Network


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

Hotspot


Dive into the research topics where Andrew D. Kroeker is active.

Publication


Featured researches published by Andrew D. Kroeker.


Antimicrobial Agents and Chemotherapy | 2006

VP35 Knockdown Inhibits Ebola Virus Amplification and Protects against Lethal Infection in Mice

Sven Enterlein; Kelly L. Warfield; Dana L. Swenson; David A. Stein; Jeffery L. Smith; C. Scott Gamble; Andrew D. Kroeker; Patrick L. Iversen; Sina Bavari; Elke Mühlberger

ABSTRACT Phosphorodiamidate morpholino oligomers (PMO) are a class of uncharged single-stranded DNA analogs modified such that each subunit includes a phosphorodiamidate linkage and morpholine ring. PMO antisense agents have been reported to effectively interfere with the replication of several positive-strand RNA viruses in cell culture. The filoviruses, Marburg virus and Ebola virus (EBOV), are negative-strand RNA viruses that cause up to 90% lethality in human outbreaks. There is currently no commercially available vaccine or efficacious therapeutic for any filovirus. In this study, PMO conjugated to arginine-rich cell-penetrating peptide (P-PMO) and nonconjugated PMO were assayed for the ability to inhibit EBOV infection in cell culture and in a mouse model of lethal EBOV infection. A 22-mer P-PMO designed to base pair with the translation start site region of EBOV VP35 positive-sense RNA generated sequence-specific and time- and dose-dependent inhibition of EBOV amplification in cell culture. The same oligomer provided complete protection to mice when administered before or after an otherwise lethal infection of EBOV. A corresponding nonconjugated PMO, as well as nonconjugated truncated versions of 16 and 19 base residues, provided length-dependent protection to mice when administered prophylactically. Together, these data suggest that antisense PMO and P-PMO have the potential to control EBOV infection and are promising therapeutic candidates.


Journal of Virology | 2005

Inhibition of Dengue Virus Serotypes 1 to 4 in Vero Cell Cultures with Morpholino Oligomers

Richard M. Kinney; Claire Y.-H. Huang; Becky C. Rose; Andrew D. Kroeker; Theo W. Dreher; Patrick L. Iversen; David A. Stein

ABSTRACT Five dengue (DEN) virus-specific R5F2R4 peptide-conjugated phosphorodiamidate morpholino oligomers (P4-PMOs) were evaluated for their ability to inhibit replication of DEN virus serotype 2 (DEN-2 virus) in mammalian cell culture. Initial growth curves of DEN-2 virus 16681 were obtained in Vero cells incubated with 20 μM P4-PMO compounds. At 6 days after infection, a P4-PMO targeting the 3′-terminal nucleotides of the DEN-2 virus genome and a random-sequence P4-PMO showed relatively little suppression of DEN-2 virus titer (0.1 and 0.9 log10, respectively). P4-PMOs targeting the AUG translation start site region of the single open reading frame and the 5′ cyclization sequence region had moderate activity, generating 1.6- and 1.8-log10 reductions. Two P4-PMO compounds, 5′SL and 3′CS (targeting the 5′-terminal nucleotides and the 3′ cyclization sequence region, respectively), were highly efficacious, each reducing the viral titer by greater than 5.7 log10 compared to controls at 6 days after infection with DEN-2 virus. Further experiments showed that 5′SL and 3′CS inhibited DEN-2 virus replication in a dose-dependent and sequence-specific manner. Treatment with 10 μM 3′CS reduced the titers of all four DEN virus serotypes, i.e., DEN-1 (strain 16007), DEN-2 (16681), DEN-3 (16562), and DEN-4 (1036) viruses by over 4 log10, in most cases to below detectable limits. The extent of 3′CS efficacy was affected by the timing of compound application in relation to viral infection of the cells. The 5′SL and 3′CS P4-PMOs did not suppress the replication of West Nile virus NY99 in Vero cells. These data indicate that further evaluation of the 5′SL and 3′CS compounds as potential DEN virus therapeutics is warranted.


Antimicrobial Agents and Chemotherapy | 2003

Inhibition of Gene Expression in Escherichia coli by Antisense Phosphorodiamidate Morpholino Oligomers

B. L. Geller; J. D. Deere; David A. Stein; Andrew D. Kroeker; Hong M. Moulton; Patrick L. Iversen

ABSTRACT Antisense phosphorodiamidate morpholino oligomers (PMOs) were tested for the ability to inhibit gene expression in Escherichia coli. PMOs targeted to either a myc-luciferase reporter gene product or 16S rRNA did not inhibit luciferase expression or growth. However, in a strain with defective lipopolysaccharide (lpxA mutant), which has a leaky outer membrane, PMOs targeted to the myc-luciferase or acyl carrier protein (acpP) mRNA significantly inhibited their targets in a dose-dependent response. A significant improvement was made by covalently joining the peptide (KFF)3KC to the end of PMOs. In strains with an intact outer membrane, (KFF)3KC-myc PMO inhibited luciferase expression by 63%. A second (KFF)3KC-PMO conjugate targeted to lacI mRNA induced β-galactosidase in a dose-dependent response. The end of the PMO to which (KFF)3KC is attached affected the efficiency of target inhibition but in various ways depending on the PMO. Another peptide-lacI PMO conjugate was synthesized with the cationic peptide CRRRQRRKKR and was found not to induce β-galactosidase. We conclude that the outer membrane of E. coli inhibits entry of PMOs and that (KFF)3KC-PMO conjugates are transported across both membranes and specifically inhibit expression of their genetic targets.


Journal of Virology | 2005

Inhibition, Escape, and Attenuated Growth of Severe Acute Respiratory Syndrome Coronavirus Treated with Antisense Morpholino Oligomers

Benjamin W. Neuman; David A. Stein; Andrew D. Kroeker; Michael J. Churchill; Alice M. Kim; Peter Kuhn; Philip E. Dawson; Hong M. Moulton; Richard K. Bestwick; Patrick L. Iversen; Michael J. Buchmeier

ABSTRACT The recently emerged severe acute respiratory syndrome coronavirus (SARS-CoV) is a potent pathogen of humans and is capable of rapid global spread. Peptide-conjugated antisense morpholino oligomers (P-PMO) were designed to bind by base pairing to specific sequences in the SARS-CoV (Tor2 strain) genome. The P-PMO were tested for their capacity to inhibit production of infectious virus as well as to probe the function of conserved viral RNA motifs and secondary structures. Several virus-targeted P-PMO and a random-sequence control P-PMO showed low inhibitory activity against SARS coronavirus. Certain other virus-targeted P-PMO reduced virus-induced cytopathology and cell-to-cell spread as a consequence of decreasing viral amplification. Active P-PMO were effective when administered at any time prior to peak viral synthesis and exerted sustained antiviral effects while present in culture medium. P-PMO showed low nonspecific inhibitory activity against translation of nontargeted RNA or growth of the arenavirus lymphocytic choriomeningitis virus. Two P-PMO targeting the viral transcription-regulatory sequence (TRS) region in the 5′ untranslated region were the most effective inhibitors tested. After several viral passages in the presence of a TRS-targeted P-PMO, partially drug-resistant SARS-CoV mutants arose which contained three contiguous base point mutations at the binding site of a TRS-targeted P-PMO. Those partially resistant viruses grew more slowly and formed smaller plaques than wild-type SARS-CoV. These results suggest PMO compounds have powerful therapeutic and investigative potential toward coronavirus infection.


Journal of Virology | 2004

Antisense Morpholino-Oligomers Directed against the 5′ End of the Genome Inhibit Coronavirus Proliferation and Growth†

Benjamin W. Neuman; David A. Stein; Andrew D. Kroeker; Amy D. Paulino; Hong M. Moulton; Patrick L. Iversen; Michael J. Buchmeier

ABSTRACT Conjugation of a peptide related to the human immunodeficiency virus type 1 Tat represents a novel method for delivery of antisense morpholino-oligomers. Conjugated and unconjugated oligomers were tested to determine sequence-specific antiviral efficacy against a member of the Coronaviridae, Mouse hepatitis virus (MHV). Specific antisense activity designed to block translation of the viral replicase polyprotein was first confirmed by reduction of luciferase expression from a target sequence-containing reporter construct in both cell-free and transfected cell culture assays. Peptide-conjugated morpholino-oligomers exhibited low toxicity in DBT astrocytoma cells used for culturing MHV. Oligomer administered at micromolar concentrations was delivered to >80% of cells and inhibited virus titers 10- to 100-fold in a sequence-specific and dose-responsive manner. In addition, targeted viral protein synthesis, plaque diameter, and cytopathic effect were significantly reduced. Inhibition of virus infectivity by peptide-conjugated morpholino was comparable to the antiviral activity of the aminoglycoside hygromycin B used at a concentration fivefold higher than the oligomer. These results suggest that this composition of antisense compound has therapeutic potential for control of coronavirus infection.


Veterinary Microbiology | 2006

Suppression of porcine reproductive and respiratory syndrome virus replication by morpholino antisense oligomers

Yan-Jin Zhang; David A. Stein; Su-Min Fan; Kai-Yu Wang; Andrew D. Kroeker; Xiang-Jin Meng; Patrick L. Iversen; David O. Matson

Abstract Porcine reproductive and respiratory syndrome virus (PRRSV) is the causative agent of a contagious disease characterized by reproductive failure in sows and respiratory disease in piglets. This infectious disease results in significant losses in the swine industry and specific anti-PRRSV drugs are needed. In this study, we evaluated a novel class of antisense compounds, peptide-conjugated phosphorodiamidate morpholino oligomers (P-PMOs), for their ability to suppress PRRSV replication in cell culture. P-PMOs are analogs of single-stranded DNA and contain a modified backbone that confers highly specific binding to RNA and resistance to nucleases. Of six P-PMOs tested, one (‘5UP1’), with sequence complementary to the 5′-terminal 21 nucleotides of the PRRSV genome, was found to be highly effective at reducing PRRSV replication in a specific and dose-dependent manner in CRL11171 cells in culture. 5UP1 treatment generated up to a 4.5log reduction in infectious PRRSV yield, while a control P-PMO had no effect on viral titer. Immunofluorescence assay with an anti-PRRSV monoclonal antibody confirmed the titer observations. The sequence-specificity of 5UP1 effect was confirmed in part by a cell-free luciferase reporter assay system, which showed that 5UP1-mediated inhibition of translation decreased if the target-RNA contained mispairings in relation to the 5UP1 P-PMO. Real-time RT-PCR showed that the production of PRRSV negative-sense RNA was reduced if 5UP1 was added to cells at up to 6h post-virus inoculation. Cell viability assays detected no cytotoxicity of 5UP1 within the concentration-range of this study. These results indicate that P-PMO 5UP1 has potential as an anti-PRRSV agent.


Advances in Experimental Medicine and Biology | 2006

Inhibition and escape of SARS-CoV treated with antisense morpholino oligomers.

Benjamin W. Neuman; David A. Stein; Andrew D. Kroeker; Richard K. Bestwick; Patrick L. Iversen; Hong M. Moulton; Michael J. Buchmeier

Identification of potential SARS-CoV antiviral compounds has progressed swiftly, thanks in part to the availability of bioinformatic and virus structural data. Antivirals that target the SARS-CoV superfamily 1 helicase and the 3C-related serine proteinase with low micromolar EC50 values have been reported. The papain-related cysteine proteinase may prove to be an unsuitable target, as a coronavirus molecular clone lacking one of the two known cleavage sites for this enzyme displayed only minor growth defects in cell culture. Other confirmed and putative viral enzymes including the polymerase, poly(U)specific endo-ribonuclease homolog, S-adenosyl-methionine-dependent ribose 2’-Omethyltransferase, and cyclic phosphodiesterase represent plausible anti-SARS targets. Antivirals targeting the interaction of the viral spike protein with the ACE-2 receptor, or with the spike-mediated fusion event, and showing micromolar-scale efficiency in cell culture, have been reported. Several groups have also reported antiviral in vitro efficacy with siRNAs. The antisense agents directed against single-stranded RNA are known to act by two general mechanisms: by causing damage to an RNA strand containing the complementary “target” sequence through priming of endogenous RNase H activity, or by stably binding to and steric interference with targeted RNA function. Phosphorodiamidate morpholino oligomers (PMO) act by the latter mechanism, duplexing to specific RNA sequence by Watson-Crick base pairing and forming a steric block. The most frequently successful targeting strategies for PMO-based gene knockdown involve interfering with translation initiation or masking splice sites. We recently demonstrated antiviral effects in vitro for one peptide-conjugated PMO (P-PMO) complementary to the AUG translation start site region of a murine coronavirus replicase


Bioconjugate Chemistry | 2005

Arginine-rich peptide conjugation to morpholino oligomers: effects on antisense activity and specificity.

Michelle H. Nelson; David A. Stein; Andrew D. Kroeker; Susie A. Hatlevig; Patrick L. Iversen; Hong M. Moulton


Archive | 2004

Compositions for enhancing transport of molecules into cells

Patrick L. Iversen; Hong M. Moulton; Michelle H. Nelson; Andrew D. Kroeker; David A. Stein


Archive | 2004

Compositions for enhancing transport and antisense efficacy of nucleic acid analog into cells

Patrick L. Iversen; Hong M. Moulton; Michelle H. Nelson; Andrew D. Kroeker; David A. Stein

Collaboration


Dive into the Andrew D. Kroeker's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David O. Matson

Eastern Virginia Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Claire Y.-H. Huang

Centers for Disease Control and Prevention

View shared research outputs
Researchain Logo
Decentralizing Knowledge