Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
JCI Insight ; 4(11)2019 06 06.
Article in English | MEDLINE | ID: mdl-31167974

ABSTRACT

Reduction/elimination of HIV-1 reservoirs that persist despite combination antiretroviral therapy (cART) will likely require induction of viral expression by residual infected cells and enhanced clearance of these cells. TLR7 agonists have potential to mediate these activities. We evaluated immunologic and virologic effects of repeated doses of the TLR7 agonist GS-9620 in SIV-infected rhesus macaques receiving cART, which was initiated at 13 days after infection and was continued for 75 weeks prior to GS-9620 administration. During cART, GS-9620 induced transient upregulation of IFN-stimulated genes in blood and tissues, increases in plasma cytokines, and changes in immune cell population activation and phenotypes but did not result in measurable increases in plasma viremia or viral RNA-to-viral DNA ratio in PBMCs or tissues nor decreases in viral DNA in PBMC or tissues. SIV-specific CD8+ T cell responses, negligible prior to GS-9620 treatment, were not measurably boosted by treatment; a second course of GS-9620 administration overlapping with later cART discontinuation was associated with increased CD8+ T cell responses during viral recrudescence. These results confirm and extend evidence for GS-9620-mediated enhancement of antiviral immune responses in SIV-infected macaques but suggest that GS-9620-mediated viral induction may depend critically on the timing of initiation and duration of cART and resulting characteristics of viral reservoirs.


Subject(s)
Anti-Retroviral Agents , Pteridines , Simian Acquired Immunodeficiency Syndrome , Toll-Like Receptor 7/agonists , Viremia , Animals , Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Drug Therapy, Combination , Macaca mulatta , Male , Pteridines/administration & dosage , Pteridines/pharmacology , Pteridines/therapeutic use , RNA, Viral/genetics , RNA, Viral/metabolism , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Up-Regulation/drug effects , Viral Load/drug effects , Viremia/drug therapy , Viremia/immunology , Viremia/virology
2.
Mol Ther Nucleic Acids ; 8: 243-249, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28918025

ABSTRACT

Synthetic oligodeoxynucleotides containing unmethylated CpG motifs (CpG ODNs) stimulate immune cells via Toll-like receptor 9 (TLR9). Because oligodeoxynucleotides (ODNs) are susceptible to gastric degradation, clinical trials designed to evaluate their therapeutic utility have relied solely on parenteral routes of administration. A strategy to improve the activity of orally delivered ODNs by reducing their susceptibility to gastrointestinal (GI) digestion via encapsulation in calcium carbonate nanoparticles (ODNcaps) was recently described. This study compares the in vitro and in vivo activity of encapsulated (ODNcaps) versus free CpG ODNs delivered orally or parenterally. ODNcaps mirrored the ability of free ODNs to stimulate splenic B cells and macrophages in vitro. ODNcaps activated immune cells in the Peyer's patches and mesenteric lymph nodes after oral delivery. Their effect on GI immunity was evaluated in studies of dextran sulfate sodium (DSS)-induced colitis and enteric infection, whereas systemic immunity was examined by monitoring their effect on lipopolysaccharide (LPS)-induced cytokine production and systemic pathogen challenge. Results indicate that orally delivered CpG ODNs predominantly induce GI rather than systemic immunity, and that calcium carbonate encapsulation does not significantly alter this behavior.

3.
J Pathol Clin Res ; 2(4): 210-222, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27785366

ABSTRACT

The limited clinical success of anti-HGF/MET drugs can be attributed to the lack of predictive biomarkers that adequately select patients for treatment. We demonstrate here that quantitative digital imaging of formalin fixed paraffin embedded tissues stained by immunohistochemistry can be used to measure signals from weakly staining antibodies and provides new opportunities to develop assays for detection of MET receptor activity. To establish a biomarker panel of MET activation, we employed seven antibodies measuring protein expression in the HGF/MET pathway in 20 cases and up to 80 cores from 18 human cancer types. The antibodies bind to epitopes in the extra (EC)- and intracellular (IC) domains of MET (MET4EC, SP44_METIC, D1C2_METIC), to MET-pY1234/pY1235, a marker of MET kinase activation, as well as to HGF, pSFK or pMAPK. Expression of HGF was determined in tumour cells (T_HGF) as well as in stroma surrounding cancer (St_HGF). Remarkably, MET4EC correlated more strongly with pMET (r = 0.47) than SP44_METIC (r = 0.21) or D1C2_METIC (r = 0.08) across 18 cancer types. In addition, correlation coefficients of pMET and T_HGF (r = 0.38) and pMET and pSFK (r = 0.56) were high. Prediction models of MET activation reveal cancer-type specific differences in performance of MET4EC, SP44_METIC and anti-HGF antibodies. Thus, we conclude that assays to predict the response to HGF/MET inhibitors require a cancer-type specific antibody selection and should be developed in those cancer types in which they are employed clinically.

4.
J Virol ; 90(21): 9942-9952, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27558423

ABSTRACT

AIDS virus infections are rarely controlled by cell-mediated immunity, in part due to viral immune evasion and immunodeficiency resulting from CD4+ T-cell infection. One likely aspect of this failure is that antiviral cellular immune responses are either absent or present at low levels during the initial establishment of infection. To test whether an extensive, timely, and effective response could reduce the establishment of infection from a high-dose inoculum, we adoptively transferred large numbers of T cells that were molecularly engineered with anti-simian immunodeficiency virus (anti-SIV) activity into rhesus macaques 3 days following an intrarectal SIV inoculation. To measure in vivo antiviral activity, we assessed the number of viruses transmitted using SIVmac239X, a molecularly tagged viral stock containing 10 genotypic variants, at a dose calculated to transmit 12 founder viruses. Single-genome sequencing of plasma virus revealed that the two animals receiving T cells expressing SIV-specific T-cell receptors (TCRs) had significantly fewer viral genotypes than the two control animals receiving non-SIV-specific T cells (means of 4.0 versus 7.5 transmitted viral genotypes; P = 0.044). Accounting for the likelihood of transmission of multiple viruses of a particular genotype, the calculated means of the total number of founder viruses transmitted were 4.5 and 14.5 in the experimental and control groups, respectively (P = 0.021). Thus, a large antiviral T-cell response timed with virus exposure can limit viral transmission. The presence of strong, preexisting T-cell responses, including those induced by vaccines, might help prevent the establishment of infection at the lower-exposure doses in humans that typically transmit only a single virus. IMPORTANCE: The establishment of AIDS virus infection in an individual is essentially a race between the spreading virus and host immune defenses. Cell-mediated immune responses induced by infection or vaccination are important contributors in limiting viral replication. However, in human immunodeficiency virus (HIV)/SIV infection, the virus usually wins the race, irreversibly crippling the immune system before an effective cellular immune response is developed and active. We found that providing an accelerated response by adoptively transferring large numbers of antiviral T cells shortly after a high-dose mucosal inoculation, while not preventing infection altogether, limited the number of individual viruses transmitted. Thus, the presence of strong, preexisting T-cell responses, including those induced by vaccines, might prevent infection in humans, where the virus exposure is considerably lower.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Macaca mulatta/immunology , Macaca mulatta/virology , Simian Immunodeficiency Virus/immunology , Adoptive Transfer/methods , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Humans , Immunity, Cellular/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Vaccination/methods , Virus Replication/genetics
5.
Nucleic Acids Res ; 44(16): 7848-65, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27439715

ABSTRACT

Although APOBEC3 cytidine deaminases A3G, A3F, A3D and A3H are packaged into virions and inhibit viral replication by inducing G-to-A hypermutation, it is not known whether they are copackaged and whether they can act additively or synergistically to inhibit HIV-1 replication. Here, we showed that APOBEC3 proteins can be copackaged by visualization of fluorescently-tagged APOBEC3 proteins using single-virion fluorescence microscopy. We further determined that viruses produced in the presence of A3G + A3F and A3G + A3H, exhibited extensive comutation of viral cDNA, as determined by the frequency of G-to-A mutations in the proviral genomes in the contexts of A3G (GG-to-AG) and A3D, A3F or A3H (GA-to-AA) edited sites. The copackaging of A3G + A3F and A3G + A3H resulted in an additive increase and a modest synergistic increase (1.8-fold) in the frequency of GA-to-AA mutations, respectively. We also identified distinct editing site trinucleotide sequence contexts for each APOBEC3 protein and used them to show that hypermutation of proviral DNAs from seven patients was induced by A3G, A3F (or A3H), A3D and A3G + A3F (or A3H). These results indicate that APOBEC3 proteins can be copackaged and can comutate the same genomes, and can cooperate to inhibit HIV replication.


Subject(s)
Cytosine Deaminase/metabolism , Genome, Viral , HIV-1/genetics , Mutation/genetics , APOBEC Deaminases , Adult , Cell Line , Cytidine Deaminase , HIV Infections/metabolism , HIV Infections/virology , Humans , Male , Mutation Rate , Nucleotides/genetics , Proviruses/physiology , Sequence Analysis, DNA , Virion/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism
6.
JCI Insight ; 1(10)2016 Jul 07.
Article in English | MEDLINE | ID: mdl-27446990

ABSTRACT

Early after HIV infection there is substantial depletion of CD4+ T cells in the gastrointestinal (GI) tract lamina propria (LP), with associated epithelial barrier damage, leading to microbial translocation and systemic inflammation and immune activation. In this study, we analyzed these early events in the GI tract in a cohort of Thai acute HIV-infected patients and determined the effect of early combination antiretroviral treatment (cART). HIV-uninfected and chronically and acutely HIV-infected patients at different Fiebig stages (I-V) underwent colonic biopsies and then received cART. Immunohistochemistry and quantitative image analysis were performed on cross-sectional and longitudinal colon biopsy specimens (day 0 to week 96) to measure GI tract damage (infiltration of polymorphonuclear cells), inflammation (M×1, TNF-α), immune activation (Ki-67), and the CD4+ T cell population in the LP. The magnitude of GI tract damage, immune activation, and inflammation was significantly increased, with significantly depleted CD4+ T cells in the LP in all acutely infected groups prior to cART compared with HIV-uninfected control participants. While most patients treated during acute infection resolved GI tract inflammation and immune activation back to baseline levels after 24 weeks of cART, most acutely infected participants did not restore their CD4+ T cells after 96 weeks of cART.

7.
Amino Acids ; 48(3): 859-872, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26660760

ABSTRACT

Proline dehydrogenase/oxidase (PRODH/POX) is a mitochondrial protein critical to multiple stress pathways. Because of the roles of PRODH/POX in signaling, and its shared localization to the mitochondrial inner membrane with the electron transport chain (ETC), we investigated whether there was a direct relationship between PRODH/POX and regulation of the ETC. We found that PRODH/POX binds directly to CoQ1 and that CoQ1-dependent PRODH/POX activity required functional Complex III and Complex IV. PRODH/POX supported respiration in living cells during nutrient stress; however, expression of PRODH/POX resulted in an overall decrease in respiratory fitness. Effects on respiratory fitness were inhibited by DHP and NAC, indicating that these effects were mediated by PRODH/POX-dependent reactive oxygen species (ROS) generation. PRODH/POX expression resulted in a dose-dependent down-regulation of Complexes I-IV of the ETC, and this effect was also mitigated by the addition of DHP and NAC. We found that succinate was an uncompetitive inhibitor of PRODH/POX activity, inhibited ROS generation by PRODH/POX, and alleviated PRODH/POX effects on respiratory fitness. The findings demonstrate novel cross-talk between proline and succinate respiration in vivo and provide mechanistic insights into observations from previous animal studies. Our results suggest a potential regulatory loop between PRODH/POX and succinate in regulation of mitochondrial respiration.


Subject(s)
Mitochondria/metabolism , Proline Oxidase/metabolism , Succinic Acid/metabolism , Animals , Cell Respiration , Electron Transport , Liver/enzymology , Liver/metabolism , Mice , Mitochondria/enzymology , Proline Oxidase/genetics , Reactive Oxygen Species/metabolism
8.
Antimicrob Agents Chemother ; 60(3): 1560-72, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26711758

ABSTRACT

Replication-competent human immunodeficiency virus (HIV) persists in infected people despite suppressive combination antiretroviral therapy (cART), and it represents a major obstacle to HIV functional cure or eradication. We have developed a model of cART-mediated viral suppression in simian human immunodeficiency virus (SIV) mac239-infected Indian rhesus macaques and evaluated the impact of the histone deacetylase inhibitor (HDACi) romidepsin (RMD) on viremia in vivo. Eight macaques virologically suppressed to clinically relevant levels (<30 viral RNA copies/ml of plasma), using a three-class five-drug cART regimen, received multiple intravenous infusions of either RMD (n = 5) or saline (n = 3) starting 31 to 54 weeks after cART initiation. In vivo RMD treatment resulted in significant transient increases in acetylated histone levels in CD4(+) T cells. RMD-treated animals demonstrated plasma viral load measurements for each 2-week treatment cycle that were significantly higher than those in saline control-treated animals during periods of treatment, suggestive of RMD-induced viral reactivation. However, plasma virus rebound was indistinguishable between RMD-treated and control-treated animals for a subset of animals released from cART. These findings suggest that HDACi drugs, such as RMD, can reactivate residual virus in the presence of suppressive antiviral therapy and may be a valuable component of a comprehensive HIV functional cure/eradication strategy.


Subject(s)
Depsipeptides/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Macaca mulatta/virology , RNA, Viral/blood , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Viral Load/drug effects , Acetylation , Animals , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/metabolism , Depsipeptides/pharmacokinetics , Histone Deacetylase Inhibitors/pharmacokinetics , Histones/metabolism , Viremia/drug therapy , Virus Activation/drug effects
9.
Nat Commun ; 6: 8020, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26282376

ABSTRACT

Mucosal damage to the gastrointestinal (GI) tract with resulting microbial translocation is hypothesized to significantly contribute to the heightened and persistent chronic inflammation and immune activation characteristic to HIV infection. Here we employ a non-human primate model of chemically induced colitis in SIV-uninfected rhesus macaques that we developed using dextran sulfate sodium (DSS), to directly test this hypothesis. DSS treatment results in GI barrier damage with associated microbial translocation, inflammation and immune activation. The progression and severity of colitis are longitudinally monitored by a magnetic resonance imaging approach. DSS treatment of SIV-infected African green monkeys, a natural host species for SIV that does not manifest GI tract damage or chronic immune activation during infection, results in colitis with elevated levels of plasma SIV RNA, sCD14, LPS, CRP and mucosal CD4+ T-cell loss. Together these results support the hypothesis that GI tract damage leading to local and systemic microbial translocation, and associated immune activation, are important determinants of AIDS pathogenesis.


Subject(s)
Colitis/chemically induced , Colitis/pathology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Immunodeficiency Virus , Animals , Dextran Sulfate/toxicity , Female , Gastrointestinal Tract/pathology , Macaca mulatta , Male
10.
J Hepatol ; 63(5): 1181-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26143441

ABSTRACT

BACKGROUND & AIMS: Liver inflammatory diseases associated with cancer promoting somatic oncogene mutations are increasing in frequency. Preclinical cancer models that allow for the study of early tumor progression are often protracted, which limits the experimental study parameters due to time and expense. Here we report a robust inexpensive approach using Sleeping Beauty transposition (SBT) delivery of oncogenes along with Gaussia Luciferase expression vector GLuc, to assess de novo liver tumor progression, as well as the detection of innate immune responses or responses induced by therapeutic intervention. METHODS: Tracking de novo liver tumor progression with GLuc was demonstrated in models of hepatocellular carcinoma (HCC) or adenoma (HCA) initiated by hydrodynamic delivery of SBT oncogenes. RESULTS: Rising serum luciferase levels correlated directly with increasing liver tumor burden and eventual morbidity. Early detection of hepatocyte apoptosis from mice with MET+CAT transfected hepatocytes was associated with a transient delay in HCC growth mediated by a CD8(+) T-cell response against transformed hepatocytes. Furthermore, mice that lack B cells or macrophages had an increase in TUNEL(+) hepatocytes following liver MET transfection demonstrating that these cells provide protection from MET-induced hepatocyte apoptosis. Treatment with IL-18+IL-12 of mice displaying established HCC decreased tumor burden which was associated with decreased levels of serum luciferase. CONCLUSIONS: Hydrodynamic delivery of the SBT vector GLuc to hepatocytes serves as a simple blood-based approach for real-time tracking of pathologically distinct types of liver cancer. This revealed tumor-induced immunologic responses and was beneficial in monitoring the efficacy of therapeutic interventions.


Subject(s)
Adenoma, Liver Cell/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/immunology , Immunity, Cellular , Liver Neoplasms, Experimental/immunology , Luciferases/blood , Recombinant Proteins/therapeutic use , Adenoma, Liver Cell/drug therapy , Adenoma, Liver Cell/pathology , Animals , Apoptosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Disease Progression , Hepatocytes/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Interleukin-12/therapeutic use , Interleukin-18/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/pathology , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Signal Transduction
11.
Virology ; 484: 334-340, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26164392

ABSTRACT

Recombination can assort polymorphic alleles to increase diversity in the HIV-1 population. To better understand the recombination potential of subtype A HIV-1, we generated viruses containing sequences from two variants circulating in Russia and analyzed the polymerase gene (pol) of the recombinants after one round of HIV-1 replication using single-genome sequencing. We observed that recombination occurred throughout pol and could easily assort alleles containing mutations that conferred resistance to currently approved antivirals. We measured the recombination rate in various regions of pol including a G-rich region that has been previously proposed to be a recombination hot spot. Our study does not support a recombination hot spot in this G-rich region. Importantly, of the 58 proviral sequences containing crossover event(s) in pol, we found that each sequence was a unique genotype indicating that recombination is a powerful genetic mechanism in assorting the genomes of subtype A HIV-1 variants.


Subject(s)
HIV-1/genetics , Recombination, Genetic , Genetic Variation , Genome, Viral , Genotype , HIV Infections/virology , HIV-1/classification , HIV-1/isolation & purification , Humans , Russia , Sequence Analysis, DNA , Virus Cultivation , pol Gene Products, Human Immunodeficiency Virus/genetics
12.
Antimicrob Agents Chemother ; 58(11): 6790-806, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25182644

ABSTRACT

Nonhuman primate models are needed for evaluations of proposed strategies targeting residual virus that persists in HIV-1-infected individuals receiving suppressive combination antiretroviral therapy (cART). However, relevant nonhuman primate (NHP) models of cART-mediated suppression have proven challenging to develop. We used a novel three-class, six-drug cART regimen to achieve durable 4.0- to 5.5-log reductions in plasma viremia levels and declines in cell-associated viral RNA and DNA in blood and tissues of simian immunodeficiency virus SIVmac239-infected Indian-origin rhesus macaques, then evaluated the impact of treatment with the histone deacetylase inhibitor (HDACi) suberoylanilide hydroxamic acid (SAHA; Vorinostat) on the residual virus pool. Ex vivo SAHA treatment of CD4(+) T cells obtained from cART-suppressed animals increased histone acetylation and viral RNA levels in culture supernatants. cART-suppressed animals each received 84 total doses of oral SAHA. We observed SAHA dose-dependent increases in acetylated histones with evidence for sustained modulation as well as refractoriness following prolonged administration. In vivo virologic activity was demonstrated based on the ratio of viral RNA to viral DNA in peripheral blood mononuclear cells, a presumptive measure of viral transcription, which significantly increased in SAHA-treated animals. However, residual virus was readily detected at the end of treatment, suggesting that SAHA alone may be insufficient for viral eradication in the setting of suppressive cART. The effects observed were similar to emerging data for repeat-dose SAHA treatment of HIV-infected individuals on cART, demonstrating the feasibility, utility, and relevance of NHP models of cART-mediated suppression for in vivo assessments of AIDS virus functional cure/eradication approaches.


Subject(s)
Anti-Retroviral Agents/therapeutic use , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Acetylation/drug effects , Animals , CD4-Positive T-Lymphocytes/drug effects , Disease Models, Animal , Drug Therapy, Combination , Histones/metabolism , Macaca mulatta , RNA, Viral/blood , RNA, Viral/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Viral Load/drug effects , Vorinostat
13.
J Virol ; 88(14): 8077-90, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24807714

ABSTRACT

Following mucosal human immunodeficiency virus type 1 transmission, systemic infection is established by one or only a few viral variants. Modeling single-variant, mucosal transmission in nonhuman primates using limiting-dose inoculations with a diverse simian immunodeficiency virus isolate stock may increase variability between animals since individual variants within the stock may have substantial functional differences. To decrease variability between animals while retaining the ability to enumerate transmitted/founder variants by sequence analysis, we modified the SIVmac239 clone to generate 10 unique clones that differ by two or three synonymous mutations (molecular tags). Transfection- and infection-derived virus stocks containing all 10 variants showed limited phenotypic differences in 9 of the 10 clones. Twenty-nine rhesus macaques were challenged intrarectally or intravenously with either a single dose or repeated, limiting doses of either stock. The proportion of each variant within each inoculum and in plasma from infected animals was determined by using a novel real-time single-genome amplification assay. Each animal was infected with one to five variants, the number correlating with the dose. Longitudinal sequence analysis revealed that the molecular tags are highly stable with no reversion to the parental sequence detected in >2 years of follow-up. Overall, the viral stocks are functional and mucosally transmissible and the number of variants is conveniently discernible by sequence analysis of a small amplicon. This approach should be useful for tracking individual infection events in preclinical vaccine evaluations, long-term viral reservoir establishment/clearance research, and transmission/early-event studies. Importance: Human immunodeficiency virus type 1 transmission is established by one or only a few viral variants. Modeling of limited variant transmission in nonhuman primates with a diverse simian immunodeficiency virus isolate stock may increase the variability between animals because of functional differences in the individual variants within the stock. To decrease such variability while retaining the ability to distinguish and enumerate transmitted/founder variants by sequence analysis, we generated a viral stock with 10 sequence-identifiable but otherwise genetically identical variants. This virus was characterized in vitro and in vivo and shown to allow discrimination of distinct transmission events. This approach provides a novel nonhuman primate challenge system for the study of viral transmission, evaluation of vaccines and other prevention approaches, and characterization of viral reservoirs and strategies to target them.


Subject(s)
Genetic Variation , Plasma/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/classification , Simian Immunodeficiency Virus/isolation & purification , Animals , Disease Models, Animal , Genotype , Longitudinal Studies , Macaca mulatta , RNA, Viral/genetics , Sequence Analysis, DNA , Simian Immunodeficiency Virus/genetics
14.
J Virol ; 88(13): 7589-601, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24760888

ABSTRACT

UNLABELLED: The genetic variation in HIV-1 in patients is due to the high rate of viral replication, the high viral load, and the errors made during viral replication. Some of the mutations in reverse transcriptase (RT) that alter the deoxynucleoside triphosphate (dNTP)-binding pocket, including those that confer resistance to nucleoside/nucleotide analogs, affect dNTP selection during replication. The effects of mutations in RT on the spectrum (nature, position, and frequency) of errors made in vivo are poorly understood. We previously determined the mutation rate and the frequency of different types of mutations and identified hot spots for mutations in a lacZα (the α complementing region of lacZ) reporter gene carried by an HIV-1 vector that replicates using wild-type RT. We show here that four mutations (Y115F, M184V, M184I, and Q151M) in the dNTP-binding pocket of RT that had relatively small effects on the overall HIV-1 mutation rate (less than 3-fold compared to the wild type) significantly increased mutations at some specific positions in the lacZα reporter gene. We also show that changes in a sequence that flanks the reporter gene can affect the mutations that arise in the reporter. These data show that changes either in HIV-1 RT or in the sequence of the nucleic acid template can affect the spectrum of mutations made during viral replication. This could, by implication, affect the generation of drug-resistant mutants and immunological-escape mutants in patients. IMPORTANCE: RT is the viral enzyme that converts the RNA genome of HIV into DNA. Errors made during replication allow the virus to escape from the host's immune system and to develop resistance to the available anti-HIV drugs. We show that four different mutations in RT which are known to be associated with resistance to anti-RT drugs modestly increased the overall frequency of errors made during viral replication. However, the increased errors were not uniformly distributed; the additional errors occurred at a small number of positions (hot spots). Moreover, some of the RT mutations preferentially affected the nature of the errors that were made (some RT mutations caused an increase in insertion and deletion errors; others caused an increase in substitution errors). We also show that sequence changes in a region adjacent to a target gene can affect the errors made within the target gene.


Subject(s)
Drug Resistance, Viral/genetics , HIV Infections/virology , HIV Reverse Transcriptase/genetics , HIV-1/physiology , Lac Operon/genetics , Mutation/genetics , Virus Replication/genetics , Cells, Cultured , DNA, Viral/genetics , HIV Infections/drug therapy , HIV Infections/genetics , HIV Reverse Transcriptase/antagonists & inhibitors , Humans , Reverse Transcriptase Inhibitors/pharmacology
15.
Carcinogenesis ; 35(5): 1078-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24403310

ABSTRACT

Silicosis is an inflammatory lung disease induced by the inhalation of silica-containing dust particles. There is conflicting data on whether patients with silicosis are more susceptible to lung cancer induced by cigarette smoke. To examine this issue experimentally, a model was developed in which one of the most abundant and potent carcinogens present in cigarette smoke [4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone (NNK)] was administered to mice at the peak of silica-induced pulmonary inflammation. Results show that the incidence of lung tumors in silicotic mice treated with NNK was significantly increased compared with mice exposed to silica or NNK alone. Synthetic oligonucleotides (ODN) containing repetitive TTAGGG motifs can block pathologic inflammation. We therefore examined whether treatment with these suppressive (Sup) ODN could block silica-induced pulmonary inflammation and thereby reduce susceptibility to lung cancer. Results show that Sup (but not control) ODN inhibit pulmonary fibrosis and other inflammatory manifestations of chronic silicosis. Of greater import, Sup ODN reduced lung tumor incidence and multiplicity in silicotic mice exposed to NNK. These findings establish an experimental model for examining the role of silicotic inflammation in cancer susceptibility and demonstrate that Sup ODN represent a novel therapy for chronic silicosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Disease Susceptibility , Lung Neoplasms/etiology , Lung Neoplasms/prevention & control , Oligodeoxyribonucleotides/pharmacology , Silicosis/complications , Animals , Anti-Inflammatory Agents/administration & dosage , Biomarkers , Disease Models, Animal , Female , Gene Expression , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lung Neoplasms/pathology , Mice , Nitrosamines/adverse effects , Oligodeoxyribonucleotides/administration & dosage , Pneumonia/complications , Pneumonia/drug therapy , Pneumonia/etiology , Pneumonia/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Silicon Dioxide/adverse effects , Silicosis/pathology
16.
Mol Cell Biol ; 33(16): 3242-58, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23775115

ABSTRACT

C/EBPß is an important regulator of oncogene-induced senescence (OIS). Here, we show that C/EBPγ, a heterodimeric partner of C/EBPß whose biological functions are not well understood, inhibits cellular senescence. Cebpg(-/-) mouse embryonic fibroblasts (MEFs) proliferated poorly, entered senescence prematurely, and expressed a proinflammatory gene signature, including elevated levels of senescence-associated secretory phenotype (SASP) genes whose induction by oncogenic stress requires C/EBPß. The senescence-suppressing activity of C/EBPγ required its ability to heterodimerize with C/EBPß. Covalently linked C/EBPß homodimers (ß∼ß) inhibited the proliferation and tumorigenicity of Ras(V12)-transformed NIH 3T3 cells, activated SASP gene expression, and recruited the CBP coactivator in a Ras-dependent manner, whereas Î³âˆ¼ß heterodimers lacked these capabilities and efficiently rescued proliferation of Cebpg(-/-) MEFs. C/EBPß depletion partially restored growth of C/EBPγ-deficient cells, indicating that the increased levels of C/EBPß homodimers in Cebpg(-/-) MEFs inhibit proliferation. The proliferative functions of C/EBPγ are not restricted to fibroblasts, as hematopoietic progenitors from Cebpg(-/-) bone marrow also displayed impaired growth. Furthermore, high CEBPG expression correlated with poorer clinical prognoses in several human cancers, and C/EBPγ depletion decreased proliferation and induced senescence in lung tumor cells. Our findings demonstrate that C/EBPγ neutralizes the cytostatic activity of C/EBPß through heterodimerization, which prevents senescence and suppresses basal transcription of SASP genes.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cellular Senescence , Gene Expression Regulation , Neoplasms/genetics , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cell Cycle , Cell Line , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Deletion , Gene Expression Regulation, Neoplastic , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neoplasms/metabolism , Protein Multimerization
17.
J Infect Dis ; 207(6): 880-92, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23087435

ABSTRACT

BACKGROUND: Human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infections induce robust, generalized inflammatory responses that begin during acute infection and lead to pathological systemic immune activation, fibrotic damage of lymphoid tissues, and CD4⁺ T-cell loss, pathogenic processes that contribute to disease progression. METHODS: To better understand the contribution of tumor necrosis factor (TNF), a key regulator of acute inflammation, to lentiviral pathogenesis, rhesus macaques newly infected with SIVmac239 were treated for 12 weeks in a pilot study with adalimumab (Humira), a human anti-TNF monoclonal antibody. RESULTS: Adalimumab did not affect plasma SIV RNA levels or measures of T-cell immune activation (CD38 or Ki67) in peripheral blood or lymph node T cells. However, compared with untreated rhesus macaques, adalimumab-treated rhesus macaques showed attenuated expression of proinflammatory genes, decreased infiltration of polymorphonuclear cells into the T-cell zone of lymphoid tissues, and weaker antiinflammatory regulatory responses to SIV infection (ie, fewer presumed alternatively activated [ie, CD163⁺] macrophages, interleukin 10-producing cells, and transforming growth factor ß-producing cells), along with reduced lymphoid tissue fibrosis and better preservation of CD4⁺ T cells. CONCLUSIONS: While HIV/SIV replication drives pathogenesis, these data emphasize the contribution of the inflammatory response to lentiviral infection to overall pathogenesis, and they suggest that early modulation of the inflammatory response may help attenuate disease progression.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Inflammation/metabolism , Lymph Nodes/pathology , Lymphocyte Activation/drug effects , Retroviruses, Simian , Simian Acquired Immunodeficiency Syndrome/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Adalimumab , Animals , CD4 Lymphocyte Count , Cell Movement/drug effects , Cytokines/genetics , Cytokines/metabolism , Fibrosis , Gene Expression Regulation/drug effects , Inflammation/genetics , Lymph Nodes/immunology , Macaca mulatta , Macrophages/drug effects , Macrophages/physiology , Male , RNA, Viral/metabolism , Random Allocation , Simian Acquired Immunodeficiency Syndrome/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Viral Load/drug effects
18.
Environ Mol Mutagen ; 53(5): 392-407, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22674528

ABSTRACT

Both gene methylation changes and genetic instability have been noted in offspring of male rodents exposed to radiation or chemicals, but few specific gene targets have been established. Previously, we identified the gene for ribosomal RNA, rDNA, as showing methylation change in sperm of mice treated with the preconceptional carcinogen, chromium(III) chloride. rDNA is a critical cell growth regulator. Here, we investigated the effects of paternal treatments on rDNA in offspring tissue. A total of 93 litters and 758 offspring were obtained, permitting rigorous mixed-effects models statistical analysis of the results. We show that the offspring of male mice treated with Cr(III) presented increased methylation in a promoter sequence of the rDNA gene, specifically in lung. Furthermore polymorphic variants of the multi-copy rDNA genes displayed altered frequencies indicative of structural changes, as a function of both tissue type and paternal treatments. Organismal effects also occurred: some groups of offspring of male mice treated with either Cr(III) or its vehicle, acidic saline, compared with those of untreated mice, had altered average body and liver weights and levels of serum glucose and leptin. Males treated directly with Cr(III) or acidic saline presented serum hormone changes consistent with a stress response. These results establish for the first time epigenetic and genetic instability effects in a gene of central physiological importance, in offspring of male mice exposed preconceptionally to chemicals, possibly related to a stress response in these males.


Subject(s)
Stress, Physiological/drug effects , Animals , DNA Methylation , DNA, Ribosomal/genetics , Genotype , Insulin-Like Growth Factor I/genetics , Male , Mice , Regulatory Sequences, Nucleic Acid
19.
EMBO J ; 30(18): 3714-28, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21804532

ABSTRACT

C/EBPß is an auto-repressed protein that becomes post-translationally activated by Ras-MEK-ERK signalling. C/EBPß is required for oncogene-induced senescence (OIS) of primary fibroblasts, but also displays pro-oncogenic functions in many tumour cells. Here, we show that C/EBPß activation by H-Ras(V12) is suppressed in immortalized/transformed cells, but not in primary cells, by its 3' untranslated region (3'UTR). 3'UTR sequences inhibited Ras-induced cytostatic activity of C/EBPß, DNA binding, transactivation, phosphorylation, and homodimerization, without significantly affecting protein expression. The 3'UTR suppressed induction of senescence-associated C/EBPß target genes, while promoting expression of genes linked to cancers and TGFß signalling. An AU-rich element (ARE) and its cognate RNA-binding protein, HuR, were required for 3'UTR inhibition. These components also excluded the Cebpb mRNA from a perinuclear cytoplasmic region that contains activated ERK1/2, indicating that the site of C/EBPß translation controls de-repression by Ras signalling. Notably, 3'UTR inhibition and Cebpb mRNA compartmentalization were absent in primary fibroblasts, allowing Ras-induced C/EBPß activation and OIS to proceed. Our findings reveal a novel mechanism whereby non-coding mRNA sequences selectively regulate C/EBPß activity and suppress its anti-oncogenic functions.


Subject(s)
3' Untranslated Regions , Aging , CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation , Oncogene Protein p21(ras)/metabolism , Cell Line , Humans , Models, Biological , RNA, Messenger/metabolism
20.
PLoS One ; 6(7): e22266, 2011.
Article in English | MEDLINE | ID: mdl-21765958

ABSTRACT

Gene rearrangement occurs during development in some cell types and this genome dynamics is modulated by intrinsic and extrinsic factors, including growth stimulants and nutrients. This raises a possibility that such structural change in the genome and its subsequent epigenetic modifications may also take place during mammalian ontogeny, a process undergoing finely orchestrated cell division and differentiation. We tested this hypothesis by comparing single nucleotide polymorphism-defined haplotype frequencies and DNA methylation of the rDNA multicopy gene between two mouse ontogenic stages and among three adult tissues of individual mice. Possible influences to the genetic and epigenetic dynamics by paternal exposures were also examined for Cr(III) and acid saline extrinsic factors. Variables derived from litters, individuals, and duplicate assays in large mouse populations were examined using linear mixed-effects model. We report here that active rDNA rearrangement, represented by changes of haplotype frequencies, arises during ontogenic progression from day 8 embryos to 6-week adult mice as well as in different tissue lineages and is modifiable by paternal exposures. The rDNA methylation levels were also altered in concordance with this ontogenic progression and were associated with rDNA haplotypes. Sperm showed highest level of methylation, followed by lungs and livers, and preferentially selected haplotypes that are positively associated with methylation. Livers, maintaining lower levels of rDNA methylation compared with lungs, expressed more rRNA transcript. In vitro transcription demonstrated haplotype-dependent rRNA expression. Thus, the genome is also dynamic during mammalian ontogeny and its rearrangement may trigger epigenetic changes and subsequent transcriptional controls, that are further influenced by paternal exposures.


Subject(s)
DNA Methylation/genetics , DNA, Ribosomal/genetics , Gene Rearrangement/genetics , Paternal Exposure , Transcription, Genetic , Animals , Base Sequence , CpG Islands/genetics , Epigenesis, Genetic , Haplotypes/genetics , Male , Mice , Models, Genetic , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...