle in solution. This is referred to as the zeta potential. Nanoparticles with a zeta potential between -10 and +10 mV are considered approximately neutral, while nanoparticles with zeta potentials of greater than +30 mV or less than -30 mV are considered strongly cationic and strongly anionic, respectively. Since most cellular membranes are negatively charged, zeta potential can affect a nanoparticles tendency to permeate membranes, with cationic particles generally displaying more toxicity associated with cell wall disruption. This technique is demonstrated for two types of nanoparticles commonly used in biological applications: colloidal gold (strongly anionic) and amine-terminated PAMAM dendrimer (strongly cationic).


Toxicology and Applied Pharmacology | 2010

Fullerenol Cytotoxicity in Kidney Cells is Associated with Cytoskeleton Disruption, Autophagic Vacuole Accumulation, and Mitochondrial Dysfunction

Denise N. Johnson-Lyles; Kimberly Peifley; Stephen J. Lockett; Barry W. Neun; Matthew Hansen; Jeffrey D. Clogston; Stephan T. Stern; Scott E. McNeil

Water soluble fullerenes, such as the hydroxylated fullerene, fullerenol (C₆₀OHx), are currently under development for diagnostic and therapeutic biomedical applications in the field of nanotechnology. These molecules have been shown to undergo urinary clearance, yet there is limited data available on their renal biocompatibility. Here we examine the biological responses of renal proximal tubule cells (LLC-PK1) exposed to fullerenol. Fullerenol was found to be cytotoxic in the millimolar range, with viability assessed by the sulforhodamine B and trypan blue assays. Fullerenol-induced cell death was associated with cytoskeleton disruption and autophagic vacuole accumulation. Interaction with the autophagy pathway was evaluated in vitro by Lysotracker Red dye uptake, LC3-II marker expression and TEM. Fullerenol treatment also resulted in coincident loss of cellular mitochondrial membrane potential and ATP depletion, as measured by the Mitotracker Red dye and the luciferin-luciferase assays, respectively. Fullerenol-induced ATP depletion and loss of mitochondrial potential were partially ameliorated by co-treatment with the autophagy inhibitor, 3-methyladenine. In vitro fullerenol treatment did not result in appreciable oxidative stress, as measured by lipid peroxide and glutathione content. Based on these data, it is hypothesized that cytoskeleton disruption may be an initiating event in fullerenol cytotoxicity, leading to subsequent autophagy dysfunction and loss of mitochondrial capacity. As nanoparticle-induced cytoskeleton disruption, autophagic vacuole accumulation and mitochondrial dysfunction are commonly reported in the literature, the proposed mechanism may be relevant for a variety of nanomaterials.


Clinical Cancer Research | 2010

A Novel Nanoparticulate Formulation of Arsenic Trioxide with Enhanced Therapeutic Efficacy in a Murine Model of Breast Cancer

Richard W. Ahn; Feng Chen; Haimei Chen; Stephan T. Stern; Jeffrey D. Clogston; Anil K. Patri; Meera R. Raja; Elden P. Swindell; Vamsi Parimi; Vincent L. Cryns; Thomas V. O'Halloran

Purpose: The clinical success of arsenic trioxide (As2O3) in hematologic malignancies has not been replicated in solid tumors due to poor pharmacokinetics and dose-limiting toxicity. We have developed a novel nanoparticulate formulation of As2O3 encapsulated in liposomal vesicles or “nanobins” [(NB(Ni,As)] to overcome these hurdles. We postulated that nanobin encapsulation of As2O3 would improve its therapeutic index against clinically aggressive solid tumors, such as triple-negative breast carcinomas. Experimental Design: The cytotoxicity of NB(Ni,As), the empty nanobin, and free As2O3 was evaluated against a panel of human breast cancer cell lines. The plasma pharmacokinetics of NB(Ni,As) and free As2O3 were compared in rats to measure drug exposure. In addition, the antitumor activity of these agents was evaluated in an orthotopic model of human triple-negative breast cancer. Results: The NB(Ni,As) agent was much less cytotoxic in vitro than free As2O3 against a panel of human breast cancer cell lines. In contrast, NB(Ni,As) dramatically potentiated the therapeutic efficacy of As2O3 in vivo in an orthotopic model of triple-negative breast cancer. Reduced plasma clearance, enhanced tumor uptake, and induction of tumor cell apoptosis were observed for NB(Ni,As). Conclusions: Nanobin encapsulation of As2O3 improves the pharmacokinetics and antitumor efficacy of this cytotoxic agent in vivo. Our findings demonstrate the therapeutic potential of this nanoscale agent and provide a foundation for future clinical studies in breast cancer and other solid tumors. Clin Cancer Res; 16(14); 3607–17. ©2010 AACR.


