Network


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

Hotspot


Dive into the research topics where Michael E. Østergaard is active.

Publication


Featured researches published by Michael E. Østergaard.


Chemical Society Reviews | 2011

Pyrene-functionalized oligonucleotides and locked nucleic acids (LNAs): tools for fundamental research, diagnostics, and nanotechnology.

Michael E. Østergaard; Patrick J. Hrdlicka

Pyrene-functionalized oligonucleotides (PFOs) are increasingly explored as tools in fundamental research, diagnostics and nanotechnology. Their popularity is linked to the ability of pyrenes to function as polarity-sensitive and quenchable fluorophores, excimer-generating units, aromatic stacking moieties and nucleic acid duplex intercalators. These characteristics have enabled development of PFOs for detection of complementary DNA/RNA targets, discrimination of single nucleotide polymorphisms (SNPs), and generation of π-arrays on nucleic acid scaffolds. This critical review will highlight the physical properties and applications of PFOs that are likely to provide high degree of positional control of the chromophore in nucleic acid complexes. Particular emphasis will be placed on pyrene-functionalized Locked Nucleic Acids (LNAs) since these materials display interesting properties such as fluorescence quantum yields approaching unity and recognition of mixed-sequence double stranded DNA (144 references).


Nucleic Acids Research | 2013

Rational design of antisense oligonucleotides targeting single nucleotide polymorphisms for potent and allele selective suppression of mutant Huntingtin in the CNS.

Michael E. Østergaard; Amber L. Southwell; Holly Kordasiewicz; Andrew T. Watt; Niels H. Skotte; Crystal N. Doty; Kuljeet Vaid; Erika B. Villanueva; Eric E. Swayze; C. Frank Bennett; Michael R. Hayden; Punit P. Seth

Autosomal dominant diseases such as Huntington’s disease (HD) are caused by a gain of function mutant protein and/or RNA. An ideal treatment for these diseases is to selectively suppress expression of the mutant allele while preserving expression of the wild-type variant. RNase H active antisense oligonucleotides (ASOs) or small interfering RNAs can achieve allele selective suppression of gene expression by targeting single nucleotide polymorphisms (SNPs) associated with the repeat expansion. ASOs have been previously shown to discriminate single nucleotide changes in targeted RNAs with ∼5-fold selectivity. Based on RNase H enzymology, we enhanced single nucleotide discrimination by positional incorporation of chemical modifications within the oligonucleotide to limit RNase H cleavage of the non-targeted transcript. The resulting oligonucleotides demonstrate >100-fold discrimination for a single nucleotide change at an SNP site in the disease causing huntingtin mRNA, in patient cells and in a completely humanized mouse model of HD. The modified ASOs were also well tolerated after injection into the central nervous system of wild-type animals, suggesting that their tolerability profile is suitable for advancement as potential allele-selective HD therapeutics. Our findings lay the foundation for efficient allele-selective downregulation of gene expression using ASOs—an outcome with broad application to HD and other dominant genetic disorders.


Molecular Therapy | 2014

In vivo evaluation of candidate allele-specific mutant huntingtin gene silencing antisense oligonucleotides.

Amber L. Southwell; Niels H. Skotte; Holly Kordasiewicz; Michael E. Østergaard; Andrew T. Watt; Jeffrey B. Carroll; Crystal N. Doty; Erika B. Villanueva; Eugenia Petoukhov; Kuljeet Vaid; Yuanyun Xie; Susan M. Freier; Eric E. Swayze; Punit P. Seth; C. Bennett; Michael R. Hayden

Huntington disease (HD) is a dominant, genetic neurodegenerative disease characterized by progressive loss of voluntary motor control, psychiatric disturbance, and cognitive decline, for which there is currently no disease-modifying therapy. HD is caused by the expansion of a CAG tract in the huntingtin (HTT) gene. The mutant HTT protein (muHTT) acquires toxic functions, and there is significant evidence that muHTT lowering would be therapeutically efficacious. However, the wild-type HTT protein (wtHTT) serves vital functions, making allele-specific muHTT lowering strategies potentially safer than nonselective strategies. CAG tract expansion is associated with single nucleotide polymorphisms (SNPs) that can be targeted by gene silencing reagents such as antisense oligonucleotides (ASOs) to accomplish allele-specific muHTT lowering. Here we evaluate ASOs targeted to HD-associated SNPs in acute in vivo studies including screening, distribution, duration of action and dosing, using a humanized mouse model of HD, Hu97/18, that is heterozygous for the targeted SNPs. We have identified four well-tolerated lead ASOs that potently and selectively silence muHTT at a broad range of doses throughout the central nervous system for 16 weeks or more after a single intracerebroventricular (ICV) injection. With further validation, these ASOs could provide a therapeutic option for individuals afflicted with HD.


PLOS ONE | 2014

Allele-Specific Suppression of Mutant Huntingtin Using Antisense Oligonucleotides: Providing a Therapeutic Option for All Huntington Disease Patients

Niels H. Skotte; Amber L. Southwell; Michael E. Østergaard; Jeffrey B. Carroll; Simon C. Warby; Crystal N. Doty; Eugenia Petoukhov; Kuljeet Vaid; Holly Kordasiewicz; Andrew T. Watt; Susan M. Freier; Gene Hung; Punit P. Seth; C. Frank Bennett; Eric E. Swayze; Michael R. Hayden

Huntington disease (HD) is an inherited, fatal neurodegenerative disorder caused by a CAG repeat expansion in the huntingtin gene. The mutant protein causes neuronal dysfunction and degeneration resulting in motor dysfunction, cognitive decline, and psychiatric disturbances. Currently, there is no disease altering treatment, and symptomatic therapy has limited benefit. The pathogenesis of HD is complicated and multiple pathways are compromised. Addressing the problem at its genetic root by suppressing mutant huntingtin expression is a promising therapeutic strategy for HD. We have developed and evaluated antisense oligonucleotides (ASOs) targeting single nucleotide polymorphisms that are significantly enriched on HD alleles (HD-SNPs). We describe our structure-activity relationship studies for ASO design and find that adjusting the SNP position within the gap, chemical modifications of the wings, and shortening the unmodified gap are critical for potent, specific, and well tolerated silencing of mutant huntingtin. Finally, we show that using two distinct ASO drugs targeting the two allelic variants of an HD-SNP could provide a therapeutic option for all persons with HD; allele-specifically for roughly half, and non-specifically for the remainder.


Journal of Medicinal Chemistry | 2016

Comprehensive Structure-Activity Relationship of Triantennary N-Acetylgalactosamine Conjugated Antisense Oligonucleotides for Targeted Delivery to Hepatocytes.

Thazha P. Prakash; Jinghua Yu; Michael T. Migawa; Garth A. Kinberger; W. Brad Wan; Michael E. Østergaard; Recaldo L. Carty; Guillermo Vasquez; Audrey Low; Alfred Chappell; Karsten Schmidt; Mariam Aghajan; Jeff Crosby; Heather M. Murray; Sheri L. Booten; Jill Hsiao; Armand Soriano; Todd Machemer; Patrick Cauntay; Sebastien A. Burel; Susan F. Murray; Hans Gaus; Mark J. Graham; Eric E. Swayze; Punit P. Seth

The comprehensive structure-activity relationships of triantennary GalNAc conjugated ASOs for enhancing potency via ASGR mediated delivery to hepatocytes is reported. Seventeen GalNAc clusters were assembled from six distinct scaffolds and attached to ASOs. The resulting ASO conjugates were evaluated in ASGR binding assays, in primary hepatocytes, and in mice. Five structurally distinct GalNAc clusters were chosen for more extensive evaluation using ASOs targeting SRB-1, A1AT, FXI, TTR, and ApoC III mRNAs. GalNAc-ASO conjugates exhibited excellent potencies (ED50 0.5-2 mg/kg) for reducing the targeted mRNAs and proteins. This work culminated in the identification of a simplified tris-based GalNAc cluster (THA-GN3), which can be efficiently assembled using readily available starting materials and conjugated to ASOs using a solution phase conjugation strategy. GalNAc-ASO conjugates thus represent a viable approach for enhancing potency of ASO drugs in the clinic without adding significant complexity or cost to existing protocols for manufacturing oligonucleotide drugs.


Nucleic Acids Research | 2016

Stabilin-1 and Stabilin-2 are specific receptors for the cellular internalization of phosphorothioate-modified antisense oligonucleotides (ASOs) in the liver

Colton M. Miller; Aaron J. Donner; Emma E. Blank; Andrew W. Egger; Brianna M. Kellar; Michael E. Østergaard; Punit P. Seth; Edward N. Harris

Phosphorothioate (PS)-modified antisense oligonucleotides (ASOs) have been extensively investigated over the past three decades as pharmacological and therapeutic agents. One second generation ASO, Kynamro™, was recently approved by the FDA for the treatment of homozygous familial hypercholesterolemia and over 35 second generation PS ASOs are at various stages of clinical development. In this report, we show that the Stabilin class of scavenger receptors, which were not previously thought to bind DNA, do bind and internalize PS ASOs. With the use of primary cells from mouse and rat livers and recombinant cell lines each expressing Stabilin-1 and each isoform of Stabilin-2 (315-HARE and 190-HARE), we have determined that PS ASOs bind with high affinity and these receptors are responsible for bulk, clathrin-mediated endocytosis within the cell. Binding is primarily dependent on salt-bridge formation and correct folding of the intact protein receptor. Increased internalization rates also enhanced ASO potency for reducing expression of the non-coding RNA Malat-1, in Stabilin-expressing cell lines. A more thorough understanding of mechanisms by which ASOs are internalized in cells and their intracellular trafficking pathways will aid in the design of next generation antisense agents with improved therapeutic properties.


Journal of Organic Chemistry | 2014

Synthesis and Biophysical Properties of C5-Functionalized LNA (Locked Nucleic Acid)

Pawan Kumar; Michael E. Østergaard; Bharat Baral; Brooke A. Anderson; Dale C. Guenther; Mamta Kaura; Daniel J. Raible; Pawan K. Sharma; Patrick J. Hrdlicka

Oligonucleotides modified with conformationally restricted nucleotides such as locked nucleic acid (LNA) monomers are used extensively in molecular biology and medicinal chemistry to modulate gene expression at the RNA level. Major efforts have been devoted to the design of LNA derivatives that induce even higher binding affinity and specificity, greater enzymatic stability, and more desirable pharmacokinetic profiles. Most of this work has focused on modifications of LNA’s oxymethylene bridge. Here, we describe an alternative approach for modulation of the properties of LNA: i.e., through functionalization of LNA nucleobases. Twelve structurally diverse C5-functionalized LNA uridine (U) phosphoramidites were synthesized and incorporated into oligodeoxyribonucleotides (ONs), which were then characterized with respect to thermal denaturation, enzymatic stability, and fluorescence properties. ONs modified with monomers that are conjugated to small alkynes display significantly improved target affinity, binding specificity, and protection against 3′-exonucleases relative to regular LNA. In contrast, ONs modified with monomers that are conjugated to bulky hydrophobic alkynes display lower target affinity yet much greater 3′-exonuclease resistance. ONs modified with C5-fluorophore-functionalized LNA-U monomers enable fluorescent discrimination of targets with single nucleotide polymorphisms (SNPs). In concert, these properties render C5-functionalized LNA as a promising class of building blocks for RNA-targeting applications and nucleic acid diagnostics.


Angewandte Chemie | 2012

Structure‐Based Design of a Highly Constrained Nucleic Acid Analogue: Improved Duplex Stabilization by Restricting Sugar Pucker and Torsion Angle γ

Benjamin R. Schroeder; Robert D. Giacometti; Bradley L. Merner; Michael E. Østergaard; Eric E. Swayze; Punit P. Seth

Dual conformational restriction: a new, highly constrained modification of the α-L-locked nucleic acid (α-L-LNA) scaffold that locks the sugar furanose ring in an N-type configuration and also restricts rotation around torsion angle γ was synthesized. This new modification increases the thermostability of an oligonucleotide duplex compared to using a single mode of constraint alone.


ChemBioChem | 2009

C5‐Functionalized LNA: Unparalleled Hybridization Properties and Enzymatic Stability

Michael E. Østergaard; Pawan Kumar; Bharat Baral; Daniel J. Raible; T. Santhosh Kumar; Brooke A. Anderson; Dale C. Guenther; Lee A. Deobald; Andrzej Paszczynski; Pawan K. Sharma; Patrick J. Hrdlicka

Antisense oligonucleotides (ONs) are widely explored as fundamental research tools and therapeutic agents against diseases of genetic origin due to their ability to modulate gene expression by interfering with target RNA. Introduction of chemically modified nucleotides into antisense ONs is crucial to increase binding affinity toward RNA targets, improve discrimination of mismatched RNA to avoid off-target effects, and enhance stability against nucleases to slow down degradation. The use of conformationally restricted nucleotides and locked nucleic acids (LNAs, Scheme 1) in particular, has to some extent addressed these challenges. Antisense LNAs are accordingly evaluated in several clinical trials. Substantial efforts have been invested to develop LNA analogues with even more desirable biophysical properties and reduced hepatotoxicity. These studies have primarily focused on modification of the oxymethylene bridge spanning the C2’and C4’-positions and/or introduction of minor-groove-oriented substituents into the bridge. Improved enzymatic stability, e, f, j] altered biodistribution, or reduced hepatotoxicity has been reported for some of the analogues, but improvements in hybridization properties relative to LNA were generally not observed. Results from comparative in vivo antisense studies must be awaited to assess if the significantly increased synthetic complexity of these conformationally restricted nucleotides is justified. C5-functionalized pyrimidine DNA building blocks have attracted considerable attention due to their ability to accommodate functional entities in the major groove of nucleic acid duplexes and straightforward synthesis. Small C5-entities are generally well tolerated in duplexes and result in small increases in thermal affinity toward DNA/RNA complements. f] In light of this, we hypothesized that C5-alkynyl-functionalized LNA monomers would synergistically integrate beneficial Scheme 1. Synthetic outline of phosphoramidites 5 W–5 Z. CAN = ceric ammonium nitrate, DMTr = 4,4’-dimethoxytrityl, TBAF = tetrabutylammonium fluoride, PCl = 2-cyanoethyl N, N’diisopropylchlorophosphoramidite.


Chemical Communications | 2009

Optimized DNA-targeting using triplex forming C5-alkynyl functionalized LNA

Sujay P. Sau; Pawan Kumar; Brooke A. Anderson; Michael E. Østergaard; Lee A. Deobald; Andrzej Paszczynski; Pawan K. Sharma; Patrick J. Hrdlicka

Triplex forming oligonucleotides (TFOs) modified with C5-alkynyl functionalized LNA (locked nucleic acid) monomers display extraordinary thermal affinity toward double stranded DNA targets, excellent discrimination of Hoogsteen-mismatched targets, and high stability against 3?-exonucleases.

Collaboration


Dive into the Michael E. Østergaard's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pawan Kumar

Kurukshetra University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amber L. Southwell

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Michael R. Hayden

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Crystal N. Doty

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge