Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
Biochem Pharmacol ; 194: 114816, 2021 12.
Article En | MEDLINE | ID: mdl-34715067

The presence of latent human immunodeficiency virus 1 (HIV-1) in quiescent memory CD4 + T cells represents a major barrier to viral eradication. Proliferation of memory CD4 + T cells is the primary mechanism that leads to persistence of the latent reservoir, despite effective antiretroviral therapy (ART). Memory CD4 + T cells are long-lived and can proliferate through two mechanisms: homeostatic proliferation via γc-cytokine stimulation or antigen-driven proliferation. Therefore, therapeutic modalities that perturb homeostatic and antigen-driven proliferation, combined with ART, represent promising strategies to reduce the latent reservoir. In this study, we investigated a library of FDA-approved oncology drugs to determine their ability to inhibit homeostatic and/or antigen-driven proliferation. We confirmed potential hits by evaluating their effects on proliferation in memory CD4 + T cells from people living with HIV-1 on ART (PLWH) and interrogated downstream signaling of γc-cytokine stimulation. We found that dasatinib and ponatinib, tyrosine kinase inhibitors, and trametinib, a MEK inhibitor, reduced both homeostatic and antigen-driven proliferationby >65%, with a reduction in viability <45%, ex vivo. In memory CD4 + T cells from PLWH, only dasatinib restricted both homeostatic and antigen-driven proliferation and prevented spontaneous rebound, consistent with promoting a smaller reservoir size. We show that dasatinib restricts IL-7 induced proliferation through STAT5 phosphorylation inhibition. Our results establish that the anti-cancer agent dasatinib is an exciting candidate to be used as an anti-proliferative drug in a clinical trial, since it efficiently blocks proliferation and iswell tolerated in patients with chronic myeloid leukemia (CML).


Antigens, Viral , Cell Proliferation/drug effects , Drug Delivery Systems/methods , HIV-1/drug effects , Homeostasis/drug effects , Protein Kinase Inhibitors/administration & dosage , Antigens, Viral/metabolism , Cell Proliferation/physiology , Cells, Cultured , Dasatinib/administration & dosage , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/immunology , HIV-1/metabolism , Homeostasis/physiology , Humans , Imidazoles/administration & dosage , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Pyridazines/administration & dosage , Pyridones/administration & dosage , Pyrimidinones/administration & dosage
2.
Cell Death Differ ; 12(4): 326-34, 2005 Apr.
Article En | MEDLINE | ID: mdl-15650754

The human immunodeficiency virus type-1 (HIV-1) accessory gene vpr encodes a conserved 96-amino-acid protein that is necessary and sufficient for the HIV-1-induced block of cellular proliferation. Expression of vpr in CD4+ lymphocytes results in G2 arrest, followed by apoptosis. In a previous study, we identified the ataxia telangiectasia-mutated (ATM) and Rad3-related protein (ATR) as a cellular factor that mediates Vpr-induced cell cycle arrest. In the present study, we report that the breast cancer-associated protein-1 (BRCA1), a known target of ATR, is activated in the presence of Vpr. In addition, the gene encoding the growth arrest and DNA damage-45 protein alpha (GADD45alpha), a known transcriptional target of BRCA1, is upregulated by Vpr in an ATR-dependent manner. We demonstrate that RNAi-mediated silencing of either ATR or GADD45alpha leads to nearly complete suppression of the proapoptotic effect of Vpr. Our results support a model in which Vpr-induced apoptosis is mediated via ATR phosphorylation of BRCA1, and consequent upregulation of GADD45alpha.


Apoptosis/physiology , Cell Cycle Proteins/metabolism , Gene Products, vpr/metabolism , HIV-1/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ataxia Telangiectasia Mutated Proteins , BRCA1 Protein/metabolism , CD4-Positive T-Lymphocytes/metabolism , Caspase Inhibitors , G2 Phase/physiology , HeLa Cells , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Serine/metabolism , T-Lymphocyte Subsets/metabolism , vpr Gene Products, Human Immunodeficiency Virus
3.
An Pediatr (Barc) ; 60(2): 125-32, 2004 Feb.
Article Es | MEDLINE | ID: mdl-14757015

OBJECTIVE: To assess the burden (incidence, treatment and complications) of acute otitis media (AOM) and otitis media with effusion (OME) in children younger than 5 years of age from Valencia, Spain. SUBJECT AND METHODS: We performed a retrospective cohort study of 1,399 children followed-up for the first 5 years of life. Seventeen pediatricians reviewed the medical records of their patients born in 1995 and 1996 and followed-up from birth until the age of 5 years. For each child, the number of otitis episodes, treatment, complications, and surgical interventions was obtained. RESULTS: There were 2,961 episodes of AOM in the first 5 years of life (2.23 cases/child). Four hundred seventy-six cases (16.1 %) occurred before 1 year of age and 1,346 between the first and second year of life (45.5 %). By the third year of life, 59.8 % had had at least one episode. In most children (80.9 %), diagnosis was made in primary care and required a median of 1.81 visits/episode for follow-up. A total of 94.5 % were treated with antibiotics (amoxicillin-clavulanate 38.8 %, cefuroxime 14.3 %, clarithromycin 8.2 % and amoxicillin 5.9 %) and 8.5 % required a change of antibiotic therapy. Two hundred seventeen children (15.2 %) had at least one episode of OME. Twenty-six patients (1.8 %, 95 % CI: 1.2-2.7 %) required insertion of ventilation tubes. Twenty-four patients (1.7 %) had secondary hypoacusis. There was one case of meningitis and two cases of chronic otorrhea. No cases of mastoiditis were recorded. CONCLUSIONS: The incidence of AOM in Valencia is 40,014 episodes/100,000 children younger than 5 years/year (95 % CI: 39,700-40,300). It represents a significant burden due to the large number of visits, antibiotic use, associated surgical procedures and need for auditory rehabilitation.


Otitis Media/epidemiology , Acute Disease , Child, Preschool , Female , Follow-Up Studies , Humans , Incidence , Infant , Infant, Newborn , Male , Otitis Media/complications , Otitis Media/drug therapy , Retrospective Studies , Spain/epidemiology
4.
An. pediatr. (2003, Ed. impr.) ; 60(2): 125-132, feb. 2004.
Article Es | IBECS | ID: ibc-29519

Objetivo: Estimar el impacto (incidencia, tratamiento y complicaciones) de la otitis media aguda (OMA) y la otitis media serosa (OMS) en los niños menores de 5 años de la Comunidad Valenciana (España). Sujetos y métodos: Cohorte retrospectiva de 1.399 niños seguidos durante los primeros 5 años de vida. Diecisiete pediatras revisaron las historias clínicas de sus pacientes nacidos en 1995 y 1996, controlados desde su nacimiento hasta los 5 años. Se obtuvo de cada niño el número de episodios de otitis, tratamiento, complicaciones e intervenciones quirúrgicas requeridas. Resultados: Hubo 2.961 episodios de OMA en los primeros 5 años de vida (2,23 casos/niño), 476 casos (16,1 por ciento) antes del año y 1.346 entre el primer y segundo año (45,5 por ciento). Al tercer año, el 59,8 por ciento habían presentado al menos un episodio. La mayoría de casos (80,9 por ciento) se diagnosticaron en atención primaria, con 1,81 visitas/episodio de media para su seguimiento. El 94,5 por ciento de los episodios fueron tratados con antibiótico (amoxicilina-ácido clavulánico 38,8 por ciento, cefuroxima-axetilo, 14,3 por ciento; claritromicina, 8,2 por ciento, y amoxicilina 5,9 por ciento). El 8,5 por ciento requirió cambio a un segundo antibiótico. Presentaron al menos un episodio de OMS 217 niños (15,2 por ciento). Necesitaron implantación de tubos de ventilación 26 pacientes (1,8 por ciento, intervalo de confianza del 95 por ciento (IC 95 por ciento), 1,2-2,7). Presentaron hipoacusia secundaria 24 niños (1,7 por ciento). Hubo un caso de meningitis y dos de otorrea crónica. No hubo mastoiditis. Conclusiones: La incidencia de OMA en la Comunidad Valenciana es de 40.014 episodios/100.000 niños menores de 5 años/año (IC 95 por ciento, 39.700-40.300). Supone un gran impacto sanitario por el elevado número de visitas, uso antibiótico, cirugía asociada y necesidad de rehabilitación auditiva (AU)


Child, Preschool , Male , Infant, Newborn , Infant , Female , Humans , Spain , Incidence , Otitis Media , Retrospective Studies , Acute Disease , Follow-Up Studies
5.
Cell Mol Biol (Noisy-le-grand) ; 49(7): 1151-6, 2003 Nov.
Article En | MEDLINE | ID: mdl-14682398

Gene therapy is a promising approach for the treatment of neurological disorders. However, current approaches to gene transfer in the central nervous system (CNS) are limited by the lack of effective, but non-invasive methods to deliver transgenes across the blood-brain barrier (BBB). In an effort to begin to explore the use of migratory monocytes as vehicles for delivery of therapeutic and antiviral genes into the CNS, we have utilized three HIV-based transfer vectors encoding cis-acting elements but lacking either structural genes (gag/pol and env), most accessory genes (vif, vpr and nef) and/or rev. These defective lentiviral vectors (DLV) encode the green fluorescent protein (GFP), display potent antiviral activity in CD4+ lymphocytes and can be mobilized by wild-type HIV-1 DLV were generated by transient transfection of 293T cells. Vector titers ranged from 4.2-6.6 x 10(6) infectious units (IU)/ml prior to concentration (by ultracentrifugation) and were equal to or higher than 1 x 10(9) IU/ml after concentration. Primary human monocyte-derived macrophages (MDM) were exposed to DLV resulting in efficiencies of transduction ranging from 14 to 26%. GFP expression in transduced MDM remained stable for more than 8 weeks without apparent cytopathic effect. Given the previously reported antiviral activities of these DLV and their lack of cytopathic effects on primary MDM, it may be possible to use these vectors to inhibit HIV-1 replication within the CNS.


Gene Transfer Techniques , Genes, Viral/genetics , Lentivirus/genetics , Macrophages/cytology , Virus Replication/physiology , Cells, Cultured , Cloning, Molecular , Green Fluorescent Proteins , Humans , Lentivirus/physiology , Luminescent Proteins/metabolism , Macrophages/virology
6.
Apoptosis ; 8(1): 71-9, 2003 Jan.
Article En | MEDLINE | ID: mdl-12510154

The human survivin gene belongs to the family of inhibitor of apoptosis proteins (IAP) and is involved in apoptosis inhibition and regulation of cell division. The survivin gene is the only member of the IAP family whose expression is known to be regulated through the cell cycle. Survivin expression reaches the highest levels during the G(2)/M transition and then is rapidly degraded during the G(1) phase. Here we report that the human immunodeficiency virus type 1 (HIV-1) upregulates Survivin expression via survivin promoter transactivation. Vpr, an HIV-1 accessory protein that induces cell cycle arrest in G(2)/M, is necessary and sufficient for this effect. Blocking Vpr-induced G(2)/M arrest leads to elimination of the survivin promoter transactivation by Vpr. Our results suggest that Survivin may be actively involved in regulating cell viability during HIV-1 infection.


Gene Products, vpr/metabolism , Microtubule-Associated Proteins/biosynthesis , Blotting, Western , Caffeine/pharmacology , Caspase 3 , Caspases/metabolism , Cell Cycle , Cell Line , Cell Survival , G1 Phase , G2 Phase , Genes, Reporter , Genetic Vectors/genetics , Green Fluorescent Proteins , HIV Infections , HIV-1/metabolism , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Luciferases/metabolism , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Models, Genetic , Neoplasm Proteins , Plasmids/metabolism , Promoter Regions, Genetic , Survivin , Time Factors , Transcriptional Activation , Transfection , Up-Regulation , beta-Galactosidase/metabolism , vpr Gene Products, Human Immunodeficiency Virus
7.
Expert Opin Biol Ther ; 1(1): 17-40, 2001 Jan.
Article En | MEDLINE | ID: mdl-11727544

The aim of gene therapy is to modify the genetic material of living cells to achieve therapeutic benefit. Gene therapy involves the insertion of a functional gene into a cell, to replace an absent or defective gene, or to fight an infectious agent or a tumour. At present, a wide variety of somatic tissues are being explored for the introduction of foreign genes with a view towards treatment. A prime requirement for successful gene therapy is the sustained expression of the therapeutic gene without any adverse effect on the recipient. A highly desirable vector would be generated at high titres, integrate into target cells (including non-dividing cells) and have little or no associated immune reactions. Lentiviruses have the ability to infect dividing and non-dividing cells and, therefore, constitute ideal candidates for development of vectors for gene therapy. This review presents a description of available lentiviral vectors, including vector design, applications to disease treatment and safety considerations. In addition, general aspects of the biology of lentiviruses with relevance to vector development will be discussed. Recent investigations have revealed that foamy viruses, another group of retroviruses, are also capable of infecting non-dividing cells. Thus, foamy virus vectors are actively being developed in parallel to lentivirus vectors. This review will also include various aspects of the biology of foamy viruses with relevance to vector development.


Genetic Therapy/methods , Genetic Vectors , Lentivirus/genetics , Spumavirus/genetics , Animals , Genetic Therapy/adverse effects , HIV-1/genetics , Humans , Models, Genetic , Transcription, Genetic , Transgenes
8.
Mol Ther ; 4(4): 375-82, 2001 Oct.
Article En | MEDLINE | ID: mdl-11592842

Lentiviral vectors, so far, have been optimized for the expression of a single open reading frame. Certain practical applications of gene therapy will, however, require expression of multiple genes. The goal of this study was to explore the feasibility of directing expression of two marker genes from a lentiviral vector. We designed two types of multigene lentiviral vectors. First, we used a strategy based on the natural splicing signals of HIV-1, by which multiple mRNAs are generated from a single transcriptional unit. A second strategy was construction of a polycistronic mRNA using a translational cis-acting element, the encephalomyocarditis virus internal ribosome entry site (IRES). Our studies show that the inclusion of multiple genes in lentiviral vectors does not result in reduction in virus titers or in the loss of ability to infect nondividing cells. We introduced mutations in tat and/or rev to test whether splicing modulates the relative levels of expression of reporter genes. We also developed a truncated version of tat, which is devoid of the apoptosis-associated domain. Inclusion of this tat mutant in a lentiviral vector resulted in the generation of virus with titers similar to those of lentivirus vectors expressing wild-type tat.


Alternative Splicing/genetics , Gene Expression Regulation, Viral , Genetic Vectors/genetics , Lentivirus/genetics , Protein Biosynthesis/genetics , Transduction, Genetic/methods , Transgenes/genetics , Cell Division , Encephalomyocarditis virus/genetics , Flow Cytometry , Gene Products, tat/chemistry , Gene Products, tat/genetics , Genes/genetics , Genes, Reporter/genetics , Genes, rev/genetics , Genes, tat/genetics , HIV-1/genetics , HeLa Cells , Humans , Lentivirus/physiology , Microscopy, Fluorescence , Mutation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Ribosomes/metabolism , Transcriptional Activation , tat Gene Products, Human Immunodeficiency Virus
9.
J Virol ; 75(20): 9671-8, 2001 Oct.
Article En | MEDLINE | ID: mdl-11559799

Cellular tRNA(Lys)(3) serves as the primer for reverse transcription of human immunodeficiency virus type 1 (HIV-1). tRNA(Lys)(3) interacts directly with HIV-1 reverse transcriptase (RT), is packaged into viral particles, and anneals to the primer-binding site (PBS) of the HIV-1 genome in order to initiate reverse transcription. Residue A58 of tRNA(Lys)(3), which lies outside the PBS-complementary region, is posttranscriptionally methylated to form 1-methyladenosine 58 (M(1)A58). This methylation is thought to serve as a pause signal for plus-strand strong-stop DNA synthesis during reverse transcription. However, formal proof that the methylation is necessary for the pausing of RT has not been obtained in vivo. In the present study, we investigated the role of tRNA(Lys)(3) residue A58 in the replication cycle of HIV-1 in living cells. We have developed a mutant tRNA(Lys)(3) derivative, tRNA(Lys)(3)A58U, in which A58 was replaced by U. This mutant tRNA was expressed in CEM cells. We demonstrate that the presence of M(1)A58 is necessary for the appropriate termination of plus-strand strong-stop DNA synthesis and that the absence of M(1)A58 allows RT to read the tRNA sequences beyond residue 58. In addition, we show that replacement of M(1)A58 with U inhibits the replication of HIV-1 in vivo. These results highlight the importance of tRNA primer residue A58 in the reverse transcription process. Inhibition of reverse transcription with mutant tRNA primers constitutes a novel approach for therapeutic intervention against HIV-1.


Adenosine/analogs & derivatives , HIV-1/genetics , RNA, Transfer, Amino Acyl/genetics , Adenosine/metabolism , Base Sequence , Cell Line , DNA, Viral/biosynthesis , HIV-1/metabolism , Humans , Methylation , Mutagenesis, Site-Directed , RNA, Transfer, Amino Acyl/metabolism , T-Lymphocytes/virology , Transcription, Genetic , Transfection , Virus Replication
10.
Mol Ther ; 3(6): 928-39, 2001 Jun.
Article En | MEDLINE | ID: mdl-11407907

Gene therapy against HIV infection should involve vector-mediated delivery of anti-HIV therapeutic genes into T-lymphocytes and macrophages or, alternatively, hematopoietic progenitors. Transduction of mature cells with defective vectors would have limited success because the vector would disappear with cell turnover. However, if a vector could be trafficked by wild-type HIV, initial transduction of a majority of the population would not be required, as the vector would be able to spread. We describe HIV-1-based lentiviral vectors that are efficiently packaged and trafficked by HIV-1, allowing a small number of cells initially transduced to spread the vector within a nontransduced cell population. We examined whether the presence or absence of the rev gene and the Rev-responsive element (RRE) would have a noticeable effect on the ability of lentiviral vectors to be trafficked and to inhibit HIV-1 replication. We found that replacement of rev/RRE with a constitutive transport element from Mason-Pfizer monkey virus had no apparent effect on trafficking and did not change the intrinsic inhibitory abilities of the vectors. We also constructed a rev/RRE-independent HIV-1-derived vector carrying a trans-dominant negative mutant of HIV-1 Rev, RevM10. This vector was less efficiently trafficked by HIV-1 and, despite the presence of an anti-HIV-1 gene, RevM10, was less efficient at inhibiting HIV-1 replication when introduced into a target T-cell population.


Gene Products, rev/metabolism , Genetic Vectors , HIV-1/physiology , Lentivirus/genetics , Virus Replication/genetics , Cells, Cultured , Defective Viruses , Gene Products, rev/antagonists & inhibitors , Gene Transfer Techniques , Genes, env/physiology , Genes, rev/physiology , Genetic Therapy/methods , HIV-1/genetics , HIV-1/growth & development , Humans , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Transduction, Genetic , Tumor Cells, Cultured , Virus Assembly , rev Gene Products, Human Immunodeficiency Virus
11.
Hum Gene Ther ; 12(7): 847-57, 2001 May 01.
Article En | MEDLINE | ID: mdl-11339901

Lentiviral vectors are attractive candidates for gene therapy because of their ability to integrate into nondividing cells. To date, conventional HIV-1-based vectors can be produced at higher titers, but concerns regarding their safety for human use exist because of the possibility of recombination leading to production of infectious virions with pathogenic potential. Development of lentivirus vectors based on nonhuman lentiviruses constitutes an active area of research. We described a novel HIV-SIV hybrid vector system in which an HIV-1-derived transfer vector is encapsidated by SIVmac1A11 core particles and pseudotyped with VSV glycoprotein G. In an effort to further develop this vector system, we modified the packaging plasmid by deletion of the SIV accessory genes. Specifically, versions of the packaging plasmid (SIVpack) lacking vif, vpr, vpx, and/or nef were constructed. Our results indicate that, as with HIV-1-based packaging plasmids, deletion of accessory genes has no significant effect on transduction in either dividing or nondividing cells. The SIV packaging plasmid was also modified with regard to the requirement for RRE and rev. Deletion of the RRE and rev from SIVpack led to dramatic loss of transduction ability. Introduction of the 5' LTR from the spleen necrosis virus to packaging plasmids lacking RRE/Rev was then sufficient to fully restore vector titer. A minimal SIV transfer vector was also developed, which does not require RRE/Rev and exhibits no reduction in transduction efficiency in two packaging systems. The SIV-based vector system described here recapitulates the biological properties of minimal HIV-1-derived systems and is expected to provide an added level of safety for human gene transfer. We suggest that the SIV-derived vector system will also be useful to deliver anti-HIV-1 gene therapy reagents that would inhibit an HIV-1-derived vector.


Gene Products, rev/physiology , Genetic Therapy/methods , Genetic Vectors/genetics , HIV-1/genetics , Membrane Glycoproteins , Simian Immunodeficiency Virus/genetics , Cell Division , Cell Line , Cloning, Molecular , DNA, Recombinant/genetics , DNA, Viral/genetics , Gene Deletion , Gene Products, rev/genetics , Genes, Viral/genetics , HIV Long Terminal Repeat/genetics , HIV-1/growth & development , HIV-1/metabolism , Humans , Response Elements/genetics , Simian Immunodeficiency Virus/growth & development , Simian Immunodeficiency Virus/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Assembly , rev Gene Products, Human Immunodeficiency Virus
12.
Apoptosis ; 6(1-2): 103-16, 2001.
Article En | MEDLINE | ID: mdl-11321033

Infection with the human immunodeficiency virus type 1 (HIV-1) leads to progressive immunodeficiency and onset of opportunistic infections and neoplasms. The loss of immune competence is associated with declines in both the functionality and the number of CD4+ lymphocytes. Multiple mechanisms have been proposed to explain death and dysfunction of CD4+ T-cells. The mechanisms of HIV-1-mediated cell death which are relevant in vivo are unclear at present. However, in vitro explorations on the cytopathic effects of HIV-1 have yielded a wealth of potential triggering events, and signaling and effector pathways leading to apoptosis. The types of pro- and anti-apoptotic stimuli that have been associated with HIV-1 are multiple and often appear overlapping or even contradictory. This review focuses on the various molecular determinants from HIV-1 that play a role in induction of apoptosis in T-lymphocytes. Special attention is devoted to the viral genes, env, nef, tat and vpr, for which a significant body of literature on apotosis-related effects is available.


Acquired Immunodeficiency Syndrome/pathology , Apoptosis , HIV-1 , Acquired Immunodeficiency Syndrome/virology , Humans , T-Lymphocytes/pathology , T-Lymphocytes/virology , Virus Replication
13.
J Virol ; 75(8): 3791-801, 2001 Apr.
Article En | MEDLINE | ID: mdl-11264368

All primate lentiviruses known to date contain one or two open reading frames with homology to the human immunodeficiency virus type 1 (HIV-1) vpr gene. HIV-1 vpr encodes a 96-amino-acid protein with multiple functions in the viral life cycle. These functions include modulation of the viral replication kinetics, transactivation of the long terminal repeat, participation in the nuclear import of preintegration complexes, induction of G2 arrest, and induction of apoptosis. The simian immunodeficiency virus (SIV) that infects African green monkeys (SIVagm) contains a vpr homologue, which encodes a 118-amino-acid protein. SIVagm vpr is structurally and functionally related to HIV-1 vpr. The present study focuses on how three specific functions (transactivation, induction of G2 arrest, and induction of apoptosis) are related to one another at a functional level, for HIV-1 and SIVagm vpr. While our study supports previous reports demonstrating a causal relationship between induction of G2 arrest and transactivation for HIV-1 vpr, we demonstrate that the same is not true for SIVagm vpr. Transactivation by SIVagm vpr is independent of cell cycle perturbation. In addition, we show that induction of G2 arrest is necessary for the induction of apoptosis by HIV-1 vpr but that the induction of apoptosis by SIVagm vpr is cell cycle independent. Finally, while SIVagm vpr retains its transactivation function in human cells, it is unable to induce G2 arrest or apoptosis in such cells, suggesting that the cytopathic effects of SIVagm vpr are species specific. Taken together, our results suggest that while the multiple functions of vpr are conserved between HIV-1 and SIVagm, the mechanisms leading to the execution of such functions are divergent.


Apoptosis , Cell Cycle , Gene Products, vpr/metabolism , HIV-1 , Simian Immunodeficiency Virus , Transcriptional Activation , Active Transport, Cell Nucleus , Animals , Apoptosis/drug effects , COS Cells , Caffeine/pharmacology , Cell Cycle/drug effects , Chlorocebus aethiops , Flow Cytometry , G2 Phase/drug effects , Gene Products, vpr/genetics , Genes, vpr/genetics , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Humans , Mitosis/drug effects , Models, Biological , Paclitaxel/pharmacology , Simian Immunodeficiency Virus/genetics , Transcriptional Activation/drug effects , Transduction, Genetic , Tumor Cells, Cultured , vpr Gene Products, Human Immunodeficiency Virus
14.
AIDS Res Hum Retroviruses ; 16(16): 1771-6, 2000 Nov 01.
Article En | MEDLINE | ID: mdl-11080825

Infection with human T cell leukemia virus type 1 (HTLV-1) can result in the development of HAM/TSP, a nonfatal, chronic inflammatory disease involving neuronal degeneration and demyelination of the central nervous system. Elevated levels of the proinflammatory cytokines tumor necrosis factor alpha (TNF-alpha), interleukin-6 (IL-6), and IL-1 observed in the cerebrospinal fluid of HAM-TSP patients suggest that cytokine dysregulation within the CNS is involved in neuropathogenesis. HTLV-1 infection and enhanced expression of TNF-alpha by microglial cells, astrocytes, and macrophages has been hypothesized to lead to the destruction of myelin and oligodendrocytes in the CNS. Although the association of HTLV-2 infection and development of neurological disease is more tenuous, HTLV-2 has also been found to be associated with peripheral neuropathies. To investigate the roles of HTLV Tax(1) and Tax(2) in the induction of cytokine disregulation in these cell types, we are currently developing gene delivery vectors based on human immunodeficiency virus type-1 (HIV-1) capable of stably coexpressing the HTLV-1 or -2 tax and eGFP reporter genes in primary human cells. Transduction frequencies of up to 50%, as assessed by eGFP expression, can be achieved in human monocyte-derived macrophages and in explanted cultures of human microglia. Preliminary data suggest that Tax(1) expression is sufficient to up-regulate the proinflammatory cytokine profile in explanted human microglial cells. Future experiments will compare and evaluate the effect of tax(1) and tax(2) gene expression on the cellular proinflammatory cytokine expression profile, as well as demonstrate the effects of transducing human fetal astrocytes and PBMC-derived macrophages.


Astrocytes/virology , Gene Products, tax/genetics , Genetic Vectors/genetics , Lentivirus/genetics , Microglia/virology , Transduction, Genetic , Astrocytes/cytology , Astrocytes/metabolism , Cell Line , Cells, Cultured , Gene Products, tax/metabolism , Green Fluorescent Proteins , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Macrophages/metabolism , Microglia/cytology , Microglia/metabolism , Monocytes/cytology
15.
Obstet Gynecol ; 95(1): 141-6, 2000 Jan.
Article En | MEDLINE | ID: mdl-10636517

OBJECTIVE: To determine the ability of the human immunodeficiency virus type 1 (HIV-1) gene vpr to induce cell-cycle arrest in cervical cancer cells with or without human papillomavirus (HPV) type 16 E6 or E7 expression. METHODS: High- and low-level expression vectors for vpr (designated pVPR(HIGH) and pVPR(LOW), respectively) were used in conjunction with HPV-16 E6 or E7 vectors to transfect HPV-negative C33A cervical cancer cells. Vpr expression vectors encode a cell surface marker gene, murine Thy-1, for specific detection of transfected cells. Dual staining for the surface molecule Thy-1 and DNA content was used to determine cell-cycle profile and G2-phase arrest. RESULTS: C33A cells not expressing HPV-16 E6 showed some but not maximal G2-phase arrest when transfected with pVPR(HIGH) alone (43.2% of cells in the G2 phase). Addition of HPV-16 E6 or E6 plus E7 to pVPR(HIGH) substantially increased the percentages of cells in the G2 phase (51.3% and 53.0%, respectively). Cotransfection with pVPR(HIGH) and HPV-16 E7 did not increase significantly the percentage of cells in the G2 phase compared with pVPR(HIGH) alone (40.6% versus 43.2%). In transfections involving pVPR(LOW), a slight degree of G2-phase arrest was observed when Vpr was expressed alone (29.0% of cells in the G2 phase) or in cotransfection with HPV-16 E7 (33.2% of cells), and G2-phase arrest was augmented with the addition of HPV-16 E6 (41.7%) or E6 plus E7 (45.7%). CONCLUSION: Cervical cancer cells are susceptible to cell-cycle arrest induced by HIV-1 vpr. This effect is exacerbated by coexpression of HPV-16 E6, although E6 alone is incapable of inducing any detectable G2-phase arrest, suggesting that E6 and VPR share links in cell-cycle signaling pathways.


Cell Cycle , Gene Expression , Gene Products, vpr/metabolism , HIV-1/genetics , Papillomaviridae/isolation & purification , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Cells, Cultured , Female , Flow Cytometry , G2 Phase , Humans , Papillomavirus Infections/metabolism , Transcription, Genetic , Transfection , Tumor Virus Infections/metabolism , Uterine Cervical Neoplasms/metabolism , vpr Gene Products, Human Immunodeficiency Virus
16.
AIDS Res Hum Retroviruses ; 15(17): 1563-71, 1999 Nov 20.
Article En | MEDLINE | ID: mdl-10580407

Sera from highly selected HIV-1-positive patients are known to have the ability to neutralize a diverse array of primary isolates of HIV-1. The human osteosarcoma cell line that expresses CD4 and chemokine receptors (GHOST cells) was adapted to study HIV-1 neutralization in 37 HIV-1-infected individuals who were selected because of slow disease progression or nonprogression. Many of these individuals were receiving combination drug therapy. Molecularly cloned HIV-1 JR-FL and NL4-3 viruses were used as prototypes to define assay conditions. Sera were then tested at a 1:40 dilution against six additional primary isolates, three of which utilized CCR5 and three of which used both CCR5 and CXCR4. The assay was highly reproducible and independent of viral input titer, with a readout at 48 hr equivalent to that at later time points. As previously reported, neutralization sensitivity was entirely independent of coreceptor usage. Only a few sera from slow progressors were able to neutralize a broad array of primary isolates at a 1:40 dilution, and the best clinical predictor of broadly neutralizing antibody for primary isolates was the present use of antiretroviral agents. In further studies it was found that purified antibody accounted for the majority of the measured neutralization. However, experiments with exogenous addition of antiviral agents showed that the use of nucleosides also greatly contributed to the measured neutralization in some patients. Measurement of neutralization of HIV-1 primary isolates by sera from patients receiving antiretroviral therapy must be carried out with some caution.


Anti-HIV Agents/therapeutic use , HIV Antibodies/immunology , HIV Infections/drug therapy , HIV Infections/immunology , HIV Long-Term Survivors , Antibody Specificity , CD4 Antigens/metabolism , Cell Line , Cohort Studies , Green Fluorescent Proteins , HIV Antibodies/metabolism , HIV Infections/virology , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Luminescent Proteins/metabolism , Neutralization Tests/methods , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , Receptors, Chemokine/metabolism , Species Specificity
17.
Exp Cell Res ; 251(1): 156-65, 1999 Aug 25.
Article En | MEDLINE | ID: mdl-10438581

The vpr gene from the human immunodeficiency virus type-1 (HIV-1) encodes a 14-kDa protein that prevents cell proliferation by causing a block in the G(2) phase of the cell cycle. This cellular function of vpr is conserved in evolution because other primate lentiviruses, including HIV-2, SIV(mac), and SIV(agm) encode related genes that also induce G(2) arrest. After G(2) arrest, cells expressing vpr undergo apoptosis. The signaling pathways that result in vpr-induced cell cycle arrest and apoptosis have yet to be determined. The p53 tumor suppressor protein is involved in signaling pathways leading to cell cycle arrest and apoptosis in a variety of cell types. In this work, we examine the potential role of p53 in mediating cell cycle block and/or apoptosis by HIV-1 vpr and demonstrate that both phenomena occur independently of the presence and function of p53. Caspases are common mediators of apoptosis. We examined the potential role of caspases in mediating vpr-induced apoptosis by treating vpr-expressing cells with Boc-D-FMK, a broad spectrum, irreversible inhibitor of the caspase family. Boc-D-FMK significantly reduced the numbers of apoptotic cells induced by vpr. Therefore, we conclude that vpr-induced apoptosis is effected via the activation of caspases.


Apoptosis , Caspases/metabolism , Cell Cycle , Gene Products, vpr/metabolism , HIV-1/physiology , Tumor Suppressor Protein p53/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , COS Cells , Caspase Inhibitors , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Fragmentation , Doxorubicin/pharmacology , Enzyme Activation , G2 Phase/drug effects , G2 Phase/radiation effects , Gamma Rays , Gene Products, vpr/genetics , Genetic Vectors/genetics , HIV-1/genetics , Humans , In Situ Nick-End Labeling , Mechlorethamine/pharmacology , Signal Transduction/drug effects , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , vpr Gene Products, Human Immunodeficiency Virus
18.
Front Biosci ; 4: D481-96, 1999 Jun 01.
Article En | MEDLINE | ID: mdl-10352135

Gene therapy is a novel method under investigation for the treatment of genetic, metabolic and neurologic diseases, cancer and AIDS. The primary goal of gene therapy is to deliver a specific gene to a pre-determined target cell, and to direct expression of such a gene in a manner which will result in a therapeutic effect. Retroviral vectors have the ability to integrate in the host cell DNA irreversibly and therefore, are suitable vectors for permanent genetic modification of cells. Retrovirus-mediated gene transfer has been limited, however, by the inability of onco-retroviruses to productively infect non-dividing cells. Lentiviruses are unique among retroviruses because of their ability to infect target cells independently of their proliferation status. This chapter presents an up-to-date description of available lentiviral vectors, including vector design, applications to disease treatment and safety considerations. In addition, general aspects of the biology of lentiviruses with relevance to vector development will be discussed.


Genetic Therapy/methods , Genetic Vectors , Lentivirus/physiology , Genes, Viral , HIV Infections/therapy , HIV-1/genetics , Lentivirus/genetics , Lentivirus/pathogenicity , Lentivirus Infections/genetics , Viral Structural Proteins/genetics , Virus Integration
19.
J Virol ; 73(4): 2832-40, 1999 Apr.
Article En | MEDLINE | ID: mdl-10074131

Lentivirus vectors based on human immunodeficiency virus (HIV) type 1 (HIV-1) constitute a recent development in the field of gene therapy. A key property of HIV-1-derived vectors is their ability to infect nondividing cells. Although high-titer HIV-1-derived vectors have been produced, concerns regarding safety still exist. Safety concerns arise mainly from the possibility of recombination between transfer and packaging vectors, which may give rise to replication-competent viruses with pathogenic potential. We describe a novel lentivirus vector which is based on HIV, simian immunodeficiency virus (SIV), and vesicular stomatitis virus (VSV) and which we refer to as HIV/SIVpack/G. In this system, an HIV-1-derived genome is encapsidated by SIVmac core particles. These core particles are pseudotyped with VSV glycoprotein G. Because the nucleotide homology between HIV-1 and SIVmac is low, the likelihood of recombination between vector elements should be reduced. In addition, the packaging construct (SIVpack) for this lentivirus system was derived from SIVmac1A11, a nonvirulent SIV strain. Thus, the potential for pathogenicity with this vector system is minimal. The transduction ability of HIV/SIVpack/G was demonstrated with immortalized human lymphocytes, human primary macrophages, human bone marrow-derived CD34(+) cells, and primary mouse neurons. To our knowledge, these experiments constitute the first demonstration that the HIV-1-derived genome can be packaged by an SIVmac capsid. We demonstrate that the lentivirus vector described here recapitulates the biological properties of HIV-1-derived vectors, although with increased potential for safety in humans.


Genetic Therapy , Genetic Vectors , HIV-1/genetics , Lentivirus/genetics , Simian Immunodeficiency Virus/genetics , Animals , Humans , Mice
20.
Blood ; 93(2): 643-54, 1999 Jan 15.
Article En | MEDLINE | ID: mdl-9885227

Herpes simplex virus amplicon vectors expressing RANTES (HSVrantes) and the T-cell costimulatory ligand B7.1 (HSVB7.1) were studied for their ability to elicit a tumor-specific T-cell response in a murine lymphoma model. HSVB7.1- and HSVrantes-transduced EL4 cells expressed high levels of B7.1 and RANTES as analyzed by flow cytometry and enzyme-linked immunosorbent assay, respectively. Inoculation of ex vivo HSVB7.1 transduced cells in syngeneic mice resulted in regression of both transduced cells and nontransduced cells inoculated contralaterally. Direct intratumoral injection of HSVB7.1 and/or HSVrantes alone or in combination into established EL4 tumors led to complete tumor regression in injected tumors as well as in nontransduced contralaterally implanted tumor, whereas control tumors or tumors injected with HSVlac expressing beta-galactosidase did not regress. Maximal protection was achieved with combined injection of HSVB7.1 and HSVrantes; mice showing tumor regression were resistant to rechallenge with parental EL4 cells, and tumor cell-specific cytolytic T-cell activity was observed in mice demonstrating regression. HSV amplicon-mediated delivery of immune effector molecules may represent a useful strategy for immunotherapy in the setting of pre-existing tumor.


Genetic Vectors , Immunotherapy , Lymphoma, T-Cell/therapy , Simplexvirus/genetics , T-Lymphocytes/immunology , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression , Humans , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , T-Lymphocytes, Cytotoxic/immunology , Transfection , Tumor Cells, Cultured
...