Cancer Letters | 2013

Synergistic Combination Therapy with Nanoliposomal C6-Ceramide and Vinblastine is Associated with Autophagy Dysfunction in Hepatocarcinoma and Colorectal Cancer Models

Pavan P. Adiseshaiah; Jeffrey D. Clogston; Christopher B. McLeland; Jamie Rodriguez; Timothy M. Potter; Barry W. Neun; Sarah L. Skoczen; Sriram S. Shanmugavelandy; Mark Kester; Stephan T. Stern; Scott E. McNeil

Autophagy, a catabolic survival pathway, is gaining attention as a potential target in cancer. In human liver and colon cancer cells, treatment with an autophagy inducer, nanoliposomal C6-ceramide, in combination with the autophagy maturation inhibitor, vinblastine, synergistically enhanced apoptotic cell death. Combination treatment resulted in a marked increase in autophagic vacuole accumulation and decreased autophagy maturation, without diminution of the autophagy flux protein P62. In a colon cancer xenograft model, a single intravenous injection of the drug combination significantly decreased tumor growth in comparison to the individual treatments. Most importantly, the combination treatment did not result in increased toxicity as assessed by body weight loss. The mechanism of combination treatment-induced cell death both in vitro and in vivo appeared to be apoptosis. Supportive of autophagy flux blockade as the underlying synergy mechanism, treatment with other autophagy maturation inhibitors, but not autophagy initiation inhibitors, were similarly synergistic with C6-ceramide. Additionally, knockout of the autophagy protein Beclin-1 suppressed combination treatment-induced apoptosis in vitro. In conclusion, in vitro and in vivo data support a synergistic antitumor activity of the nanoliposomal C6-ceramide and vinblastine combination, potentially mediated by an autophagy mechanism.


Molecular Pharmaceutics | 2010

Biological assessment of triazine dendrimer: toxicological profiles, solution behavior, biodistribution, drug release and efficacy in a PEGylated, paclitaxel construct.

Su Tang Lo; Stephan T. Stern; Jeffrey D. Clogston; Jiwen Zheng; Pavan P. Adiseshaiah; Marina A. Dobrovolskaia; Jongdoo Lim; Anil K. Patri; Xiankai Sun; Eric E. Simanek

The physicochemical characteristics, in vitro properties, and in vivo toxicity and efficacy of a third generation triazine dendrimer bearing approximately nine 2 kDa polyethylene glycol chains and twelve ester linked paclitaxel groups are reported. The hydrodynamic diameter of the neutral construct varies slightly with aqueous solvent ranging from 15.6 to 19.4 nm. Mass spectrometry and light scattering suggest radically different molecular weights with the former approximately 40 kDa mass consistent with expectation, and the latter 400 kDa mass consistent with a decameric structure and the observed hydrodynamic radii. HPLC can be used to assess purity as well as paclitaxel release, which is insignificant in organic solvents or aqueous solutions at neutral and low pH. Paclitaxel release occurs in vitro in human, rat, and mouse plasma and is nonlinear, ranging from 7 to 20% cumulative release over a 48 h incubation period. The construct is 2-3 orders of magnitude less toxic than Taxol by weight in human hepatocarcinoma (Hep G2), porcine renal proximal tubule (LLC-PK1), and human colon carcinoma (LS174T) cells, but shows similar cytotoxicity to Abraxane in LS174T cells. Both Taxol and the construct appear to induce caspase 3-dependent apoptosis. The construct shows a low level of endotoxin, is not hemolytic and does not induce platelet aggregation in vitro, but does appear to reduce collagen-induced platelet aggregation in vitro. Furthermore, the dendrimer formulation slightly activates the complement system in vitro due most likely to the presence of trace amounts (<1%) of free paclitaxel. An animal study provided insight into the maximum tolerated dose (MTD) wherein 10, 25, 50, and 100 mg of paclitaxel/kg of construct or Abraxane were administered once per week for three consecutive weeks to non tumor bearing athymic nude mice. The construct showed in vivo toxicity comparable to that of Abraxane. Both formulations were found to be nontoxic at the administered doses, and the dendrimer had an acute MTD greater than the highest dose administered. In a prostate tumor model (PC-3-h-luc), efficacy was observed over 70 days with an arrest of tumor growth and lack of luciferase activity observed in the twice treated cohort.


Methods of Molecular Biology | 2011

Measuring the hydrodynamic size of nanoparticles in aqueous media using batch-mode dynamic light scattering.

Vincent A. Hackley; Jeffrey D. Clogston

Particle size characterization is of particular importance to nanomedicine. The size similarity of nanoparticles to biological moieties is believed to impart many of their unique medical properties. Here we present a method for sample preparation and the determination of mean nanoparticle size (hydrodynamic diameter) using batch-mode dynamic light scattering (DLS) in dilute aqueous suspensions. We then demonstrate this method for 30 nm colloidal gold.


Methods of Molecular Biology | 2011

Lipid Component Quantitation by Thin Layer Chromatography

Jeffrey D. Clogston; Anil K. Patri

This chapter describes a thin layer chromatography (TLC) method for the quantitation of various lipids (such as phospholipids, sphingolipids, acylglycerols, and fatty acids) in lipid-based nanoparticle formulations such as liposomes and nanoemulsions. We illustrate this technique to quantify C6-ceramide (N-hexanoyl-D: -erythro-sphingosine) in a nanoemulsion formulation. C6-ceramide is a powerful chemotherapeutic that is poorly soluble in aqueous buffers.


Methods of Molecular Biology | 2011

Characterization of Nanoparticles by Matrix Assisted Laser Desorption Ionization Time-of-Flight Mass Spectrometry

Uma Ramalinga; Jeffrey D. Clogston; Anil K. Patri; John T. Simpson

Determining the molecular weight of nanoparticles can be challenging. The molecular weight characterization of dendrimers, for example, with varying covalent and noncovalent modifications is critical to their use as therapeutics. As such, we describe in this chapter a protocol for the analysis of these molecules by matrix assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS).


Methods of Molecular Biology | 2011

Detecting and measuring free gadolinium in nanoparticles for MRI imaging.

Jeffrey D. Clogston; Anil K. Patri

This chapter describes a method for the measurement of free gadolinium in nanoparticle samples. Conjugation of a gadolinium-chelate to a nanoparticle allows the particles distribution to be imaged via magnetic resonance imaging (MRI). Free (unchelated) gadolinium is a known toxin, being a heavy metal, and may contribute towards total gadolinium concentration. Determining the amount of free gadolinium is therefore an important aspect of the preclinical characterization of gadolinium-chelate MRI imaging agent nanoparticles.

Collaboration


Dive into the Jeffrey D. Clogston's collaboration.

Top Co-Authors

Avatar

Anil K. Patri

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Stephan T. Stern

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Vincent A. Hackley

National Institute of Standards and Technology

View shared research outputs
Top Co-Authors

Avatar

Scott E. McNeil

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Barry W. Neun

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Marina A. Dobrovolskaia

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Pavan P. Adiseshaiah

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Jiwen Zheng

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Christopher B. McLeland

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

De-Hao Tsai

National Institute of Standards and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge