Network


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

Hotspot


Dive into the research topics where C D Morrow is active.

Publication


Featured researches published by C D Morrow.


Virology | 2003

Immunogenicity in pig-tailed macaques of poliovirus replicons expressing HIV-1 and SIV antigens and protection against SHIV-89.6P disease.

Patricia N. Fultz; Jackie Stallworth; D C Porter; Miroslav Novak; Marie J Anderson; C D Morrow

In the search for an effective vaccine against the human immunodeficiency virus (HIV), novel ways to deliver viral antigens are being evaluated. One such approach is the use of nonreplicating viral vectors encoding HIV and/or SIV genes that are expressed after infection of host cells. Nonreplicating poliovirus vectors, termed replicons, that expressed HIV-1/HXB2 and SIVmac239 gag and various HIV-1 env genes from different clades were tested for immunogenicity and protective efficacy against intravenous challenge of pig-tailed macaques with SHIV-89.6P. To maximize both cellular and humoral immune responses, a prime-boost regimen was used. Initially, macaques were immunized four times over 35 weeks by either the intranasal and intrarectal or the intramuscular (im) route with mixtures of poliovirus replicons expressing HIV-1 gag and multiple env genes. Immunization with replicons alone induced both serum antibodies and lymphocyte proliferative responses. After boosting with purified Env protein, neutralizing antibodies to SHIV-89.6P were induced in four of five immunized animals. In a second experiment, four macaques were immunized im three times over 27 weeks with replicons expressing the SIVmac239 gag and HIV-1/HXB2 env genes. All immunized animals were then boosted twice with purified HIV-1-89.6 rgp140-Env and SIVmac239 p55-Gag proteins. Four control animals received only the two protein inoculations. Immunized and control animals were then challenged intravenously with the pathogenic SHIV-89.6P. After challenge the animals were monitored for virus isolation from peripheral blood mononuclear cells and plasma viremia and for changes in virus-specific antibody titers. Naïve pig-tailed macaques experienced rapid loss of CD4(+) T cells and died between 38 and 62 weeks after infection. In contrast, macaques immunized with replicons and proteins rapidly cleared plasma virus and did not experience sustained loss of CD4(+) lymphocytes. Furthermore, two of the four macaques that were immunized only with purified proteins maintained high viral burdens and lost greater than 95% of their CD4(+) lymphocytes within 2 to 4 weeks after challenge. Thus, poliovirus replicons expressing HIV-1 and SIV antigens were immunogenic in pig-tailed macaques and appeared to enhance the protective effects observed after administration of purified proteins alone.


Journal of Virology | 2000

Inherent instability of poliovirus genomes containing two internal ribosome entry site (IRES) elements supports a role for the IRES in encapsidation.

Lisa K. Johansen; C D Morrow

ABSTRACT Previous studies have described poliovirus genomes in which the internal ribosome entry (IRES) for encephalomyocarditis virus (EMCV) is positioned between the P1 and P2-P3 open reading frames of the poliovirus genome. Although these dicistronic poliovirus genomes were replication competent, most exhibited evidence of genetic instability, and the EMCV IRES was deleted upon serial passage. One possible reason for instability of the genome is that the dicistronic genome was at least 108% larger than the wild-type poliovirus genome, which could reduce the efficiency of encapsidation. To address this possibility, we have constructed dicistronic poliovirus replicons by substituting the EMCV IRES and the gene encoding luciferase in place of the poliovirus P1 region; the resulting dicistronic replicons are smaller than the wild-type poliovirus genome. One dicistronic genome was constructed in which the poliovirus 5′ nontranslated region was fused to the gene encoding luciferase, followed by the complete EMCV IRES fused to the P2-P3 region of the poliovirus genome (PV-Luc-EMCV). A second dicistronic genome, EMCV-Luc-PV, was constructed with the first 108 nucleotides of the poliovirus genome fused to the EMCV IRES, followed by the gene encoding luciferase and the poliovirus IRES fused to the remaining P2-P3 region of the poliovirus genome. Both dicistronic replicons expressed abundant luciferase following transfection of in vitro-transcribed RNA into HeLa cells at 30, 33, or 37°C. The luciferase activity detected from PV-Luc-EMCV increased rapidly during the first 4 h following transfection and then plateaued, peaking after approximately 24 h. In contrast, the luciferase activity detected from EMCV-Luc-PV increased for approximately 12 h following transfection; by 24 h posttransfection, the overall levels of luciferase activity were similar to that of PV-Luc-EMCV. To analyze encapsidation of the dicistronic replicons, we used a system in which the capsid protein (P1) is provided in trans from a recombinant vaccinia virus (VV-P1). The PV-Luc-EMCV replicon was unstable upon serial passage in the presence of VV-P1, with deletions of the EMCV IRES region detected even during the initial transfection at 37°C. Following serial passage in the presence of VV-P1 at 33 or 30°C, we detected deleted genomes in which the luciferase gene was fused with the P2-P3 genes of the poliovirus genome so as to maintain the translational reading frame. In contrast, the EMCV-Luc-PV replicon was genetically stable during passage with VV-P1 at 33 or 30°C. The encapsidation of EMCV-Luc-PV was compared to that of monocistronic replicons encoding luciferase with either a poliovirus or EMCV IRES. Analysis of the encapsidated replicons after four serial passages with VV-P1 revealed that the dicistronic replicon was encapsidated more efficiently than the monocistronic replicon with the EMCV IRES but less efficiently than the monicistronic replicon with the poliovirus IRES. The results of this study suggest a genetic predisposition for picornavirus genomes to contain a single IRES region and are discussed with respect to a role of the IRES in encapsidation.


Methods in molecular medicine | 1997

Methods for the Use of Poliovirus Vectors for Gene Delivery

C D Morrow; David C. Ansardi; D C Porter

Poliovirus is a member of the Picornaviridae family of viruses. Characteristic of all members of this family, the poliovirus genome consrsis of approx 7500 bp of RNA of the plus sense polarity (1,2). Poliovirus is undoubtedly one of the most thoroughly characterized animal viruses. The three-dimensional structure of the entire virion is known (3), an infectious cDNA clone of the poliovirus genome has been generated (4,5), the entire nucleic acid sequence of poliovirus has been determined (1), and the cellular receptor that pohovirus uses to enter cells has been cloned and sequenced (6). The availability of a transgenic mouse expressing the poliovirus receptor has facilitated further description of the pathogenesis of poliovirus (7,8).


Journal of Virology | 1992

In vitro enzymatic activity of human immunodeficiency virus type 1 reverse transcriptase mutants in the highly conserved YMDD amino acid motif correlates with the infectious potential of the proviral genome.

John K. Wakefield; Sandra Jablonski; C D Morrow


Journal of Virology | 1997

Identification of a sequence within U5 required for human immunodeficiency virus type 1 to stably maintain a primer binding site complementary to tRNA(Met).

Sang-Moo Kang; Zhijun Zhang; C D Morrow


Journal of Virology | 1995

Mutation of the Aspartic Acid Residues of the GDD Sequence Motif of Poliovirus RNA-Dependent RNA Polymerase Results in Enzymes with Altered Metal Ion Requirements for Activity

S A Jablonski; C D Morrow


Journal of Virology | 1995

Encapsidation of poliovirus replicons encoding the complete human immunodeficiency virus type 1 gag gene by using a complementation system which provides the P1 capsid protein in trans.

D C Porter; D C Ansardi; C D Morrow


Journal of Virology | 1991

Coinfection with recombinant vaccinia viruses expressing poliovirus P1 and P3 proteins results in polyprotein processing and formation of empty capsid structures.

David C. Ansardi; D C Porter; C D Morrow


Journal of Virology | 1993

Complementation of a poliovirus defective genome by a recombinant vaccinia virus which provides poliovirus P1 capsid precursor in trans.

David C. Ansardi; D C Porter; C D Morrow


Journal of Virology | 1992

Myristylation of poliovirus capsid precursor P1 is required for assembly of subviral particles.

David C. Ansardi; D C Porter; C D Morrow

Collaboration


Dive into the C D Morrow's collaboration.

Top Co-Authors

Avatar

David C. Ansardi

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donna C. Porter

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John K. Wakefield

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhijun Zhang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge