Network


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

Hotspot


Dive into the research topics where Brandon Ason is active.

Publication


Featured researches published by Brandon Ason.


Journal of Lipid Research | 2011

Improved efficacy for ezetimibe and rosuvastatin by attenuating the induction of PCSK9

Brandon Ason; Samnang Tep; Harry R. Davis; Yiming Xu; Glen Tetzloff; Beverly Galinski; Ferdie Soriano; Natalya Dubinina; Lei Zhu; Alice Stefanni; Kenny K. Wong; Marija Tadin-Strapps; Steven R. Bartz; Brian K. Hubbard; Mollie Ranalletta; Alan B. Sachs; Alison M. Strack; Nelly A. Kuklin

Reducing circulating LDL-cholesterol (LDL-c) reduces the risk of cardiovascular disease in people with hypercholesterolemia. Current approaches to reduce circulating LDL-c include statins, which inhibit cholesterol synthesis, and ezetimibe, which blocks cholesterol absorption. Both elevate serum PCSK9 protein levels in patients, which could attenuate their efficacy by reducing the amount of cholesterol cleared from circulation. To determine whether PCSK9 inhibition could enhance LDL-c lowering of both statins and ezetimibe, we utilized small interfering RNAs (siRNAs) to knock down Pcsk9, together with ezetimibe, rosuvastatin, and an ezetimibe/rosuvastatin combination in a mouse model with a human-like lipid profile. We found that ezetimibe, rosuvastatin, and ezetimibe/rosuvastatin combined lower serum cholesterol but induce the expression of Pcsk9 as well as the Srebp-2 hepatic cholesterol biosynthesis pathway. Pcsk9 knockdown in combination with either treatment led to greater reductions in serum non-HDL with a near-uniform reduction of all LDL-c subfractions. In addition to reducing serum cholesterol, the combined rosuvastatin/ezetimibe/Pcsk9 siRNA treatment exhibited a significant reduction in serum APOB protein and triglyceride levels. Taken together, these data provide evidence that PCSK9 inhibitors, in combination with current therapies, have the potential to achieve greater reductions in both serum cholesterol and triglycerides.


Journal of Lipid Research | 2011

siRNA-induced liver ApoB knockdown lowers serum LDL-cholesterol in a mouse model with human-like serum lipids

Marija Tadin-Strapps; Laurence B. Peterson; Anne-Marie Cumiskey; Ray Rosa; Vivienne Mendoza; Jose Castro-Perez; Oscar Puig; Liwen Zhang; Walter Strapps; Satyasri Yendluri; Lori Andrews; Victoria Pickering; Julie Rice; Lily Luo; Zhu Chen; Samnang Tep; Brandon Ason; Elizabeth Polizzi Somers; Alan B. Sachs; Steven R. Bartz; Jenny Tian; Jayne Chin; Brian K. Hubbard; Kenny K. Wong; Lyndon J. Mitnaul

Increased serum apolipoprotein (apo)B and associated LDL levels are well-correlated with an increased risk of coronary disease. ApoE–/– and low density lipoprotein receptor (LDLr)–/– mice have been extensively used for studies of coronary atherosclerosis. These animals show atherosclerotic lesions similar to those in humans, but their serum lipids are low in apoB-containing LDL particles. We describe the development of a new mouse model with a human-like lipid profile. Ldlr CETP+/– hemizygous mice carry a single copy of the human CETP transgene and a single copy of a LDL receptor mutation. To evaluate the apoB pathways in this mouse model, we used novel short-interfering RNAs (siRNA) formulated in lipid nanoparticles (LNP). ApoB siRNAs induced up to 95% reduction of liver ApoB mRNA and serum apoB protein, and a significant lowering of serum LDL in Ldlr CETP+/– mice. ApoB targeting is specific and dose-dependent, and it shows lipid-lowering effects for over three weeks. Although specific triglycerides (TG) were affected by ApoB mRNA knockdown (KD) and the total plasma lipid levels were decreased by 70%, the overall lipid distribution did not change. Results presented here demonstrate a new mouse model for investigating additional targets within the ApoB pathways using the siRNA modality.


Molecular Pharmacology | 2011

RNA-induced silencing complex-bound small interfering RNA is a determinant of RNA interference-mediated gene silencing in mice.

Jie Wei; Jeffrey P. Jones; Jing Kang; Ananda Card; Michael Krimm; Paula J. Hancock; Yi Pei; Brandon Ason; Elmer Payson; Natalya Dubinina; Mark T. Cancilla; Mark Stroh; Julja Burchard; Alan B. Sachs; Jerome Hochman; William Michael Flanagan; Nelly A. Kuklin

Deeper knowledge of pharmacokinetic and pharmacodynamic (PK/PD) concepts for RNA therapeutics is important to streamline the drug development process and for rigorous selection of best performing drug candidates. Here we characterized the PK/PD relationship for small interfering RNAs (siRNAs) targeting luciferase by examining siRNA concentration in plasma and liver, the temporal RNA-induced silencing complex binding profiles, mRNA reduction, and protein inhibition measured by noninvasive bioluminescent imaging. A dose-dependent and time-related decrease in bioluminescence was detected over 25 days after a single treatment of a lipid nanoparticle-formulated siRNA targeting luciferase messenger RNA. A direct relationship was observed between the degree of in vivo mRNA and protein reduction and the Argonaute2 (Ago2)-bound siRNA fraction but not with the total amount of siRNA found in the liver, suggesting that the Ago2-siRNA complex is the key determinant of target inhibition. These observations were confirmed for an additional siRNA that targets endogenously expressed Sjögren syndrome antigen B (Ssb) mRNA, indicating that our observations are not limited to a transgenic mouse system. Our data provide detailed information of the temporal regulation of siRNA liver delivery, Ago2 loading, mRNA reduction, and protein inhibition that are essential for the rapid and cost-effective clinical development of siRNAs therapeutics.


Journal of Lipid Research | 2014

PCSK9 inhibition fails to alter hepatic LDLR, circulating cholesterol, and atherosclerosis in the absence of ApoE.

Brandon Ason; José W.A. van der Hoorn; Joyce Chi Yee Chan; Edward Lee; Elsbet J. Pieterman; Kathy Nguyen; Mei Di; Susan Shetterly; Jie Tang; Wen-Chen Yeh; Margrit Schwarz; J. Wouter Jukema; Rob Scott; Scott M. Wasserman; Simon Jackson

LDL cholesterol (LDL-C) contributes to coronary heart disease. Proprotein convertase subtilisin/kexin type 9 (PCSK9) increases LDL-C by inhibiting LDL-C clearance. The therapeutic potential for PCSK9 inhibitors is highlighted by the fact that PCSK9 loss-of-function carriers exhibit 15–30% lower circulating LDL-C and a disproportionately lower risk (47–88%) of experiencing a cardiovascular event. Here, we utilized pcsk9−/− mice and an anti-PCSK9 antibody to study the role of the LDL receptor (LDLR) and ApoE in PCSK9-mediated regulation of plasma cholesterol and atherosclerotic lesion development. We found that circulating cholesterol and atherosclerotic lesions were minimally modified in pcsk9−/− mice on either an LDLR- or ApoE-deficient background. Acute administration of an anti-PCSK9 antibody did not reduce circulating cholesterol in an ApoE-deficient background, but did reduce circulating cholesterol (−45%) and TGs (−36%) in APOE*3Leiden.cholesteryl ester transfer protein (CETP) mice, which contain mouse ApoE, human mutant APOE3*Leiden, and a functional LDLR. Chronic anti-PCSK9 antibody treatment in APOE*3Leiden.CETP mice resulted in a significant reduction in atherosclerotic lesion area (−91%) and reduced lesion complexity. Taken together, these results indicate that both LDLR and ApoE are required for PCSK9 inhibitor-mediated reductions in atherosclerosis, as both are needed to increase hepatic LDLR expression.


Lipids | 2011

ApoB siRNA-induced Liver Steatosis is Resistant to Clearance by the Loss of Fatty Acid Transport Protein 5 (Fatp5)

Brandon Ason; Jose Castro-Perez; Samnang Tep; Alice Stefanni; Marija Tadin-Strapps; Thomas P. Roddy; Thomas Hankemeier; Brian K. Hubbard; Alan B. Sachs; Nelly A. Kuklin; Lyndon J. Mitnaul

The association between hypercholesterolemia and elevated serum apolipoprotein B (APOB) has generated interest in APOB as a therapeutic target for patients at risk of developing cardiovascular disease. In the clinic, mipomersen, an antisense oligonucleotide (ASO) APOB inhibitor, was associated with a trend toward increased hepatic triglycerides, and liver steatosis remains a concern. We found that siRNA-mediated knockdown of ApoB led to elevated hepatic triglycerides and liver steatosis in mice engineered to exhibit a human-like lipid profile. Many genes required for fatty acid synthesis were reduced, suggesting that the observed elevation in hepatic triglycerides is maintained by the cell through fatty acid uptake as opposed to fatty acid synthesis. Fatty acid transport protein 5 (Fatp5/Slc27a5) is required for long chain fatty acid (LCFA) uptake and bile acid reconjugation by the liver. Fatp5 knockout mice exhibited lower levels of hepatic triglycerides due to decreased fatty acid uptake, and shRNA-mediated knockdown of Fatp5 protected mice from diet-induced liver steatosis. Here, we evaluated if siRNA-mediated knockdown of Fatp5 was sufficient to alleviate ApoB knockdown-induced steatosis. We determined that, although Fatp5 siRNA treatment was sufficient to increase the proportion of unconjugated bile acids 100-fold, consistent with FATP5s role in bile acid reconjugation, Fatp5 knockdown failed to influence the degree, zonal distribution, or composition of the hepatic triglycerides that accumulated following ApoB siRNA treatment.


Journal of Pharmacology and Experimental Therapeutics | 2015

Anti-PCSK9 Antibody Pharmacokinetics and Low-Density Lipoprotein-Cholesterol Pharmacodynamics in Nonhuman Primates Are Antigen Affinity–Dependent and Exhibit Limited Sensitivity to Neonatal Fc Receptor–Binding Enhancement

Kirk Henne; Brandon Ason; Monique Howard; Wei Wang; Jeonghoon Sun; Jared Higbee; Jie Tang; Katherine Matsuda; Ren Xu; Lei Zhou; Joyce Chi Yee Chan; Chadwick Terence King; Derek E. Piper; Randal R. Ketchem; Mark Leo Michaels; Simon Jackson; Marc W. Retter

Proprotein convertase subtilisin/kexin type 9 (PCSK9) has emerged as an attractive therapeutic target for cardiovascular disease. Monoclonal antibodies (mAbs) that bind PCSK9 and prevent PCSK9:low-density lipoprotein receptor complex formation reduce serum low-density lipoprotein-cholesterol (LDL-C) in vivo. PCSK9-mediated lysosomal degradation of bound mAb, however, dramatically reduces mAb exposure and limits duration of effect. Administration of high-affinity mAb1:PCSK9 complex (1:2) to mice resulted in significantly lower mAb1 exposure compared with mAb1 dosed alone in normal mice or in PCSK9 knockout mice lacking antigen. To identify mAb-binding characteristics that minimize lysosomal disposition, the pharmacokinetic behavior of four mAbs representing a diverse range of PCSK9-binding affinities at neutral (serum) and acidic (endosomal) pH was evaluated in cynomolgus monkeys. Results revealed an inverse correlation between affinity and both mAb exposure and duration of LDL-C lowering. High-affinity mAb1 exhibited the lowest exposure and shortest duration of action (6 days), whereas mAb2 displayed prolonged exposure and LDL-C reduction (51 days) as a consequence of lower affinity and pH-sensitive PCSK9 binding. mAbs with shorter endosomal PCSK9:mAb complex dissociation half-lives (<20 seconds) produced optimal exposure-response profiles. Interestingly, incorporation of previously reported Fc-region amino acid substitutions or novel loop-insertion peptides that enhance in vitro neonatal Fc receptor binding, led to only modest pharmacokinetic improvements for mAbs with pH-dependent PCSK9 binding, with only limited augmentation of pharmacodynamic activity relative to native mAbs. A pivotal role for PCSK9 in mAb clearance was demonstrated, more broadly suggesting that therapeutic mAb-binding characteristics require optimization based on target pharmacology.


Journal of Proteome Research | 2011

Attenuation of Slc27a5 gene expression followed by LC-MS measurement of bile acid reconjugation using metabolomics and a stable isotope tracer strategy.

Jose Castro-Perez; Thomas P. Roddy; Vinit Shah; Sheng-Ping Wang; Xuesong Ouyang; Anthony Ogawa; David G. McLaren; Marija Tadin-Strapps; Michael Robinson; Steven R. Bartz; Brandon Ason; Ying Chen; Stephen F. Previs; Kenny K. Wong; Rob J. Vreeken; Douglas G. Johns; Brian K. Hubbard; Thomas Hankemeier; Lyndon J. Mitnaul


Archive | 2015

JLR 2014 Ason et al. No PCSK9-induced change in LDLR in E-ko

Brandon Ason; José W.A. van der Hoorn; Joyce Chi Yee Chan; Edward Lee; Elsbet J. Pieterman; Kathy Nguyen; Mei Di; Susan Shetterly; Jie Tang; Wen-Chen Yeh; Margrit Schwarz; J. Wouter Jukema; Robert W. Scott; Scott Wasserman; Simon Jackson


Atherosclerosis | 2015

Elucidating the mechanism for PCSK9 inhibition-mediated reduction in LP(A)

Matthew Peach; Ren Xu; Mei Di; S. Shetterly; Narimon Honarpour; Ransi Somaratne; Brandon Ason; Simon Jackson


Archive | 2014

JLR 2014 Ason et al.Suppl Data; no PCSK9-induced change in LDLR in E-ko

Brandon Ason; José W.A. van der Hoorn; Joyce Chi Yee Chan; Edward Lee; Elsbet J. Pieterman; Kathy Nguyen; Mei Di; Susan Shetterly; Jie Tang; Wen-Chen Yeh; Margrit Schwarz; J. Wouter Jukema; Robert W. Scott; Scott Wasserman; Simon Jackson

Collaboration


Dive into the Brandon Ason's collaboration.

Researchain Logo
Decentralizing Knowledge