Network


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

Hotspot


Dive into the research topics where Bonnie J. Howell is active.

Publication


Featured researches published by Bonnie J. Howell.


The Journal of Neuroscience | 2014

A Sensitive Aβ Oligomer Assay Discriminates Alzheimer's and Aged Control Cerebrospinal Fluid

Mary J. Savage; Juliya Kalinina; Abigail Wolfe; Katherine Tugusheva; Rachel Korn; Tanesha Cash-Mason; Jill Maxwell; Nathan G. Hatcher; Sharie J. Haugabook; Guoxin Wu; Bonnie J. Howell; John J. Renger; Paul J. Shughrue; Alexander Mccampbell

A hallmark of Alzheimers disease (AD) brain is the amyloid β (Aβ) plaque, which is comprised of Aβ peptides. Multiple lines of evidence suggest that Aβ oligomers are more toxic than other peptide forms. We sought to develop a robust assay to quantify oligomers from CSF. Antibody 19.3 was compared in one-site and competitive ELISAs for oligomer binding specificity. A two-site ELISA for oligomers was developed using 19.3 coupled to a sensitive, bead-based fluorescent platform able to detect single photons of emitted light. The two-site ELISA was >2500× selective for Aβ oligomers over Aβ monomers with a limit of detection ∼0.09 pg/ml in human CSF. The lower limit of reliable quantification of the assay was 0.18 pg/ml and the antibody pairs recognized Aβ multimers comprised of either synthetic standards, or endogenous oligomers isolated from confirmed human AD and healthy control brain. Using the assay, a significant 3- to 5-fold increase in Aβ oligomers in human AD CSF compared with comparably aged controls was demonstrated. The increase was seen in three separate human cohorts, totaling 63 AD and 54 controls. CSF oligomers ranged between 0.1 and 10 pg/ml. Aβ oligomer levels did not strongly associate with age or gender, but had an inverse correlation with MMSE score. The C statistic for the Aβ oligomer ROC curve was 0.86, with 80% sensitivity and 88% specificity to detect AD, suggesting reasonable discriminatory power for the AD state and the potential for utility as a diagnostic marker.


Journal of Histochemistry and Cytochemistry | 2011

Biodistribution of small interfering RNA at the organ and cellular levels after lipid nanoparticle-mediated delivery.

Bin Shi; Ed Keough; Andrea Matter; Karen R. Leander; Stephanie Young; Ed Carlini; Alan B. Sachs; Weikang Tao; Marc T. Abrams; Bonnie J. Howell; Laura Sepp-Lorenzino

Chemically stabilized small interfering RNA (siRNA) can be delivered systemically by intravenous injection of lipid nanoparticles (LNPs) in rodents and primates. The biodistribution and kinetics of LNP–siRNA delivery in mice at organ and cellular resolution have been studied using immunofluorescence (IF) staining and quantitative polymerase chain reaction (qPCR). At 0.5 and 2 hr post tail vein injection of Cy5-labeled siRNA encapsulated in LNP, the organ rank-order of siRNA levels is liver > spleen > kidney, with only negligible accumulation in duodenum, lung, heart, and brain. Similar conclusions were drawn by using qPCR to measure tissue siRNA levels as a secondary end point. siRNA levels in these tissues decreased by more than 10-fold after 24 hr. Within the liver, LNPs delivered siRNA to hepatocytes, Kupffer cells, and sinusoids in a time-dependent manner, as revealed by IF staining and signal quantitation methods established using OPERA/Columbus software. siRNA first accumulated in liver sinusoids and trafficked to hepatocytes by 2 hr post dose, corresponding to the onset of target mRNA silencing. Fluorescence in situ hybridization methods were used to detect both strands of siRNA in fixed tissues. Collectively, the authors have implemented a platform to evaluate biodistribution of siRNA across cell types and across tissues in vivo, with the objective of elucidating the pharmacokinetic and pharmacodynamic relationship to guide optimization of delivery vehicles.


Cancer Biology & Therapy | 2010

An allosteric Akt inhibitor effectively blocks Akt signaling and tumor growth with only transient effects on glucose and insulin levels in vivo

Craig Cherrin; Kathleen M. Haskell; Bonnie J. Howell; Raymond E. Jones; Karen R. Leander; Ronald G. Robinson; Aubrey Watkins; Mark T. Bilodeau; Jacob M. Hoffman; Philip E. Sanderson; George D. Hartman; Elizabeth Mahan; Thomayant Prueksaritanont; Guoqiang Jiang; Qing-Bai She; Neal Rosen; Laura Sepp-Lorenzino; Deborah Defeo-Jones; Hans E. Huber

The PI3K-Akt pathway is dysregulated in the majority of solid tumors. Pharmacological inhibition of Akt is a promising strategy for treating tumors resistant to growth factor receptor antagonists due to mutations in PI3K or PTEN. We have developed allosteric, isozyme-specific inhibitors of Akt activity and activation, as well as ex vivo kinase assays to measure inhibition of individual Akt isozymes in tissues. Here we describe the relationship between PK, Akt inhibition, hyperglycemia and tumor efficacy for a selective inhibitor of Akt1 and Akt2 (AKTi). In nude mice, AKTi treatment caused transient insulin resistance and reversible, dose-dependent hyperglycemia and hyperinsulinemia. Akt1 and Akt2 phosphorylation was inhibited in mouse lung with EC50 values of 1.6 and 7 μM, respectively, and with similar potency in other tissues and xenograft tumors. Weekly subcutaneous dosing of AKTi resulted in dose-dependent inhibition of LNCaP prostate cancer xenografts, an AR-dependent tumor with PTEN deletion and constitutively activated Akt. Complete tumor growth inhibition was achieved at 200 mpk, a dose that maintained inhibition of Akt1 and Akt2 of greater than 80% and 50%, respectively, for at least 12 hours in xenograft tumor and mouse lung. Hyperglycemia could be controlled by reducing Cmax, while maintaining efficacy in the LNCaP model, but not by insulin administration. AKTi treatment was well tolerated, without weight loss or gross toxicities. These studies supported the rationale for clinical development of allosteric Akt inhibitors and provide the basis for further refining of pharmacokinetic properties and dosing regimens of this class of inhibitors.


International Journal of Pharmaceutics | 2011

Analysis of lipid nanoparticles by Cryo-EM for characterizing siRNA delivery vehicles

Randy Crawford; Belma Dogdas; Edward Keough; R. Matthew Haas; Wickliffe Wepukhulu; Steven Krotzer; Paul A. Burke; Laura Sepp-Lorenzino; Ansuman Bagchi; Bonnie J. Howell

Lipid nanoparticles are self-assembling, dynamic structures commonly used as carriers of siRNA, DNA, and small molecular therapeutics. Quantitative analysis of particle characteristics such as morphological features can be very informative as biophysical properties are known to influence biological activity, biodistribution, and toxicity. However, accurate characterization of particle attributes and population distributions is difficult. Cryo-Electron Microscopy (Cryo-EM) is a leading characterization method and can reveal diversity in particle size, shape and lamellarity, however, this approach is traditionally used for qualitative review or low throughput image analysis due to inherent EM micrograph contrast characteristics and artifacts in the images which limit extraction of quantitative feature values. In this paper we describe the development of a semiautomatic image analysis framework to facilitate reliable image enhancement, object segmentation, and quantification of nanoparticle attributes in Cryo-EM micrographs. We apply this approach to characterize two formulations of siRNA-loaded lipid nanoparticles composed of cationic lipid, cholesterol, and poly(ethylene glycol)-lipid, where the formulations differ only by input component ratios. We found Cryo-EM image analysis provided reliable size and morphology information as well as the detection of smaller particle populations that were not detected by standard dynamic light scattering (DLS) analysis.


Bioconjugate Chemistry | 2013

Endosomolytic Bioreducible Poly(amido amine disulfide) Polymer Conjugates for the in Vivo Systemic Delivery of siRNA Therapeutics

Rubina Parmar; Marina Busuek; Eileen S. Walsh; Karen R. Leander; Bonnie J. Howell; Laura Sepp-Lorenzino; Eric Kemp; Louis S. Crocker; Anthony Leone; Christopher J. Kochansky; Brian A. Carr; Robert M. Garbaccio; Steven L. Colletti; Weimin Wang

Efficient siRNA delivery is dependent not only on the ability of the delivery vehicle to target a specific organ but also on its ability to enable siRNA entry into the cytoplasm of the target cells. Polymers with endosomolytic properties are increasingly being used as siRNA delivery vehicles due to their potential to facilitate endosomal escape and intracellular delivery. Addition of disulfide bonds in the backbone of these polymers was expected to provide degradability through reduction by glutathione in cytosol. This paper describes the synthesis of new endosomolytic bioreducible poly(amido amine disulfide) polymers whose lytic potential can be masked at physiological pH, but can be restored at acidic endosomal pH. These polymer conjugates gave good in vitro knockdown (KD) and did not demonstrate cytotoxicity in a MTS assay. Efficient mRNA KD for apolipoprotein B in mouse liver was observed with these polyconjugates following intravenous dosing.


Journal of Controlled Release | 2014

Development of a liver-targeted siRNA delivery platform with a broad therapeutic window utilizing biodegradable polypeptide-based polymer conjugates.

Stephanie E. Barrett; Rob Burke; Marc T. Abrams; Carol Bason; Marina Busuek; Edward Carlini; Brian A. Carr; Louis S. Crocker; Haihong Fan; Robert M. Garbaccio; Erin N. Guidry; Jun H. Heo; Bonnie J. Howell; Eric Kemp; Robert A. Kowtoniuk; Andrew H. Latham; Anthony Leone; Michael Lyman; Rubina G. Parmar; Mihir Patel; Sergey Pechenov; Tao Pei; Nicole T. Pudvah; Conrad E. Raab; Sean Riley; Laura Sepp-Lorenzino; Sheri Smith; Eric Soli; Steven J. Staskiewicz; Melissa Stern

The greatest challenge standing in the way of effective in vivo siRNA delivery is creating a delivery vehicle that mediates a high degree of efficacy with a broad therapeutic window. Key structure-activity relationships of a poly(amide) polymer conjugate siRNA delivery platform were explored to discover the optimized polymer parameters that yield the highest activity of mRNA knockdown in the liver. At the same time, the poly(amide) backbone of the polymers allowed for the metabolism and clearance of the polymer from the body very quickly, which was established using radiolabeled polymers to demonstrate the time course of biodistribution and excretion from the body. The fast degradation and clearance of the polymers provided for very low toxicity at efficacious doses, and the therapeutic window of this poly(amide)-based siRNA delivery platform was shown to be much broader than a comparable polymer platform. The results of this work illustrate that the poly(amide) platform has a promising future in the development of a siRNA-based drug approved for human use.


Journal of Antimicrobial Chemotherapy | 2016

A long-acting formulation of the integrase inhibitor raltegravir protects humanized BLT mice from repeated high-dose vaginal HIV challenges

Martina Kovarova; Michael D. Swanson; Rosa I. Sanchez; Caroline E. Baker; Justin Steve; Rae Ann Spagnuolo; Bonnie J. Howell; Daria J. Hazuda; J. Victor Garcia

OBJECTIVES Pre-exposure prophylaxis (PrEP) using antiretroviral drugs (ARVs) has been shown to reduce HIV transmission in people at high risk of HIV infection. Adherence to PrEP strongly correlates with the level of HIV protection. Long-acting injectable ARVs provide sustained systemic drug exposures over many weeks and can improve adherence due to infrequent parenteral administration. Here, we evaluated a new long-acting formulation of raltegravir for prevention of vaginal HIV transmission. METHODS Long-acting raltegravir was administered subcutaneously to BALB/c, NSG (NOD-scid-gamma) and humanized BLT (bone marrow-liver-thymus) mice and rhesus macaques. Raltegravir concentration in peripheral blood and tissue was analysed. Suppression of HIV replication was assessed in infected BLT mice. Two high-dose HIV vaginal challenges were used to evaluate protection from HIV transmission in BLT mice. RESULTS Two weeks after a single subcutaneous injection of long-acting raltegravir in BLT mice (7.5 mg) and rhesus macaques (160 mg), the plasma concentration of raltegravir was comparable to 400 mg orally, twice daily in humans. Serum collected from mice 3 weeks post-administration of long-acting raltegravir efficiently blocked HIV infection of TZM-bl indicator cells in vitro. Administration of long-acting raltegravir suppressed viral RNA in plasma and cervico-vaginal fluids of infected BLT mice, demonstrating penetration of active raltegravir into the female reproductive tract. Using transmitted/founder HIV we observed that BLT mice administered a single subcutaneous dose of long-acting raltegravir were protected from two high-dose HIV vaginal challenges 1 week and 4 weeks after drug administration. CONCLUSIONS These preclinical results demonstrated the efficacy of long-acting raltegravir in preventing vaginal HIV transmission.


Retrovirology | 2016

In vivo analysis of the effect of panobinostat on cell-associated HIV RNA and DNA levels and latent HIV infection

Perry Tsai; Guoxin Wu; Caroline E. Baker; William O. Thayer; Rae Ann Spagnuolo; Rosa I. Sanchez; Stephanie E. Barrett; Bonnie J. Howell; David J. Margolis; Daria J. Hazuda; Nancie M. Archin; J. Victor Garcia

BackgroundThe latent reservoir in resting CD4+ T cells presents a major barrier to HIV cure. Latency-reversing agents are therefore being developed with the ultimate goal of disrupting the latent state, resulting in induction of HIV expression and clearance of infected cells. Histone deacetylase inhibitors (HDACi) have received a significant amount of attention for their potential as latency-reversing agents.ResultsHere, we have investigated the in vitro and systemic in vivo effect of panobinostat, a clinically relevant HDACi, on HIV latency. We showed that panobinostat induces histone acetylation in human PBMCs. Further, we showed that panobinostat induced HIV RNA expression and allowed the outgrowth of replication-competent virus ex vivo from resting CD4+ T cells of HIV-infected patients on suppressive antiretroviral therapy (ART). Next, we demonstrated that panobinostat induced systemic histone acetylation in vivo in the tissues of BLT humanized mice. Finally, in HIV-infected, ART-suppressed BLT mice, we evaluated the effect of panobinostat on systemic cell-associated HIV RNA and DNA levels and the total frequency of latently infected resting CD4+ T cells. Our data indicate that panobinostat treatment resulted in systemic increases in cellular levels of histone acetylation, a key biomarker for in vivo activity. However, panobinostat did not affect the levels of cell-associated HIV RNA, HIV DNA, or latently infected resting CD4+ T cells.ConclusionWe have demonstrated robust levels of systemic histone acetylation after panobinostat treatment of BLT humanized mice; and we did not observe a detectable change in the levels of cell-associated HIV RNA, HIV DNA, or latently infected resting CD4+ T cells in HIV-infected, ART-suppressed BLT mice. These results are consistent with the modest effects noted in vitro and suggest that combination therapies may be necessary to reverse latency and enable clearance. Animal models will contribute to the progress towards an HIV cure.


Bioconjugate Chemistry | 2014

Improving the In Vivo Therapeutic Index of siRNA Polymer Conjugates through Increasing pH Responsiveness

Erin N. Guidry; Julie Farand; Arash Soheili; Craig A. Parish; Nancy J. Kevin; Brenda Pipik; Kathleen Calati; Nori Ikemoto; Jacob H. Waldman; Andrew H. Latham; Bonnie J. Howell; Anthony Leone; Robert M. Garbaccio; Stephanie E. Barrett; Rubina Parmar; Quang T. Truong; Bing Mao; Ian W. Davies; Steven L. Colletti; Laura Sepp-Lorenzino

Polymer based carriers that aid in endosomal escape have proven to be efficacious siRNA delivery agents in vitro and in vivo; however, most suffer from cytotoxicity due in part to a lack of selectivity for endosomal versus cell membrane lysis. For polymer based carriers to move beyond the laboratory and into the clinic, it is critical to find carriers that are not only efficacious, but also have margins that are clinically relevant. In this paper we report three distinct categories of polymer conjugates that improve the selectivity of endosomal membrane lysis by relying on the change in pH associated with endosomal trafficking, including incorporation of low pKa heterocycles, acid cleavable amino side chains, or carboxylic acid pH sensitive charge switches. Additionally, we determine the therapeutic index of our polymer conjugates in vivo and demonstrate that the incorporation of pH responsive elements dramatically expands the therapeutic index to 10-15, beyond that of the therapeutic index (less than 3), for polymer conjugates previously reported.


Methods in Enzymology | 2006

Cell imaging assays for G protein-coupled receptor internalization: application to high-throughput screening.

Seungtaek Lee; Bonnie J. Howell; Priya Kunapuli

There are a number of assays currently available to study G protein-coupled receptors (GPCRs), including ligand binding and functional assays. The latter category, albeit more complex, offers some obvious advantages over traditional ligand-binding assays. Functional cell-based assays typically include second messenger and reporter gene assays, which depend directly or indirectly on the cellular signaling cascade initiated upon receptor activation, respectively. More recently, cell imaging assays monitoring receptor trafficking are becoming increasingly popular. These assays, described in greater detail in this chapter, are independent of receptor signaling and are thus ideally suited for orphan receptors. In addition, these assays provide a valuable measure of receptor desensitization, an important feature for the use of GPCR agonists as potential therapeutic agents. The most popular GPCR imaging assays are based on the principles of receptor desensitization and internalization monitored directly or indirectly by green fluorescent protein.

Collaboration


Dive into the Bonnie J. Howell's collaboration.

Researchain Logo
Decentralizing Knowledge