Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Gene Ther ; 16(3): 349-58, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19052634

ABSTRACT

Neural progenitor cells are potential vehicles for delivery of therapeutic agents into the brain. Differentiation-dependent promoters may be useful to target the therapeutic transgene expression to specific neural cell types. Here we explored the potential of vectors based on the foamy virus (FV) for genetic engineering of neural progenitor cells. We demonstrate that FV vectors can mediate stable long-term constitutive expression of the enhanced green fluorescent protein (EGFP) in neural progenitor cells. For differentiation-dependent gene expression, we constructed a FV vector with an internal expression cassette containing the human 2.2 kb promoter (Gfa2) of the astrocyte-specific glial fibrillary acidic protein (GFAP) and sequences encoding EGFP. We show FV-vector-mediated delivery of the Gfa2-egfp transgene into the human neural stem cell line HNSC.100 and differentiation-dependent expression in stably transduced cell populations. Differentiation of the FV-transduced HNSC.100 cells to astrocytes upregulated expression of both the Gfa2-egfp transgene and the native gfap gene, confirming differentiation-dependent activation of the transduced Gfa2 promoter. These results demonstrate that differentiation-dependent gene expression can be achieved by FV-vector-mediated gene transfer to neural progenitor cells. Our findings support the use of FV vectors for the genetic engineering of neural progenitor cells for therapeutic and research applications.


Subject(s)
Genetic Vectors , Neurons/metabolism , Spumavirus/genetics , Stem Cells/metabolism , Transduction, Genetic , Astrocytes/cytology , Astrocytes/physiology , Cell Differentiation/genetics , Cell Line , Gene Expression Regulation/genetics , Genetic Engineering/methods , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Neurons/cytology , Stem Cells/cytology
2.
Nucleic Acids Res ; 33(16): 5139-44, 2005.
Article in English | MEDLINE | ID: mdl-16157863

ABSTRACT

The C-terminal domain (CTD) of mammalian RNA polymerase II consists of 52 repeats of the consensus hepta-peptide YSPTSPS, and links transcription to the processing of pre-mRNA. Although Pol II with a CTD shortened to five repeats (Pol II Delta5) is transcriptionally inactive on chromatin templates, it is not clear whether CTD is required for promoter recognition in vivo. Here, we demonstrate that in the context of chromatin, Pol II Delta5 can bind to the c-myc promoter with the same efficiency as wild type Pol II. However, Pol II Delta5 does not form a stable initiation complex, and does not transcribe promoter proximal sequences. Fluorescence recovery after photobleaching (FRAP) experiments with cells expressing enhanced green fluorescent protein (EGFP)-tagged Delta5 or wildtype Pol II revealed a single, highly mobile Pol II Delta5 fraction whereas wildtype Pol II yielded less mobile fractions. These data suggest that CTD is not required for promoter recognition, but rather for subsequent formation of a stable initiation complex and isomerization to an elongation competent complex.


Subject(s)
Promoter Regions, Genetic , RNA Polymerase II/chemistry , Transcription, Genetic , Binding Sites , Cell Line, Tumor , Cell Nucleus/enzymology , Consensus Sequence , Fluorescence Recovery After Photobleaching , Genes, myc , Green Fluorescent Proteins/analysis , Humans , Protein Structure, Tertiary , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Repetitive Sequences, Amino Acid , Sequence Deletion
3.
Neuropathol Appl Neurobiol ; 29(4): 378-88, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12887598

ABSTRACT

Reactive astrocytosis is a well-documented feature of HIV encephalitis (HIVE), but it is unclear whether restricted infection of astrocytes contributes to this phenomenon. In addition, the part played by reactive and/or infected astrocytes in AIDS-related dementia is not fully understood. In this study of patients at different stages of the human immunodeficiency virus (HIV) infection, who had been treated at most with one antiretroviral drug, reactive astrocytes were identified by immunopositivity for glial fibrillary acidic protein (GFAP) and infected astrocytes by positivity for HIV Nef protein. Results were compared for drug-using AIDS patients with (n=9) and without (n=7) HIVE, for presymptomatic HIV-positive drug users (n=12) and for control HIV-negative subjects (n=20), including a group who used drugs (n=10). GFAP-reactive astrocytes in both grey and white matter were significantly more numerous in HIVE subjects than in each of the other groups but did not correlate with viral load. Nef-positive astrocytes were confined to HIVE cases and to white matter, but were numerous in only one subject who was treatment-naive. Nef-positive microglia were identified in all HIVE cases and in occasional AIDS and presymptomatic subjects who did not have HIVE. The results suggest that astrocytes may form an additional viral reservoir in late HIV infection and may contribute to HIVE. However, the number of GFAP-positive astrocytes was neither increased in pre-AIDS nor in drug abuse, in contrast with microglia which we have shown previously to be up-regulated in both states.


Subject(s)
AIDS Dementia Complex/pathology , Astrocytes/chemistry , Astrocytes/virology , Gene Products, nef/analysis , Glial Fibrillary Acidic Protein/analysis , AIDS Dementia Complex/complications , AIDS Dementia Complex/metabolism , Adult , Astrocytes/pathology , Cell Count , Cognition Disorders/metabolism , Cognition Disorders/pathology , Cognition Disorders/virology , Female , Gliosis/metabolism , Gliosis/pathology , Gliosis/virology , Humans , Immunohistochemistry , Male , Middle Aged , Proviruses , Substance-Related Disorders/complications , Substance-Related Disorders/pathology , Viral Load , nef Gene Products, Human Immunodeficiency Virus
4.
J Cell Sci ; 114(Pt 9): 1717-29, 2001 May.
Article in English | MEDLINE | ID: mdl-11309202

ABSTRACT

Astrocytes are cellular targets for the human immunodeficiency virus (HIV) that limit virus production, owing, at least in part, to the diminished functionality of the viral post-transcriptional stimulatory factor Rev. To understand the trafficking process in astrocytes, we compared nucleocytoplasmic transport of Rev and various proteins with well-characterized nucleocytoplasmic transport features in human astrocytes and control cells (HeLa). Localization and trafficking characteristics of several cellular and viral proteins, as well as nuclear trafficking of classical peptide signals upon microinjection were similar in both cell types, indicating maintenance of general features of nucleocytoplasmic transport in astrocytes. Quantification of fluorescence in living cells expressing Rev fused to green fluorescent protein (GFP) indicated a strong shift in intracellular distribution of Rev in astrocytes, with 50-70% of Rev in the cytoplasm, whereas the cytoplasmic proportion of Rev in HeLa cells is around 10%. The dynamics of nucleocytoplasmic trafficking of Rev were compared in astrocytes and Rev-permissive cells by monitoring migration of Rev-GFP in cell fusions using highly sensitive time-lapse imaging. Nuclear uptake of Rev was dramatically retarded in homo-polykaryons of astrocytes compared with control cells. Diminished nuclear uptake of Rev was also observed in hetero-polykaryons of Rev-permissive cells and astrocytes. These results indicate that astrocytes contain a cytoplasmic activity that interferes with nuclear uptake of Rev. Our studies suggest a model in which Rev is prevented from functioning efficiently in astrocytes by specific alterations of its nucleocytoplasmic trafficking properties.


Subject(s)
Astrocytes/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Gene Products, rev/metabolism , HIV/metabolism , Amino Acid Sequence , Fluorescent Antibody Technique , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/metabolism , Molecular Sequence Data , Nuclear Localization Signals , Protein Transport , Recombinant Fusion Proteins/metabolism , rev Gene Products, Human Immunodeficiency Virus
5.
Genome Res ; 11(3): 333-40, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11230158

ABSTRACT

The publication of the first almost complete sequence of a human chromosome (chromosome 22) is a major milestone in human genomics. Together with the sequence, an excellent annotation of genes was published which certainly will serve as an information resource for numerous future projects. We noted that the annotation did not cover regulatory regions; in particular, no promoter annotation has been provided. Here we present an analysis of the complete published chromosome 22 sequence for promoters. A recent breakthrough in specific in silico prediction of promoter regions enabled us to attempt large-scale prediction of promoter regions on chromosome 22. Scanning of sequence databases revealed only 20 experimentally verified promoters, of which 10 were correctly predicted by our approach. Nearly 40% of our 465 predicted promoter regions are supported by the currently available gene annotation. Promoter finding also provides a biologically meaningful method for "chromosomal scaffolding", by which long genomic sequences can be divided into segments starting with a gene. As one example, the combination of promoter region prediction with exon/intron structure predictions greatly enhances the specificity of de novo gene finding. The present study demonstrates that it is possible to identify promoters in silico on the chromosomal level with sufficient reliability for experimental planning and indicates that a wealth of information about regulatory regions can be extracted from current large-scale (megabase) sequencing projects. Results are available on-line at http://genomatix.gsf.de/chr22/.


Subject(s)
Chromosomes, Human, Pair 22/genetics , Computational Biology , Human Genome Project , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA , Algorithms , Computational Biology/methods , Computational Biology/trends , Forecasting , Humans , Reproducibility of Results , Sequence Analysis, DNA/trends , Software Validation
6.
J Neurovirol ; 6(1): 75-83, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10786999

ABSTRACT

Human immunodeficiency virus (HIV) infection of the central nervous system (CNS) affects primarily microglial cells and astrocytes. Infection of these latter cells occurs independently of CD4 and is characterised by preferential accumulation of 2 Kb mRNA, encoding mostly Nef, and by low levels of 4.5 and 9 Kb RNAs. We have investigated the potential role of chronic HIV infection of human astrocytic cells on the expression of pro-inflammatory cytokines, chemokines and their receptors by comparing the infected TH4-7-5 with its parental uninfected 85HG66 cell lines. Upregulated levels of tumour necrosis factor-alpha (TNF-alpha) and of certain chemokines, namely interleukin-8 (IL-8) and regulated upon activation normal T cell expressed and secreted (RANTES), were observed in the infected versus uninfected cells, whereas monocyte chemotactic protein-1 (MCP-1) was comparably expressed in both cell lines. This pattern of expression was confirmed in primary foetal astrocytes transiently transfected with HIV. In addition, CXCR1, CXCR2 and CCR2b, receptors for IL-8 and MCP-1, respectively, were also found to be upregulated in TH4-7-5 versus 85HG66. CXCR4, the receptor of stromal cell derived factor-1 (SDF-1) and co-receptor for syncytium inducing HIVs, was comparably expressed in infected and uninfected astrocytic cells, whereas CCR5 was not detected in either cell line. Furthermore, treatment of TH4-7-5 cells with TNF-alpha or IL-1beta stimulated RNA and protein secretion of IL-8, MCP-1, and RANTES as well as HIV expression. Thus, our findings suggest that HIV infection of astrocytic cells can contribute to the establishment of a chronic inflammatory state in the CNS, eventually resulting in HIV encephalitis, by increasing the secretion of pro-inflammatory cytokines, such as TNF-alpha and several chemokines. Overexpression of chemokine receptors including CCR2b, CXCR1 and CXCR2 in infected astrocytic cells may contribute to HIV-induced damage of the CNS via autocrine/paracrine activation of astrocytes.


Subject(s)
Astrocytes/metabolism , Chemokine CCL5/metabolism , HIV-1/metabolism , Interleukin-8/metabolism , Receptors, Chemokine/metabolism , Astrocytes/cytology , Astrocytes/virology , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL5/genetics , Chemokine CXCL12 , Chemokines/metabolism , Chemokines, CXC/metabolism , Humans , Inflammation/metabolism , Interleukin-1/metabolism , Interleukin-1/pharmacology , Interleukin-8/genetics , RNA, Messenger/biosynthesis , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects
7.
AIDS ; 13(17): 2331-41, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10597774

ABSTRACT

OBJECTIVE: Nef was shown to be the predominant viral protein expressed in HIV-1-infected astrocytes in vivo and in vitro suggesting a distinct role of Nef in this cell type. Nef-induced activation of T cells is well described, whereas the functional activities of Nef in astrocytes are unknown. Our aim was to examine the effect of Nef on growth properties and activation of astrocytes. DESIGN: Human Nef-expressing astrocytic cell lines were established by stable transfection with different wild-type and mutant nef genes derived from laboratory isolates and brain tissue. METHODS: Nef-expressing astrocytes were characterized in terms of growth properties (proliferation, growth in soft agar, focus formation) and morphology. Apoptotic cell death and expression of activation markers were determined by fluorescent antibody cell sorting. RESULTS: Astrocytic cell lines revealed persistent Nef expression--detectable at the levels of mRNA and protein--and showed altered growth properties and morphology. Elevated expression of activation markers such as glial fibrillary acidic protein and CD88 (complement receptor C5a) was observed; these are regarded as markers for inflammatory processes in the brain. This effect was independent of the nef type or the expression level of the Nef protein. In contrast with previous reports no evidence for increased apoptotic cell death was found in astrocytes expressing Nef stably. CONCLUSIONS: Our findings suggest that Nef changes the cellular properties of astrocytes, thus contributing to astrocyte activation and induction of astrogliosis in the central nervous system of individuals with AIDS.


Subject(s)
Astrocytes/pathology , Astrocytes/virology , Genes, nef , HIV-1/genetics , HIV-1/pathogenicity , Apoptosis , Astrocytes/physiology , Base Sequence , Cell Division , Cell Line , DNA Primers/genetics , Gene Expression , Gene Products, nef/genetics , Glial Fibrillary Acidic Protein/physiology , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , HIV-1/immunology , Humans , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/virology , Virulence/genetics , nef Gene Products, Human Immunodeficiency Virus
8.
J Virol ; 73(10): 8279-89, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10482578

ABSTRACT

Astrocytes are target cells for human immunodeficiency virus type 1 (HIV-1) in the central nervous system with attenuated virus replication in vivo and in vitro. In infected astrocytes, viral gene expression is restricted mainly to nonstructural (early) viral components like Nef, suggesting inhibition of Rev-dependent posttranscriptional processes in these cells. Because of the heterogeneity of astrocytic cells, the objective of this study was to determine whether restriction of HIV-1 Rev-associated activities is a common property of human astrocytes. To this end, we compared the trans activation capacity and intracellular distribution of Rev in four astrocytoma cell lines previously shown to be infectible by HIV-1 and in primary human fetal astrocytes from different sources with Rev-permissive nonglial control cell lines. In all astrocytic cell cultures, the Rev response was reduced to about 10% of that of Rev-permissive control cells. Rev was apparent both in cytoplasmic and in nuclear compartments of living astrocytes, in contrast to the typical nuclear and/or nucleolar localization of Rev in permissive control cells. Nuclear accumulation of Rev in astrocytes was restored by blocking export of Rev. The trans activation capacity and nuclear localization of Tat were not affected in astrocytes. These results demonstrate that inhibition of Rev-dependent posttranscriptional regulation of HIV-1 is a hallmark of human astrocytes and may contribute to suppression of HIV-1 production in these HIV-1 reservoirs. Astrocytes constitute the first example of a human cell type showing an impaired Rev response, indicating that posttranscriptional control of HIV-1 gene expression can be modulated in a cell-dependent manner.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Astrocytes/virology , Gene Products, rev/physiology , HIV-1/physiology , Acquired Immunodeficiency Syndrome/pathology , Cell Line , Humans , Organ Specificity , Transcriptional Activation , Virus Replication , rev Gene Products, Human Immunodeficiency Virus
9.
DNA Seq ; 10(2): 115-9, 1999.
Article in English | MEDLINE | ID: mdl-10376214

ABSTRACT

Pseudoautosomal boundary-like (PABL) elements have been found at transition sites between genomic regions with different GC-contents. A new PAB related sequence was found immediately adjacent to the S71 provirus on human chromosome 18q21.1-2 (officially designated the SSAV1 locus). The S71 PABL element was full-length as defined by comparison with elements identified at the pseudoautosomal boundaries of the sex chromosomes and in the MHC region. The 3'-ends of all PABL elements showed significant homology to functional CpG-islands, indicating that this similarity is a new common feature of PABL elements.


Subject(s)
Chromosomes, Human, Pair 18 , Endogenous Retroviruses/genetics , Base Sequence , DNA , DNA, Complementary , Humans , Molecular Sequence Data , Sequence Homology, Nucleic Acid
11.
J Gen Virol ; 79 ( Pt 4): 731-40, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9568968

ABSTRACT

Antimicrobial peptides are effectors of innate immunity, providing their hosts with rapid non-specific defence against parasitic invaders. In this report, the effects are assessed of two well-characterized antimicrobial amphipathic peptides (melittin and cecropin) on human immunodeficiency virus 1 (HIV-1) replication and gene expression in acutely infected cells at subtoxic concentrations. Production of infectious, cell-free virus was inhibited in a dose-dependent manner, with ID50 values in the range 0.9-1.5 microM for melittin and 2-3 microM for cecropin. Analysis of the effect of melittin on cell-associated virus production revealed decreased levels of Gag antigen and HIV-1 mRNAs. Transient transfection assays with HIV long terminal repeat (LTR)-driven reporter gene plasmids indicated that melittin has a direct suppressive effect on activity of the HIV LTR. HIV LTR activity was also reduced in human cells stably transfected with retroviral expression plasmids for the melittin or cecropin gene. It is concluded that antimicrobial peptides such as melittin and cecropin are capable of inhibiting cell-associated production of HIV-1 by suppressing HIV-1 gene expression.


Subject(s)
Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides , HIV-1/drug effects , Melitten/pharmacology , Peptides/pharmacology , Base Sequence , Cell Line , DNA Primers/genetics , Gene Expression/drug effects , Genes, Viral/drug effects , HIV Long Terminal Repeat/drug effects , HIV-1/genetics , HIV-1/physiology , Humans , Melitten/genetics , Peptides/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Viral/biosynthesis , Transfection , Viral Proteins/biosynthesis , Virus Replication/drug effects , Virus Replication/genetics
12.
Genomics ; 43(1): 52-61, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9226372

ABSTRACT

The current genome sequencing projects reveal megabases of unknown genomic sequences. About 1% of these sequences can be expected to be of retroviral origin. These are often severely deleted or mutated. Therefore, identification of the retroviral origin of these sequences can be very difficult due to the absence of convincing overall sequence similarity. There are also many copies of solo-LTRs (long terminal repeats) distributed throughout genomic sequences. LTR and envelope sequences in general are among the most divergent parts of the retroviral genome and thus especially hard to detect in mutated endogenous sequences. We took advantage of the fact that these retroviral sections contain short highly conserved sequence regions providing retroviral hallmarks even after loss of overall similarity. We defined several sequence elements and peptide motifs within LTR and Env sequences and used these elements to construct models for LTRs and Env proteins of mammalian C-type retroviruses. We then used this strategy to identify successfully the hitherto missing LTRs and an env-like region in the S71 human retroviral sequence. Our approach provides a new strategy for identifying remotely related retroviral sequences in genomic DNA (especially human DNA), of potential significance for the interpretation of genomic sequences obtained from the current large-scale sequencing projects.


Subject(s)
Genome, Human , Genome, Viral , Proviruses/genetics , Retroviridae/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA/genetics , DNA/isolation & purification , DNA Primers/genetics , DNA, Viral/genetics , DNA, Viral/isolation & purification , Gammaretrovirus/genetics , Genes, env , Humans , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Repetitive Sequences, Nucleic Acid , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
14.
Virology ; 224(2): 533-8, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8874513

ABSTRACT

RFB virus is an ecotropic C-type retrovirus isolated from CF-1 mice, in which it is associated with induction of osteomas. Sequence analysis of the RFB provirus revealed no evidence for presence of an oncogene or a recombined env gene. RFB virus is a member of the murine leukemia virus (MuLV) group (RFB MuLV), sharing 97% nucleotide identity with the endogenous ecotropic provirus of AKR mice (Akv). Like Akv, expression of RFB MuLV mRNAs is inducible by dexamethasone treatment, indicating that FRB MuLV also shares transcriptional control signals with Akv. We assessed the pathogenic potential of RFB MuLV in NMRI mice, which, in contrast to CF-1 mice, do not contain endogenous ecotropic retroviruses. RFB MuLV induced osteomas, osteopetrosis, and lymphomas in newborn NMRI mice. Another CF-1 mouse-derived leukemia virus, FBJ MuLV, the helper virus of the FBJ osteosarcoma virus stock, as well as Akv, also induced osteomas, osteopetrosis, and lymphomas in NMRI mice similar to RFB MuLV. These findings indicate that endogenous retroviruses carry a pathogenic potential in hematopoietic tissues and in the skeleton.


Subject(s)
Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/pathogenicity , Lymphoma/virology , Oncogenes , Osteoma/virology , Osteopetrosis/virology , Animals , Cell Transformation, Viral , Gene Products, gag , Leukemia Virus, Murine/metabolism , Mice , Molecular Sequence Data , Point Mutation , Protein Precursors , RNA, Messenger , RNA, Viral
15.
Virology ; 224(1): 256-67, 1996 Oct 01.
Article in English | MEDLINE | ID: mdl-8862420

ABSTRACT

Retroviruses are expressed under the control of viral control regions designated long terminal repeats (LTRs), which contain all signals for transcriptional initiation as well as transcriptional termination. However, retroviral LTRs from different species within a common genus, such as Lentivirus, do not show significant overall sequence homology. We compiled a model of the functional organization of 20 Lentivirus LTRs which we show to recognize all known Lentivirus LTRs. To this end we combined our previously published methods for identification of transcription elements with secondary structure element analysis in a novel modular approach. We deduced descriptions for three new Lentivirus-specific sequence elements present in most of the Lentivirus LTRs but absent in LTRs of other retrovirus families (B, C, D-type, BLV-HTLV, Spuma). Four of the 10 elements defined in our study were primate-specific. We were able to deduce a phylogeny based on our model which agrees in general with the phylogeny derived from the polymerase genes of these viruses. Our model indicated that more than 100 LTRs from the databases are of Lentivirus origin and can be clearly separated from all other LTR types (B, C, D, BLV-HTLV, Spuma). This selectivity appears to be a unique feature of our modular approach.


Subject(s)
Consensus Sequence , Lentivirus/genetics , Repetitive Sequences, Nucleic Acid , Animals , Base Sequence , DNA, Viral , Humans , Lentivirus/classification , Molecular Sequence Data , Phylogeny
17.
Comput Appl Biosci ; 12(3): 197-203, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8872388

ABSTRACT

Advanced sequencing techniques allow rapid deduction of individual amino acid sequences of highly related proteins. Due to their quasi-species nature, viral genomes (e.g. HIV-1) represent one of the most common sources of related proteins. Another example of related proteins are immunoglobulins. Local differences in amino acid conservation are useful indicators of potential domain structures and immunological or functional epitopes prior to structural analysis of proteins. Although variability indices can be calculated by several methods, delineation of boundaries between sequence stretches with similar variability indices is left to the user. We use algorithmic scale-space filtering for delineation of conserved and variable sequence stretches within a protein which is performed on an algorithmic basis avoiding arbitrary assignments. Out method correctly identified variable regions for the human immunoglobulin lambda-chain V-regions (subgroup I). Prediction of the variable regions of the HIV-1 gp120 env protein was in agreement with empirical derived definitions. These examples indicate that our method is useful for the regional assignment of protein variability solely on the basis of amino acid sequences.


Subject(s)
Algorithms , Proteins/genetics , Sequence Alignment/methods , Software , Amino Acid Sequence , Conserved Sequence , Genetic Variation , HIV Envelope Protein gp120/genetics , Humans , Immunoglobulin Variable Region/genetics , Immunoglobulin lambda-Chains/genetics , Molecular Sequence Data , Sequence Homology, Amino Acid
18.
Virology ; 216(1): 245-51, 1996 Feb 01.
Article in English | MEDLINE | ID: mdl-8614997

ABSTRACT

The objective of this study was to assess the inhibitory effect of potential negative regulatory elements on human immunodeficiency virus (HIV)-1 long terminal repeat (LTR) activity. This was carried out by pairwise comparisons of reporter gene activities of HIV-LTR-CAT constructs differing in the presence and absence of nef sequences in transient transfection assays. Parallel transfections were performed in two persistently HIV-infected cell lines and the uninfected parental cell lines. The negative regulatory element (NRE) of the LTR did not suppress HIV LTR activity in any of the cell lines examined. However, a non-LTR-derived fragment of the nef gene had a distinct suppressive effect on activity of the full-length LTR in chronically infected astrocytoma cells. A weaker negative effect of this nef partial sequence (nps) was detected in the other cell lines with constructs lacking the NRE. The nps was capable of suppressing LTR activity in trans in chronically infected astrocytoma cells in a concentration-dependent manner. These results stress a negative role of a non-LTR nef partial sequence in a cell-specific manner. In addition, our data indicate that nps functions in trans with promoters unrelated to HIV LTR such as SV40 early and CMV immediate-early promoters.


Subject(s)
Gene Expression Regulation, Viral , Genes, nef/genetics , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Cell Line , Down-Regulation , Humans
19.
J Virol ; 69(4): 2159-67, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7884864

ABSTRACT

Chronically human immunodeficiency virus type 1 (HIV-1) strain IIIB-infected human TH4-7-5 astrocytoma cells show low-level virus production. Cocultivation of TH4-7-5 cells with myelomonocytic cells led to active virus production in these target cells after a lag period, indicating cell-determined restriction of virus replication in the glial cells. HIV-1 transcript patterns of TH4-7-5 cells contained only a small proportion of Rev-dependent mRNA species, mimicking a Rev-negative phenotype despite the presence of rev mRNAs and protein. Sequencing of the single provirus integrated in TH4-7-5 cells demonstrated that the rev gene and the Rev-responsive element are intact. These results suggested inhibited function of the Rev-regulatory unit in these astrocytoma cells. Transfection of TH4-7-5 cells with a Rev expression plasmid resulted in weak or no induction of proviral p24gag antigen levels compared with the dramatic increase observed in Rev-permissive HeLa cells. Immunofluorescence analysis of TH4-7-5 cells transfected with a rev-expressing plasmid revealed prominent cytoplasmic and nuclear-nucleolar localization of Rev, in contrast to the predominant nuclear-nucleolar localization pattern of Rev in HeLa cells. We conclude that restriction of virus production in TH4-7-5 cells is at least partially due to a block in Rev-dependent posttranscriptional regulation of HIV expression.


Subject(s)
Astrocytes/virology , Gene Products, rev/physiology , HIV-1/physiology , Virus Replication , Amino Acid Sequence , Astrocytoma , Base Sequence , DNA Primers , Gene Products, rev/antagonists & inhibitors , Gene Products, rev/genetics , HIV-1/genetics , Humans , Molecular Sequence Data , Proviruses/genetics , Subcellular Fractions/metabolism , Tumor Cells, Cultured , rev Gene Products, Human Immunodeficiency Virus
20.
Clin Diagn Virol ; 3(1): 61-72, 1995 Jan.
Article in English | MEDLINE | ID: mdl-15566788

ABSTRACT

BACKGROUND: Data concerning the distribution of HIV in the brains of AIDS patients at different stages of viral infection might contribute towards: (1) understanding the route(s) of HIV entry into the brain and virus dissemination within the brain and (2) establishing a possible correlation between the extent of CNS damage and the distribution of virus in AIDS brains. OBJECTIVE: To determine the distribution of HIV-1 genomic DNA within the brains of three deceased AIDS patients by polymerase chain reaction (PCR). STUDY DESIGN: The brains of three deceased AIDS patients were examined. Two brains had limited neuropathologic findings (brains I and II), and one brain (brain III) showed primary HIV-specific neuropathologic damage. Tissues were taken from different locations within each brain, and high molecular weight DNA isolated from the tissues was assessed for HIV-1 genomic DNA by PCR. RESULTS: HIV-1 genomic DNA was found in all three brains, but the amount was low: order of magnitude of 1 viral genome per 1,000 cells. Multiple PCR analyses of DNA from brain I showed that the viral genomic DNA in this brain was non-uniformly distributed; only samples taken from the brainstem were clearly positive for HIV-1. HIV-1 genomic DNA in brain II was found in portions of the lower and upper hemispheres. All but one of the brain III samples were clearly positive for HIV-1, and they had been taken from locations spread throughout this brain. CONCLUSIONS: Our results suggest that in early or latent stages of HIV-infection of the brain, viral genomic DNA is localized at restricted regions. At later stages this DNA is distributed more uniformly throughout the brain. Our data are compatible with the concept of rare infection events followed by viral spreading within brain tissues.

SELECTION OF CITATIONS
SEARCH DETAIL
...