Human immunodeficiency virus type 1 Vif-derived peptides inhibit the viral protease and arrest virus production

Author(s):  
C. Gilon ◽  
A. Friedler ◽  
L. Baraz ◽  
I. Blumenzweig ◽  
O. Nussinuv ◽  
...  
FEBS Letters ◽  
1998 ◽  
Vol 441 (3) ◽  
pp. 419-426 ◽  
Author(s):  
Lea Baraz ◽  
Assaf Friedler ◽  
Immanuel Blumenzweig ◽  
Orna Nussinuv ◽  
Nissim Chen ◽  
...  

2004 ◽  
Vol 78 (14) ◽  
pp. 7319-7328 ◽  
Author(s):  
Diane M. P. Lawrence ◽  
Linda C. Durham ◽  
Lynnae Schwartz ◽  
Pankaj Seth ◽  
Dragan Maric ◽  
...  

ABSTRACT Although cells of monocytic lineage are the primary source of human immunodeficiency virus type 1 (HIV-1) in the brain, other cell types in the central nervous system, including astrocytes, can harbor a latent or persistent HIV-1 infection. In the present study, we examined whether immature, multipotential human brain-derived progenitor cells (nestin positive) are also permissive for infection. When exposed to IIIB and NL4-3 strains of HIV-1, progenitor cells and progenitor-derived astrocytes became infected, with peak p24 levels of 100 to 500 pg/ml at 3 to 6 days postinfection. After 10 days, virus production was undetectable but could be stimulated by the addition of tumor necrosis factor alpha (TNF-α). To bypass limitations to receptor entry, we compared the fate of infection in these cell populations by transfection with the infectious HIV-1 clone, pNL4-3. Again, transfected progenitors and astrocytes produced virus for 7 days but diminished to low levels beyond 8 days posttransfection. During the nonproductive phase, TNF-α stimulated virus production from progenitors as late as 5 weeks posttransfection. Astrocytes produced 5- to 20-fold more infectious virus (27 ng of p24/106 cells) than progenitors at the peak of 3 days posttransfection. Differentiation of infected progenitors toward an astrocyte phenotype increased virus production to levels consistent with infected astrocytes, suggesting a phenotypic difference in viral replication. Using this cell culture system of multipotential human brain-derived progenitor cells, we provide evidence that progenitor cells may be a reservoir for HIV-1 in the brains of AIDS patients.


2001 ◽  
Vol 82 (11) ◽  
pp. 2719-2733 ◽  
Author(s):  
Martine Bardy ◽  
Bernard Gay ◽  
Stéphanie Pébernard ◽  
Nathalie Chazal ◽  
Marianne Courcoul ◽  
...  

Interactions of human immunodeficiency virus type 1 (HIV-1) Vif protein with various forms of Gag and Gag–Pol precursors expressed in insect cells were investigated in vivo and in vitro by co-encapsidation, co-precipitation and viral protease (PR)-mediated Gag processing assays. Addressing of Gag to the plasma membrane, its budding as extracellular virus-like particles (VLP) and the presence of the p6 domain were apparently not required for Vif encapsidation, as non-N-myristoylated Δp6-Gag and Vif proteins were co-encapsidated into intracellular VLP. Encapsidation of Vif occurred at significantly higher copy numbers in extracellular VLP formed from N-myristoylated, budding-competent Gag–Pol precursors harbouring an inactive PR domain or in chimaeric VLP composed of Gag and Gag–Pol precursors compared with the Vif content of Pr55Gag VLP. Vif encapsidation efficiency did not seem to correlate directly with VLP morphology, since these chimaeric VLP were comparable in size and shape to Pr55Gag VLP. Vif apparently inhibited PR-mediated Pr55Gag processing in vitro, with preferential protection of cleavage sites at the MA–CA and CA–NC junctions. Vif was resistant to PR action in vitro under conditions that allowed full Gag processing, and no direct interaction between Vif and PR was detected in vivo or in vitro. This suggested that inhibition by Vif of PR-mediated Gag processing resulted from interaction of Vif with the Gag substrate and not with the enzyme. Likewise, the higher efficiency of Vif encapsidation by Gag–Pol precursor compared with Pr55Gag was probably not mediated by direct binding of Vif to the Gag–Pol-embedded PR domain, but more likely resulted from a particular conformation of the Gag structural domains of the Gag–Pol precursor.


1993 ◽  
Vol 2 (12) ◽  
pp. 2167-2176 ◽  
Author(s):  
Alfredo G. Tomasselli ◽  
Jean L. Sarcich ◽  
Linda J. Barrett ◽  
Ilene M. Reardon ◽  
W. Jeffrey Howe ◽  
...  

2002 ◽  
Vol 76 (17) ◽  
pp. 8963-8965 ◽  
Author(s):  
Viktor Müller ◽  
Javier Flavio Vigueras-Gómez ◽  
Sebastian Bonhoeffer

ABSTRACT Antiviral therapy induces a rapid drop in human immunodeficiency virus type 1 viremia, but the decline of virus levels decelerates over time. Mathematical modeling demonstrates that the source of residual virus production might be a single compartment of latently infected cells with an extended distribution of activation rates.


1990 ◽  
Vol 172 (4) ◽  
pp. 1035-1042 ◽  
Author(s):  
C D Pauza ◽  
J E Galindo ◽  
D D Richman

High levels of unintegrated viral DNA accumulate during human immunodeficiency virus type 1 (HIV-1) infection of CEM T cells. Reinfection of already infected cells is required to attain these levels and reinfection also promotes the development of HIV-induced cytopathology. Rates of virus production, however, are independent of the accumulation of unintegrated viral DNA. Neutralizing antibody added soon after infection reduced viral DNA levels without appreciably affecting the production of cell-free viral p24 antigen or reverse transcriptase activity. Only 50 pM AZT were required to reduce the accumulation of unintegrated viral DNA by 50% in contrast to the 25 nM required to inhibit virus production by 50%. Cytopathology, as measured by number of syncytia in infected cell cultures, was correlated with highly elevated levels of unintegrated viral DNA. The minimal levels of unintegrated viral DNA present constitutively in the persistently infected HCEM cell line were consonant with the absence of cytopathic effects in these cells. These data demonstrate that inhibiting the reinfection of already infected cells modulates cytopathic HIV-1 infection to a form that is persistent and noncytopathic.


2004 ◽  
Vol 78 (19) ◽  
pp. 10747-10754 ◽  
Author(s):  
Angela Ciuffi ◽  
Gabriela Bleiber ◽  
Miguel Muñoz ◽  
Raquel Martinez ◽  
Corinne Loeuillet ◽  
...  

ABSTRACT Isolated primary human cells from different donors vary in their permissiveness—the ability of cells to be infected and sustain the replication of human immunodeficiency virus type 1 (HIV-1). We used replicating HIV-1 and single-cycle lentivirus vectors in a population approach to identify polymorphic steps during viral replication. We found that phytohemagglutinin-stimulated CD4+ CD45RO+ CD57− T cells from healthy blood donors (n = 128) exhibited a 5.2-log-unit range in virus production. For 20 selected donors representing the spectrum of CD4 T-cell permissiveness, we could attribute up to 42% of the total variance in virus production to entry factors and 48% to postentry steps. Efficacy at key intracellular steps of the replicative cycle (reverse transcription, integration, transcription and splicing, translation, and budding and release) varied from 0.71 to 1.45 log units among donors. However, interindividual differences in transcription efficiency alone accounted for 64 to 83% of the total variance in virus production that was attributable to postentry factors. While vesicular stomatitis virus G protein-mediated fusion was more efficacious than CCR5/CD4 entry, the latter resulted in greater transcriptional activity per proviral copy. The phenotype of provirus transcription was stable over time, indicating that it represents a genetic trait.


Blood ◽  
1991 ◽  
Vol 77 (8) ◽  
pp. 1699-1705
Author(s):  
K Kitano ◽  
CN Abboud ◽  
DH Ryan ◽  
SG Quan ◽  
GC Baldwin ◽  
...  

To define the relationship between human immunodeficiency virus type 1 (HIV-1) infection in hematopoietic stem cells and virus production by their progeny, we performed kinetic studies infecting bone marrow (BM) stem cells and culturing them in the presence of hematopoietic growth factors. CD34-positive (CD34+), CD4-negative (CD4-) BM cells were isolated and infected in vitro with the monocytotropic HIV-1JR-FL strain or the laboratory-maintained HTLV-IIIB strain at a high multiplicity of infection. The cells were susceptible to productive infection only with HIV-1JR-FL, and virus production as measured by p24 protein release was markedly increased (more than fivefold) in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-3 (IL-3). Macrophage CSF (M-CSF) was less stimulatory and granulocyte CSF (G-CSF) had no effect on virus production. Virus production coincided with proliferation of mononuclear phagocytes but was not related to granulocytic proliferation in G-CSF-treated BM cultures. Although peak virus production from GM-CSF-treated macrophages occurred 2 to 3 weeks after infection, peak virus production in infected stem cells was observed 5 to 6 weeks after. Enhancement in virus production had a more rapid onset when CD34+/CD4- cells were cultured in the presence of both GM-CSF and IL-3 for 7 or 14 days. Under these conditions there was a 10-fold enhancement in virus production after 7 days of preincubation and a 50-fold enhancement after 14 days. These data indicate that while the stem cell compartment may be susceptible to infection with a monocytotropic HIV-1 strain, productive and sustained infection is realized only after macrophage differentiation. The lack of effect of G-CSF on virus production is likely because of the limited effect of this hematopoietin on mononuclear phagocyte generation and function.


Sign in / Sign up

Export Citation Format

Share Document