Network


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

Hotspot


Dive into the research topics where David W. Lazinski is active.

Publication


Featured researches published by David W. Lazinski.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Replicating hepatitis delta virus RNA is edited in the nucleus by the small form of ADAR1

Swee Kee Wong; David W. Lazinski

Hepatitis delta virus (HDV) uses a host-encoded RNA-editing activity to express its two essential proteins from the same coding sequence. Adenosine deaminase that acts on RNA (ADAR)1 and ADAR2 are enzymes that catalyze such reactions, and each, when overexpressed, are capable of editing HDV RNA in vivo. However, the enzyme responsible for editing HDV RNA during replication has not been determined. Mammalian cells express two forms of ADAR1, a large form (ADAR1-L) that mainly localizes to the cytoplasm and a small form (ADAR1-S) that resides in the nucleus. Recently, we found that the specific activity of ADAR1-L within cells is much higher than that of ADAR1-S but only when the substrate can be edited in the cytoplasm. Here we observed that although both ADAR1-S and ADAR1-L were expressed throughout HDV replication, no ADAR2 could be observed at any time. Using expression vectors that individually overexpress either form of ADAR1, we found that ADAR1-S could stimulate editing during replication more efficiently. We next reduced ADAR1 levels during HDV replication. After transfection of an ADAR1-L-specific small interfering RNA (siRNA), we observed a significant loss of that protein and its associated cytoplasmic editing activity while the level of ADAR1-S remained unchanged. Transfection of this siRNA, however, did not reduce editing during HDV replication. In contrast, transfection of an siRNA that targets both forms of ADAR1 greatly reduced the expression of both proteins and potently inhibited editing during replication. We conclude that ADAR1-S edits HDV RNA during replication and that editing occurs in the nucleus.


Journal of Virology | 2001

Hepatitis Delta Virus Minimal Substrates Competent for Editing by ADAR1 and ADAR2

Shuji Sato; Swee Kee Wong; David W. Lazinski

ABSTRACT A host-mediated RNA-editing event allows hepatitis delta virus (HDV) to express two essential proteins, the small delta antigen (HDAg-S) and the large delta antigen (HDAg-L), from a single open reading frame. One or several members of the ADAR (adenosine deaminases that act on RNA) family are thought to convert the adenosine to an inosine (I) within the HDAg-S amber codon in antigenomic RNA. As a consequence of replication, the UIG codon is converted to a UGG (tryptophan [W]) codon in the resulting HDAg-L message. Here, we used a novel reporter system to monitor the editing of the HDV amber/W site in the absence of replication. In cultured cells, we observed that both human ADAR1 (hADAR1) and hADAR2 were capable of editing the amber/W site with comparable efficiencies. We also defined the minimal HDV substrate required for hADAR1- and hADAR2-mediated editing. Only 24 nucleotides from the amber/W site were sufficient to enable efficient editing by hADAR1. Hence, the HDV amber/W site represents the smallest ADAR substrate yet identified. In contrast, the minimal substrate competent for hADAR2-mediated editing contained 66 nucleotides.


Journal of Virology | 2004

By Inhibiting Replication, the Large Hepatitis Delta Antigen Can Indirectly Regulate Amber/W Editing and Its Own Expression

Shuji Sato; Cromwell Cornillez-Ty; David W. Lazinski

ABSTRACT Hepatitis delta virus (HDV) expresses two essential proteins with distinct functions. The small hepatitis delta antigen (HDAg-S) is expressed throughout replication and is needed to promote that process. The large form (HDAg-L) is farnesylated, is expressed only at later times via RNA editing of the amber/W site, and is required for virion assembly. When HDAg-L is artificially expressed at the onset of replication, it strongly inhibits replication. However, there is controversy concerning whether HDAg-L expressed naturally at later times as a consequence of editing and replication can similarly inhibit replication. Here, by stabilizing the predicted secondary structure downstream from the amber/W site, a replication-competent HDV mutant that exhibited levels of editing higher than those of the wild type was created. This mutant expressed elevated levels of HDAg-L early during replication, and at later times, its replication aborted prematurely. No further increase in amber/W editing was observed following the cessation of replication, indicating that editing was coupled to replication. A mutation in HDAg-L and a farnesyl transferase inhibitor were both used to abolish the ability of HDAg-L to inhibit replication. Such treatments rescued the replication defect of the overediting mutant, and even higher levels of amber/W editing resulted. It was concluded that when expressed naturally during replication, HDAg-L is able to inhibit replication and thereby inhibit amber/W editing and its own synthesis. In addition, the structure adjacent to the amber/W site is suboptimal for editing, and this creates a window of time in which replication can occur in the absence of HDAg-L.


Journal of Virology | 2005

Roles of Carboxyl-Terminal and Farnesylated Residues in the Functions of the Large Hepatitis Delta Antigen

Brendan O'Malley; David W. Lazinski

ABSTRACT The large hepatitis delta antigen (HDAg-L) mediates hepatitis delta virus (HDV) assembly and inhibits HDV RNA replication. Farnesylation of the cysteine residue within the HDAg-L carboxyl terminus is required for both functions. Here, HDAg-L proteins from different HDV genotypes and genotype chimeric proteins were analyzed for their ability to incorporate into virus-like particles (VLPs). Observed differences in efficiency of VLP incorporation could be attributed to genotype-specific differences within the HDAg-L carboxyl terminus. Using a novel assay to quantify the extent of HDAg-L farnesylation, we found that genotype 3 HDAg-L was inefficiently farnesylated when expressed in the absence of the small hepatitis delta antigen (HDAg-S). However, as the intracellular ratio of HDAg-S to HDAg-L was increased, so too was the extent of HDAg-L farnesylation for all three genotypes. Single point mutations within the carboxyl terminus of HDAg-L were screened, and three mutants that severely inhibited assembly without affecting farnesylation were identified. The observed assembly defects persisted under conditions where the mutants were known to have access to the site of VLP assembly. Therefore, the corresponding residues within the wild-type protein are likely required for direct interaction with viral envelope proteins. Finally, it was observed that when HDAg-S was artificially myristoylated, it could efficiently inhibit HDV RNA replication. Hence, a general association with membranes enables HDAg to inhibit replication. In contrast, although myristoylated HDAg-S was incorporated into VLPs far more efficiently than HDAg-S or nonfarnesylated HDAg-L, it was incorporated far less efficiently than wild-type HDAg-L; thus, farnesylation was required for efficient assembly.


Advances in Virus Research | 1994

Recent Developments in Hepatitis Delta Virus Research

David W. Lazinski; John M. Taylor

Publisher Summary Although delta virus requires HBV proteins for its assembly, it is fully capable of replicating its genome in the absence of helper virus functions. A rolling-circle mechanism for HDV replication has been proposed, in which a host-encoded RNA-directed polymerase proceeds multiple times around the circular genome to generate a complementary multimeric intermediate; cis-Acting catalytic regions on this RNA (ribozymes) promote site-specific cleavage and ligation to generate a circular unit-length complement of the genome, termed as the antigenome. The antigenome is thought to serve as the template for genome synthesis via the same mechanism. The size, conformation, and structure of the genome, as well as its replication strategy, make HDV unique among animal viruses and as a result, the delta virus has no taxonomic classification. There is, however, a group of plant pathogens, most notable of which are the viroids and virusoids, which share many similarities with HDV. For example, the viroid genome is also a single-stranded circular RNA able to fold into an unbranched rod structure with extensive intramolecular base-pairing, is replicated by a host polymerase via a rolling-circle mechanism, and encodes ribozymes capable of both self-cleavage and ligation. Unlike the viroids, however, HDV does encode a protein, the small delta antigen (GAg-S) that binds viral RNA to form ribonucleoprotein complexes (RNPs) both within the virions and in the nuclei of the infected cells. This protein functions to facilitate genome replication in a manner that is not yet understood. As replication proceeds, a very specific RNA-editing event is ultimately responsible for a single nucleotide substitution that eliminates the GAg-S stop codon. The resulting protein, GAg-L, contains an additional 19 amino acids at its C terminus. RNA-editing represents an essential step in the completion of the viral life cycle, because only GAg-L is able to function with the HBV envelope proteins to promote the packaging and release of the infectious virions.


Journal of Virology | 2003

Determination of the Multimerization State of the Hepatitis Delta Virus Antigens In Vivo

Cromwell Cornillez-Ty; David W. Lazinski

ABSTRACT Hepatitis delta virus expresses two essential proteins, the small and large delta antigens, and both are required for viral propagation. Proper function of each protein depends on the presence of a common amino-terminal multimerization domain. A crystal structure, solved using a peptide fragment that contained residues 12 to 60, depicts the formation of an octameric ring composed of antiparallel coiled-coil dimers. Because this crystal structure was solved for only a fragment of the delta antigens, it is unknown whether octamers actually form in vivo at physiological protein concentrations and in the context of either intact delta antigen. To test the relevance of the octameric structure, we developed a new method to probe coiled-coil structures in vivo. We generated a panel of mutants containing cysteine substitutions at strategic locations within the predicted monomer-monomer interface and the dimer-dimer interface. Since the small delta antigen contains no cysteine residues, treatment of cell extracts with a mild oxidizing reagent was expected to induce disulfide bond formation only when the appropriate pairs of cysteine substitution mutants were coexpressed. We indeed found that, in vivo, both the small and large delta antigens assembled as antiparallel coiled-coil dimers. Likewise, we found that both proteins could assume an octameric quaternary structure in vivo. Finally, during the course of these experiments, we found that unprenylated large delta antigen molecules could be disulfide cross-linked via the sole cysteine residue located within the carboxy terminus. Therefore, in vivo, the C terminus likely provides an additional site of protein-protein interaction for the large delta antigen.


Journal of Virology | 2002

A Hepatitis B Surface Antigen Mutant That Lacks the Antigenic Loop Region Can Self-Assemble and Interact with the Large Hepatitis Delta Antigen

Brendan O'Malley; David W. Lazinski

ABSTRACT A novel hepatitis B virus surface antigen mutant harboring a deletion of most of the major antigenic loop region was competent for self-assembly and secretion. Although the mutant protein was competent for interaction with and incorporation of free large hepatitis delta antigen, it was partially defective in hepatitis delta virus RNP incorporation.


Biochimie | 1994

RNA editing in the replication cycle of human hepatitis delta virus

Ting-Ting Wu; Hans J. Netter; V. Bichko; David W. Lazinski; John M. Taylor

For some time it has been known that the RNA genome of human hepatitis delta virus (HDV) undergoes a specific RNA editing event. This review describes the editing phenomenon and its potential biological significance, and evaluates the data regarding the mechanism involved, including the possible relationship to other RNA editing phenomena.


Methods in Molecular Genetics | 1995

[20] Molecular studies of hepatitis delta virus RNAs

Tie-Bo Fu; Ting-Ting Wu; David W. Lazinski; John M. Taylor

Publisher Summary This chapter discusses the molecular studies of hepatitis delta virus RNAs. During the past few years, the application of new techniques of molecular biology for studying hepatitis delta virus (HDV) replication has revealed some of the special and unique aspects by which this virus is reproduced. The chapter describes two of the quantitative techniques used in the laboratory at Fox Chase Cancer Center (Philadelphia, PA) to study the molecular biology of HDV. These techniques are based on the use of the nested polymerase chain reaction (PCR). The first technique is a sensitive quantitative assay for HDV genomic and antigenomic HDV RNAs. The second makes use of the immobilization of PCR products via the avidin-biotin reaction so as to quantitate RNA editing. The immobilization procedure can also be used for other purposes, such as dideoxysequencing of both strands of the PCR products.


Journal of Virology | 1992

Editing on the genomic RNA of human hepatitis delta virus.

Haoqiang Zheng; Tie-Bo Fu; David W. Lazinski; John M. Taylor

Collaboration


Dive into the David W. Lazinski's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ting-Ting Wu

Fox Chase Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tie-Bo Fu

Fox Chase Cancer Center

View shared research outputs
Top Co-Authors

Avatar

V. Bichko

Fox Chase Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge