Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
1.
Nat Commun ; 10(1): 5524, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31797930

ABSTRACT

CRISPR-Cas systems have revolutionized genome editing across a broad range of biotechnological endeavors. Many CRISPR-Cas nucleases have been identified and engineered for improved capabilities. Given the modular structure of such enzymes, we hypothesized that engineering chimeric sequences would generate non-natural variants that span the kinetic parameter landscape, and thus provide for the rapid selection of nucleases fit for a particular editing system. Here, we design a chimeric Cas12a-type library with approximately 560 synthetic chimeras, and select several functional variants. We demonstrate that certain nuclease domains can be recombined across distantly related nuclease templates to produce variants that function in bacteria, yeast, and human cell lines. We further characterize selected chimeric nucleases and find that they have different protospacer adjacent motif (PAM) preferences and the M44 chimera has higher specificity relative to wild-type (WT) sequences. This demonstration opens up the possibility of generating nuclease sequences with implications across biotechnology.


Subject(s)
CRISPR-Cas Systems , Endonucleases/metabolism , Gene Editing/methods , Recombinant Fusion Proteins/metabolism , Bacteria/genetics , Biotechnology/methods , Endonucleases/genetics , Gene Library , HEK293 Cells , Humans , Mutation , Recombinant Fusion Proteins/genetics , Reproducibility of Results , Yeasts/genetics
2.
Annu Rev Virol ; 4(1): 309-325, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28715971

ABSTRACT

The strong dependence of retroviruses, such as human immunodeficiency virus type 1 (HIV-1), on host cell factors is no more apparent than when the endosomal sorting complex required for transport (ESCRT) machinery is purposely disengaged. The resulting potent inhibition of retrovirus release underscores the importance of understanding fundamental structure-function relationships at the ESCRT-HIV-1 interface. Recent studies utilizing advanced imaging technologies have helped clarify these relationships, overcoming hurdles to provide a range of potential models for ESCRT-mediated virus abscission. Here, we discuss these models in the context of prior work detailing ESCRT machinery and the HIV-1 release process. To provide a template for further refinement, we propose a new working model for ESCRT-mediated HIV-1 release that reconciles disparate and seemingly conflicting studies.


Subject(s)
Endosomal Sorting Complexes Required for Transport/physiology , HIV-1/metabolism , Virus Release , Biological Transport , Cell Line , Gene Products, gag/genetics , Gene Products, gag/metabolism , Humans , Membranes/metabolism , Models, Biological , Protein Transport
3.
Br J Cancer ; 101(3): 465-72, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19603024

ABSTRACT

BACKGROUND: We address the prognostic and predictive value of KRAS, PIK3CA and BRAF mutations for clinical outcomes in response to active agents in the treatment of metastatic colorectal cancer (mCRC). METHODS: We determined KRAS, BRAF and PIK3CA mutations in tumours from 168 patients treated for mCRC at two institutions. All patients received 5-FU-based first-line chemotherapy and treatment outcome was analysed retrospectively. RESULTS: KRAS, BRAF and PIK3CA mutations were present in 62 (37%), 13 (8%) and 26 (15%) cases, respectively. Multivariate analysis uncovered BRAF mutation as an independent prognostic factor for decreased survival (hazard ratio (HR) 4.0, 95% confidence interval (CI) 2.1-7.6). In addition, patients with BRAF-mutant tumours had significantly lower progression-free survival (PFS: HR 4.0, 95% CI 2.2-7.4) than those whose tumors that carried wild-type BRAF. Among 92 patients treated using chemotherapy and cetuximab as salvage therapy, KRAS mutation was associated with lack of response (P=0.002) and shorter PFS (P=0.09). BRAF (P=0.0005) and PIK3CA (P=0.01) mutations also predicted reduced PFS in response to cetuximab salvage therapy. CONCLUSIONS: These results underscore the potential of mutational profiling to identify CRCs with different natural histories or treatment responses. The adverse significance of BRAF mutation should inform patient selection and stratification in clinical trials.


Subject(s)
Colorectal Neoplasms/genetics , Mutation , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Cetuximab , Class I Phosphatidylinositol 3-Kinases , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Predictive Value of Tests , Prognosis , Proto-Oncogene Proteins p21(ras) , Salvage Therapy
4.
Proc Natl Acad Sci U S A ; 100(23): 13555-60, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14573704

ABSTRACT

New HIV therapies are urgently needed to address the growing problem of drug resistance. In this article, we characterize the anti-HIV drug candidate 3-O-(3',3'-dimethylsuccinyl) betulinic acid (PA-457). We show that PA-457 potently inhibits replication of both WT and drug-resistant HIV-1 isolates and demonstrate that the compound acts by disrupting a late step in Gag processing involving conversion of the capsid precursor (p25) to mature capsid protein (p24). We find that virions from PA-457-treated cultures are noninfectious and exhibit an aberrant particle morphology characterized by a spherical, acentric core and a crescent-shaped, electron-dense shell lying just inside the viral membrane. To identify the determinants of compound activity we selected for PA-457-resistant virus in vitro. Consistent with the effect on Gag processing, we found that mutations conferring resistance to PA-457 map to the p25 to p24 cleavage site. PA-457 represents a unique class of anti-HIV compounds termed maturation inhibitors that exploit a previously unidentified viral target, providing additional opportunities for HIV drug discovery.


Subject(s)
Anti-HIV Agents/pharmacology , Gene Products, gag/chemistry , Succinates/pharmacology , Triterpenes/pharmacology , Binding Sites , Chromobox Protein Homolog 5 , Drug Design , Gene Products, gag/antagonists & inhibitors , Genotype , HIV Core Protein p24/metabolism , HeLa Cells , Humans , Inhibitory Concentration 50 , Microscopy, Electron , Models, Chemical , Models, Genetic , Mutation , Plasmids/metabolism , Precipitin Tests , Succinates/chemistry , Triterpenes/chemistry , gag Gene Products, Human Immunodeficiency Virus
5.
Proc Natl Acad Sci U S A ; 98(24): 13925-30, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11717449

ABSTRACT

HIV-1 particle production occurs in a series of steps promoted by the viral Gag protein. Although it is well established that assembly and release take place at the plasma membrane, the nature of membrane assembly sites remains poorly understood. We show here that Gag specifically associates with cholesterol-enriched microdomains ("rafts") at the plasma membrane. Kinetic studies demonstrate that raft association follows membrane binding, and the analysis of Gag mutants reveals that, whereas the N terminus of Gag mediates raft binding, this association is greatly enhanced by Gag-Gag interaction domains. We observe that depletion of cellular cholesterol markedly and specifically reduces HIV-1 particle production. Furthermore, treatment of virus-producing cells or virus particles with raft-disrupting agents significantly impairs virus infectivity. These results identify the association of Gag with plasma membrane rafts as an important step in HIV-1 replication. These findings may lead to novel strategies for suppressing HIV-1 replication in vivo.


Subject(s)
HIV-1/physiology , Membrane Microdomains/physiology , Virus Assembly/physiology , Binding Sites , Cell Membrane/drug effects , Cell Membrane/metabolism , Cholesterol/metabolism , Detergents/pharmacology , Drug Resistance , Gene Products, gag/genetics , Gene Products, gag/metabolism , HIV-1/metabolism , HeLa Cells , Humans , Membrane Microdomains/metabolism , Octoxynol/pharmacology , Protein Binding , Protein Precursors/genetics , Protein Precursors/metabolism
6.
J Virol ; 75(19): 9357-66, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533199

ABSTRACT

A group of conserved hydrophobic residues faces the interior of the coiled-coil-like structure within the N-terminal domain of the human immunodeficiency virus type 1 (HIV-1) capsid protein (CA). It has been suggested that these residues are important for maintaining stable structure and functional activity. To investigate this possibility, we constructed two HIV-1 clones, in which Trp23 or Phe40 was changed to Ala. We also constructed a third mutant, D51A, which has a mutation that destroys a salt bridge between Pro1 and Asp51. All three mutants are replication defective but produce virus particles. Mutant virions contain all of the viral proteins, although the amount and stability of CA are decreased and levels of virion-associated integrase are reduced. The mutations do not affect endogenous reverse transcriptase activity; however, the mutants are blocked in their ability to initiate reverse transcription in infected cells and no minus-strand strong-stop DNA is detected. The defect in reverse transcription is associated with striking defects in the morphology of mutant virus cores, as determined by transmission electron microscopy. Our data indicate that the mutations made in this study disrupt CA structure and prevent proper maturation of virus cores. We propose that this results in a defect in core stability or in an early postentry event preceding reverse transcription.


Subject(s)
Gene Products, gag/genetics , HIV Infections/virology , HIV-1/physiology , HeLa Cells , Humans , Mutation , Transcription, Genetic/genetics , Virus Replication/genetics
7.
Cell ; 105(5): 645-55, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389834

ABSTRACT

We have discovered an early mitotic inhibitor, Emi1, which regulates mitosis by inhibiting the anaphase promoting complex/cyclosome (APC). Emi1 is a conserved F box protein containing a zinc binding region essential for APC inhibition. Emi1 accumulates before mitosis and is ubiquitylated and destroyed in mitosis, independent of the APC. Emi1 immunodepletion from cycling Xenopus extracts strongly delays cyclin B accumulation and mitotic entry, whereas nondestructible Emi1 stabilizes APC substrates and causes a mitotic block. Emi1 binds the APC activator Cdc20, and Cdc20 can rescue an Emi1-induced block to cyclin B destruction. Our results suggest that Emi1 regulates progression through early mitosis by preventing premature APC activation, and may help explain the well-known delay between cyclin B/Cdc2 activation and cyclin B destruction.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Drosophila Proteins , Ligases/metabolism , Mitosis/physiology , Proteins/metabolism , Ubiquitin-Protein Ligase Complexes , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Animals , Cdc20 Proteins , Conserved Sequence , Cyclin A/metabolism , Cyclin B/metabolism , Drosophila , In Vitro Techniques , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Oocytes/cytology , Oocytes/physiology , Rabbits , Ubiquitin-Protein Ligases , Xenopus , Xenopus Proteins
8.
J Virol ; 75(10): 4681-91, 2001 May.
Article in English | MEDLINE | ID: mdl-11312339

ABSTRACT

Blood-borne human immunodeficiency virus type 1 (HIV-1) crosses the blood-brain barrier (BBB) to induce brain dysfunction. How HIV-1 crosses the BBB is unclear. Most work has focused on the ability of infected immune cells to cross the BBB, with less attention devoted to the study of free virus. Since the HIV-1 coat glycoprotein gp120 can cross the BBB, we postulated that gp120 might be key in determining whether free virus can cross the BBB. We used radioactive virions which do (Env+) or do not (Env-) bear the envelope proteins to characterize the ability of HIV-1 to be taken up by the murine BBB. In vivo and in vitro studies showed that the envelope proteins are key to the uptake of free virus and that uptake was enhanced by wheat germ agglutinin, strongly suggesting that the envelope proteins induce viral adsorptive endocytosis and transcytosis in brain endothelia. Capillary depletion showed that Env+ virus completely crossed the vascular BBB to enter the parenchyma of the brain. Virus also entered the cerebrospinal fluid, suggesting passage across the choroid plexus as well. About 0.22% of the intravenously injected dose was taken up per g of brain. In vitro studies showed that postinternalization membrane cohesion (membrane binding not reversed with acid wash or cell lysis) was a regulated event. Intact virus was recovered from the brain endothelial cytosol and was effluxed from the endothelial cells. These results show that free HIV-1 can cross the BBB by an event related to adsorptive endocytosis and mediated by the envelope proteins.


Subject(s)
Blood-Brain Barrier/physiology , Endocytosis/physiology , HIV Envelope Protein gp120/physiology , HIV Envelope Protein gp41/physiology , HIV-1/metabolism , Animals , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp41/genetics , HIV-1/physiology , HeLa Cells , Humans , Male , Mice
9.
Somat Cell Mol Genet ; 26(1-6): 13-33, 2001 Nov.
Article in English | MEDLINE | ID: mdl-12465460

ABSTRACT

In general terms, the replication cycle of lentiviruses, including HIV-1, closely resembles that of other retroviruses. There are, however, a number of unique aspects of HIV replication; for example, the HIVs and SIVs target receptors and coreceptors distinct from those used by other retroviruses. Lentiviruses encode a number of regulatory and accessory proteins not encoded by the genomes of the prototypical "simple" retroviruses. Of particular interest from the gene therapy perspective, lentiviruses possess the ability to productively infect some types of non-dividing cells. This chapter, while reiterating certain points discussed in Chapter 1, will attempt to focus on issues unique to HIV-1 replication. The HIV-1 genome encodes the major structural and non-structural proteins common to all replication-competent retroviruses (Fig. 1, and Chapter 1). From the 5'- to 3'-ends of the genome are found the gag (for group-specific antigen), pol (for polymerase), and env (for envelope glycoprotein) genes. The gag gene encodes a polyprotein precursor whose name, Pr55Gag, is based on its molecular weight. Pr55Gag is cleaved by the viral protease (PR) to the mature Gag proteins matrix (also known as MA or p17), capsid (CA or p24), nucleocapsid (NC or p7), and p6. Two spacer peptides, p2 and p1, are also generated upon Pr55Gag processing. The pol-encoded enzymes are initially synthesized as part of a large polyprotein precursor, Pr160GagPol, whose synthesis results from a rare frameshifting event during Pr55Gag translation. The individual pol-encoded enzymes, PR, reverse transcriptase (RT), and integrase (IN), are cleaved from Pr160GagPol by the viral PR. The envelope (Env) glycoproteins are also synthesized as a polyprotein precursor (Fig. 1). Unlike the Gag and Pol precursors, which are cleaved by the viral PR, the Env precursor, known as gp160, is processed by a cellular protease during Env trafficking to the cell surface, gp160 processing results in the generation of the surface (SU) Env glycoprotein gp120 and the transmembrane (TM) glycoprotein gp41. gp120 contains the determinants that interact with receptor and coreceptor, while gp41 not only anchors the gp120/gp41 complex in the membrane (Fig. 2), but also contains domains that are critical for catalyzing the membrane fusion reaction between viral and host lipid bilayers during virus entry. Comparison of env sequences from a large number of virus isolates revealed that gp120 is organized into five conserved regions (C1-C5) and five highly variable domains (V1-V5). The variable regions tend to be located in disulfide-linked loops. gp41 is composed of three major domains: the ectodomain (which contains determinants essential for membrane fusion), the transmembrane anchor sequence, and the cytoplasmic tail. In addition to the gag, pol, and env genes, HIV-1 also encodes a number of regulatory and accessory proteins. Tat is critical for transcription from the HIV-1 LTR and Rev plays a major [figure: see text] role in the transport of viral RNAs from the nucleus to the cytoplasm. Vpu, Vif, Vpr and Nef have been termed "accessory" or "auxiliary" proteins to reflect the fact that they are not uniformly required for virus replication. The functions of these very interesting proteins will be discussed in more detail at the end of this chapter. HIV replication proceeds in a series of events that can be divided into two overall phases: "early" and "late" (Fig. 3). Although some events occur in a concerted or simultaneous fashion, the replication cycle can be viewed most simply as proceeding in an ordered, step-wise manner. In this chapter, each step in virus replication will be considered; additional information can be obtained from the more detailed reviews and primary references that are cited.


Subject(s)
HIV-1/physiology , Virus Replication , Genes, Viral , HIV-1/genetics , Humans , RNA-Directed DNA Polymerase , Retroviridae/genetics , Retroviridae/physiology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Transcription, Genetic , Viral Structural Proteins/genetics
10.
Trends Cell Biol ; 10(10): 429-39, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10998601

ABSTRACT

Recently, many new examples of E3 ubiquitin ligases or E3 enzymes have been found to regulate a host of cellular processes. These E3 enzymes direct the formation of multiubiquitin chains on specific protein substrates, and - typically - the subsequent destruction of those proteins. We discuss how the modular architecture of E3 enzymes connects one of two distinct classes of catalytic domains to a wide range of substrate-binding domains. In one catalytic class, a HECT domain transfers ubiquitin directly to substrate bound to a non-catalytic domain. Members of the other catalytic class, found in the SCF, VBC and APC complexes, use a RING finger domain to facilitate ubiquitylation. The separable substrate-recognition domains of E3 enzymes provides a flexible means of linking a conserved ubiquitylation function to potentially thousands of ubiquitylated substrates in eukaryotic cells.


Subject(s)
Ligases/metabolism , Animals , Catalytic Domain , Eukaryotic Cells/enzymology , Humans , Ligases/chemistry , Substrate Specificity/physiology , Ubiquitin-Protein Ligases
11.
J Virol ; 74(14): 6669-74, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10864683

ABSTRACT

The antiretroviral nucleoside analog 2',3'-dideoxy-3'-thiacytidine (3TC) is a potent inhibitor of wild-type human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT). A methionine-to-valine or methionine-to-isoleucine substitution at residue 184 in the HIV-1 YMDD motif, which is located at the RT active site, leads to a high level of resistance to 3TC. We sought to determine whether 3TC can inhibit the replication of wild-type murine leukemia virus (MLV), which contains V223 at the YVDD active site motif of the MLV RT, and of the V223M, V223I, V223A, and V223S mutant RTs. Surprisingly, the wild type and all four of the V223 mutants of MLV RT were highly resistant to 3TC. These results indicate that determinants outside the YVDD motif of MLV RT confer a high level of resistance to 3TC. Therefore, structural differences among similar RTs might result in widely divergent sensitivities to antiretroviral nucleoside analogs.


Subject(s)
Lamivudine/pharmacology , Leukemia Virus, Murine/drug effects , RNA-Directed DNA Polymerase/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Amino Acid Substitution , Animals , Cell Line , Drug Resistance, Microbial , Humans , Leukemia Virus, Murine/enzymology , Mice , Mutation , RNA-Directed DNA Polymerase/genetics , Virus Replication/drug effects
12.
J Virol ; 74(11): 5142-50, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10799589

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) Gag precursor, Pr55(Gag), is necessary and sufficient for the assembly and release of viruslike particles. Binding of Gag to membrane and Gag multimerization are both essential steps in virus assembly, yet the domains responsible for these events have not been fully defined. In addition, the relationship between membrane binding and Gag-Gag interaction remains to be elucidated. To investigate these issues, we analyzed, in vivo, the membrane-binding and assembly properties of a series of C-terminally truncated Gag mutants. Pr55(Gag) was truncated at the C terminus of matrix (MAstop), between the N- and C-terminal domains of capsid (CA146stop), at the C terminus of capsid (p41stop), at the C terminus of p2 (p43stop), and after the N-terminal 35 amino acids of nucleocapsid (NC35stop). The ability of these truncated Gag molecules to assemble and release viruslike particles and their capacity to copackage into particles when coexpressed with full-length Gag were determined. We demonstrate that the amount of truncated Gag incorporated into particles is incrementally increased by extension from CA146 to NC35, suggesting that multiple sites in this region are involved in Gag multimerization. Using membrane flotation centrifugation, we observe that MA shows significantly reduced membrane binding relative to full-length Gag but that CA146 displays steady-state membrane-binding properties comparable to those of Pr55(Gag). The finding that the CA146 mutant, which contains only matrix and the N-terminal domain of capsid, exhibits levels of steady-state membrane binding equivalent to those of full-length Gag indicates that strong Gag-Gag interaction domains are not required for the efficient binding of HIV-1 Gag to membrane.


Subject(s)
Gene Products, gag/metabolism , HIV-1/metabolism , Protein Precursors/metabolism , Cell Membrane/metabolism , Gene Products, gag/genetics , HIV-1/genetics , HIV-1/physiology , HeLa Cells , Humans , Mutagenesis , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Precursors/genetics , Virion/physiology , Virus Assembly/physiology , gag Gene Products, Human Immunodeficiency Virus
13.
J Virol ; 74(8): 3548-54, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10729129

ABSTRACT

The incorporation of envelope (Env) glycoproteins into virions is an essential step in the retroviral replication cycle. Lentiviruses, including human immunodeficiency virus type 1 (HIV-1), encode Env glycoproteins with unusually long cytoplasmic tails, the functions of which have not been fully elucidated. In this study, we examine the effects on virus replication of a number of mutations in a helical motif (alpha-helix 2) located near the center of the HIV-1 gp41 cytoplasmic tail. We find that, in T-cell lines, small deletions in this domain disrupt the incorporation of Env glycoproteins into virions and markedly impair virus infectivity. Through the analysis of viral revertants, we demonstrate that a single amino acid change (34VI) in the matrix domain of Gag reverses the Env incorporation and infectivity defect imposed by a small deletion near the C terminus of alpha-helix 2. These results provide genetic evidence, in the context of infected T cells, for an interaction between HIV-1 matrix and the gp41 cytoplasmic tail and identify domains of both proteins involved in this putative interaction.


Subject(s)
Gene Products, gag/metabolism , HIV Antigens/metabolism , HIV Envelope Protein gp41/metabolism , HIV-1/physiology , Viral Proteins , Amino Acid Motifs , Amino Acid Sequence , Cell Line , Flow Cytometry , Gene Products, gag/chemistry , HIV Antigens/chemistry , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/genetics , HIV-1/genetics , HIV-1/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Protein Structure, Secondary , Radioimmunoprecipitation Assay , Sequence Deletion , T-Lymphocytes/virology , Virion/metabolism , Virus Replication , gag Gene Products, Human Immunodeficiency Virus
14.
J Virol ; 74(6): 2855-66, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10684302

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) particle formation and the subsequent initiation of protease-mediated maturation occur predominantly on the plasma membrane. However, the mechanism by which HIV-1 assembly is targeted specifically to the plasma membrane versus intracellular membranes is largely unknown. Previously, we observed that mutations between residues 84 and 88 of the matrix (MA) domain of HIV-1 Gag cause a retargeting of virus particle formation to an intracellular site. In this study, we demonstrate that the mutant virus assembly occurs in the Golgi or in post-Golgi vesicles. These particles undergo core condensation in a protease-dependent manner, indicating that virus maturation can occur not only on the plasma membrane but also in the Golgi or post-Golgi vesicles. The intracellular assembly of mutant particles is dependent on Gag myristylation but is not influenced by p6(Gag) or envelope glycoprotein expression. Previous characterization of viral revertants suggested a functional relationship between the highly basic domain of MA (amino acids 17 to 31) and residues 84 to 88. We now demonstrate that mutations in the highly basic domain also retarget virus particle formation to the Golgi or post-Golgi vesicles. Although the basic domain has been implicated in Gag membrane binding, no correlation was observed between the impact of mutations on membrane binding and Gag targeting, indicating that these two functions of MA are genetically separable. Plasma membrane targeting of Gag proteins with mutations in either the basic domain or between residues 84 and 88 was rescued by coexpression with wild-type Gag; however, the two groups of MA mutants could not rescue each other. We propose that the highly basic domain of MA contains a major determinant of HIV-1 Gag plasma membrane targeting and that mutations between residues 84 and 88 disrupt plasma membrane targeting through an effect on the basic domain.


Subject(s)
Gene Products, gag/physiology , HIV-1/physiology , Viral Proteins , Virus Assembly/physiology , Binding Sites , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression , Gene Products, env/metabolism , Gene Products, gag/genetics , Gene Products, gag/metabolism , Golgi Apparatus/metabolism , HIV Antigens/genetics , HIV Antigens/metabolism , HIV-1/ultrastructure , HeLa Cells , Humans , Mutagenesis , Myristic Acid/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , gag Gene Products, Human Immunodeficiency Virus
15.
Proc Natl Acad Sci U S A ; 97(1): 343-8, 2000 Jan 04.
Article in English | MEDLINE | ID: mdl-10618420

ABSTRACT

Lentiviruses, including HIV-1, have transmembrane envelope (Env) glycoproteins with cytoplasmic tails that are quite long compared with those of other retroviruses. However, mainly because of the lack of biochemical studies performed in cell types that are targets for HIV-1 infection, no clear consensus exists regarding the function of the long lentiviral Env cytoplasmic tail in virus replication. In this report, we characterize the biological and biochemical properties of an HIV-1 mutant lacking the gp41 cytoplasmic tail. We find that the gp41 cytoplasmic tail is necessary for the efficient establishment of a productive, spreading infection in the majority of T cell lines tested, peripheral blood mononuclear cells, and monocyte-derived macrophages. Biochemical studies using a high-level, transient HIV-1 expression system based on pseudotyping with the vesicular stomatitis virus glycoprotein demonstrate that in HeLa and MT-4 cells, mutant Env incorporation into virions is reduced only 3-fold relative to wild type. In contrast, gp120 levels in virions produced from a number of other T cell lines and primary macrophages are reduced more than 10-fold by the gp41 truncation. The Env incorporation defect imposed by the cytoplasmic tail truncation is not the result of increased shedding of gp120 from virions or reduced cell-surface Env expression. These results demonstrate that in the majority of T cell lines, and in primary cell types that serve as natural targets for HIV-1 infection in vivo, the gp41 cytoplasmic tail is essential for efficient Env incorporation into virions.


Subject(s)
HIV Envelope Protein gp120/metabolism , HIV-1/metabolism , Membrane Glycoproteins/metabolism , Cell Line , Gene Expression Regulation, Viral , HIV-1/genetics , Humans , Kinetics , Membrane Glycoproteins/genetics , Mutation , Precipitin Tests , T-Lymphocytes/virology , Transfection , Virus Replication/genetics
16.
J Virol ; 73(12): 10508-13, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10559369

ABSTRACT

Previously we demonstrated that murine retroviral Gag proteins associate with a cellular motor protein, KIF-4. Using the yeast two-hybrid assay, we also found an association of KIF-4 with Gag proteins of Mason-Pfizer monkey virus (MPMV), simian immunodeficiency virus (SIV), and human immunodeficiency virus type 1 (HIV-1). Studies performed with mammalian cell systems confirmed that the HIV-1 Gag protein associates with KIF-4. Soluble cytoplasmic proteins from cells infected with recombinant vaccinia virus expressing the entire Gag-Pol precursor protein of HIV-1 or transfected with HIV-1 molecular clone pNL4-3 were fractionated by sucrose gradient centrifugation and further separated by size-exclusion and anion-exchange chromatographies. KIF-4 and HIV-1 Gag cofractionated in both chromatographic separations. Immunoprecipitation assays have also verified the KIF-4-Gag association. KIF-4 binds mainly to the Gag precursor (Pr55 Gag) and a matrix-capsid processing intermediate (Pr42) but not to other processed Gag products. The binding of Gag is mediated by a domain of KIF-4 proximal to the C terminus. These results, and our previous studies, raise the possibility that KIF-4 may play an important role in retrovirus Gag protein transport.


Subject(s)
Gene Products, gag/metabolism , Kinesins/metabolism , Retroviridae/metabolism , Animals , Avian Sarcoma Viruses , Gene Products, gag/genetics , HIV-1/metabolism , HeLa Cells , Humans , Mason-Pfizer monkey virus , Precipitin Tests , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Simian Immunodeficiency Virus
17.
Genes Dev ; 13(17): 2242-57, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10485847

ABSTRACT

Centrosomes organize the mitotic spindle to ensure accurate segregation of the chromosomes in mitosis. The mechanism that ensures accurate duplication and separation of the centrosomes underlies the fidelity of chromosome segregation, but remains unknown. In Saccharomyces cerevisiae, entry into S phase and separation of spindle pole bodies each require CDC4 and CDC34, which encode components of an SCF (Skp1-cullin-F-box) ubiquitin ligase, but a direct (SCF) connection to the spindle pole body is unknown. Using immunofluorescence microscopy, we show that in mammalian cells the Skp1 protein and the cullin Cul1 are localized to interphase and mitotic centrosomes and to the cytoplasm and nucleus. Deconvolution and immunoelectron microscopy suggest that Skp1 forms an extended pericentriolar structure that may function to organize the centrosome. Purified centrosomes also contain Skp1, and Cul1 modified by the ubiquitin-like molecule NEDD8, suggesting a role for NEDD8 in targeting. Using an in vitro assay for centriole separation in Xenopus extracts, antibodies to Skp1 or Cul1 block separation. Proteasome inhibitors block both centriole separation in vitro and centrosome duplication in Xenopus embryos. We identify candidate centrosomal F-box proteins, suggesting that distinct SCF complexes may direct proteolysis of factors mediating multiple steps in the centrosome cycle.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/genetics , Centrosome/enzymology , F-Box Proteins , Peptide Synthases/metabolism , Ubiquitin-Protein Ligase Complexes , Ubiquitin-Protein Ligases , 3T3 Cells , Anaphase-Promoting Complex-Cyclosome , Animals , CHO Cells , Cell Cycle Proteins/genetics , Centrioles/physiology , Centrioles/ultrastructure , Centrosome/ultrastructure , Cricetinae , F-Box-WD Repeat-Containing Protein 7 , Female , Ligases/genetics , Ligases/metabolism , Mice , NEDD8 Protein , Ovum , S Phase , S-Phase Kinase-Associated Proteins , SKP Cullin F-Box Protein Ligases , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins , Spindle Apparatus/physiology , Spindle Apparatus/ultrastructure , Tissue Extracts/physiology , Ubiquitin-Conjugating Enzymes , Ubiquitins/metabolism , Xenopus laevis
18.
Aust N Z J Psychiatry ; 33(3): 416-21, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10442799

ABSTRACT

OBJECTIVE: This study compared the tolerability and efficacy of paroxetine and amitriptyline in the treatment of depression in general practice. METHODS: In this double-blind, multicentre study conducted in the general practice, patients with depression (Montgomery Asberg Depression Rating Scale [MADRS] score > or = 20) who were regarded as requiring antidepressant therapy were randomly assigned to receive paroxetine (20 mg, n = 184) or amitriptyline (50-100 mg, n = 191) once daily for 9 weeks. RESULTS: More patients completed treatment with paroxetine than with amitriptyline (71.1% vs 56.1%, p = 0.009). Depression rating scores (MADRS and Clinical Global Impression [CGI]) were improved with both agents, but at week 9, paroxetine achieved more favourable scores compared with amitriptyline on MADRS (p=0.019), CGI severity of depression (p=0.044), and CGI efficacy index (p = 0.038). CONCLUSIONS: Depressed patients treated in general practice respond more quickly and are more likely to complete the treatment regimen with paroxetine than with amitriptyline.


Subject(s)
Amitriptyline/therapeutic use , Antidepressive Agents, Second-Generation/therapeutic use , Antidepressive Agents, Tricyclic/therapeutic use , Depressive Disorder/drug therapy , Paroxetine/therapeutic use , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adult , Aged , Aged, 80 and over , Australia , Chi-Square Distribution , Double-Blind Method , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Primary Health Care/statistics & numerical data , Time Factors , Treatment Outcome
19.
J Virol ; 73(6): 4728-37, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10233933

ABSTRACT

We previously characterized mutations in the human immunodeficiency virus type 1 matrix (MA) protein that displayed reduced infectivity in single-round assays, defects in the stable synthesis of viral DNA in infected cells, and impaired endogenous reverse transcriptase activity. The mutants, which contained substitutions in a highly conserved Leu at MA amino acid 20, also increased binding of Gag to membrane. To elucidate further the role of MA in the virus replication cycle, we have characterized a viral revertant of an amino acid 20 mutant (20LK). The revertant virus, which replicates with essentially wild-type kinetics in H9 cells, contains second-site compensatory changes at MA amino acids 73 (E-->K) and 82 (A-->T), while retaining the original 20LK mutation. Single-cycle infectivity assays, performed with luciferase-expressing viruses, show that the 20LK/73EK/82AT triple mutant displays markedly improved infectivity relative to the original 20LK mutant. The stable synthesis of viral DNA in infected cells is also significantly increased compared with that of 20LK DNA. Furthermore, activity of revertant virions in endogenous reverse transcriptase assays is restored to near-wild-type-levels. Interestingly, although 20LK/73EK/82AT reverses the defects in replication kinetics, postentry events, and endogenous reverse transcriptase activity induced by the 20LK mutation, the reversion does not affect the 20LK-imposed increase in Gag membrane binding. Mutants containing single and double amino acid substitutions were constructed, and their growth kinetics were examined. Only virus containing all three changes (20LK/73EK/82AT) grew with significantly accelerated kinetics; 73EK, 73EK/82AT, and 20LK/82AT mutants displayed pronounced defects in virus particle production. Viral core-like complexes were isolated by sucrose density gradient centrifugation of detergent-treated virions. Intriguingly, the protein composition of wild-type and mutant detergent-resistant complexes differed markedly. In wild-type and 20LK complexes, MA was removed following detergent solubilization of the viral membrane. In contrast, in revertant preparations, the majority of MA cosedimented with the detergent-resistant complex. These results suggest that the 20LK/73EK/82AT mutations induced a significant alteration in MA-MA or MA-core interactions.


Subject(s)
Gene Products, gag/metabolism , HIV Reverse Transcriptase/metabolism , HIV-1/physiology , Viral Matrix Proteins/physiology , Amino Acid Sequence , Cell Membrane/metabolism , Detergents/pharmacology , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Precursors/metabolism , Structure-Activity Relationship , Virion/physiology , Virus Replication
20.
J Virol ; 73(5): 4136-44, 1999 May.
Article in English | MEDLINE | ID: mdl-10196310

ABSTRACT

Binding of the human immunodeficiency virus type 1 (HIV-1) Gag protein precursor, Pr55(Gag), to membrane is an indispensable step in virus assembly. Previously, we reported that a matrix (MA) residue 6 substitution (6VR) imposed a virus assembly defect similar to that observed with myristylation-defective mutants, suggesting that the 6VR change impaired membrane binding. Intriguingly, the 6VR mutation had no effect on Gag myristylation. The defective phenotype imposed by 6VR was reversed by changes at other positions in MA, including residue 97. In this study, we use several biochemical methods to demonstrate that the residue 6 mutation, as well as additional substitutions in MA amino acids 7 and 8, reduce membrane binding without affecting N-terminal myristylation. This effect is observed in the context of Pr55(Gag), a truncated Gag containing only MA and CA, and in MA itself. The membrane binding defect imposed by the 6VR mutation is reversed by second-site changes in MA residues 20 and 97, both of which, when present alone, increase membrane binding to levels greater than those for the wild type. Both reduced and enhanced membrane binding imposed by the MA substitutions depend upon the presence of the N-terminal myristate. The results support the myristyl switch model recently proposed for the regulation of Gag membrane binding, according to which membrane binding is determined by the degree of exposure or sequestration of the N-terminal myristate moiety. Alternatively, insertion of the myristate into the lipid bilayer might be a prerequisite event for the function of other distinct MA-encoded membrane binding domains.


Subject(s)
Capsid Proteins , Gene Products, gag/metabolism , Gene Products, gag/physiology , HIV Antigens/physiology , HIV-1/metabolism , Protein Precursors/metabolism , Viral Proteins , Amino Acid Sequence , Capsid/metabolism , Cell Membrane/metabolism , Gene Products, gag/genetics , HIV Antigens/genetics , HIV Antigens/metabolism , HIV-1/genetics , HIV-1/physiology , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis , Myristic Acid/metabolism , Protein Precursors/genetics , Virus Assembly , gag Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...