Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
2.
Antimicrob Agents Chemother ; 68(7): e0020124, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38829049

ABSTRACT

Limited cellular levels of the HIV transcriptional activator Tat are one contributor to proviral latency that might be targeted in HIV cure strategies. We recently demonstrated that lipid nanoparticles containing HIV tat mRNA induce HIV expression in primary CD4 T cells. Here, we sought to further characterize tat mRNA in the context of several benchmark latency reversal agents (LRAs), including inhibitor of apoptosis protein antagonists (IAPi), bromodomain and extra-Terminal motif inhibitors (BETi), and histone deacetylase inhibitors (HDACi). tat mRNA reversed latency across several different cell line models of HIV latency, an effect dependent on the TAR hairpin loop. Synergistic enhancement of tat mRNA activity was observed with IAPi, HDACi, and BETi, albeit to variable degrees. In primary CD4 T cells from durably suppressed people with HIV, tat mRNA profoundly increased the frequencies of elongated, multiply-spliced, and polyadenylated HIV transcripts, while having a lesser impact on TAR transcript frequencies. tat mRNAs alone resulted in variable HIV p24 protein induction across donors. However, tat mRNA in combination with IAPi, BETi, or HDACi markedly enhanced HIV RNA and protein expression without overt cytotoxicity or cellular activation. Notably, combination regimens approached or in some cases exceeded the latency reversal activity of maximal mitogenic T cell stimulation. Higher levels of tat mRNA-driven HIV p24 induction were observed in donors with larger mitogen-inducible HIV reservoirs, and expression increased with prolonged exposure time. Combination LRA strategies employing both small molecule inhibitors and Tat delivered to CD4 T cells are a promising approach to effectively target the HIV reservoir.


Subject(s)
CD4-Positive T-Lymphocytes , HIV Infections , HIV-1 , Histone Deacetylase Inhibitors , Nanoparticles , Virus Latency , tat Gene Products, Human Immunodeficiency Virus , Virus Latency/drug effects , Humans , tat Gene Products, Human Immunodeficiency Virus/genetics , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/metabolism , HIV-1/drug effects , HIV-1/genetics , Histone Deacetylase Inhibitors/pharmacology , HIV Infections/drug therapy , HIV Infections/virology , RNA, Messenger/genetics , RNA, Messenger/metabolism , HIV Antigens/genetics , Anti-HIV Agents/pharmacology
3.
Vaccines (Basel) ; 12(3)2024 Feb 25.
Article in English | MEDLINE | ID: mdl-38543871

ABSTRACT

Chronic infection with hepatitis B virus (HBV) develops in millions of patients per year, despite the availability of effective prophylactic vaccines. Patients who resolve acute HBV infection develop HBV-specific polyfunctional T cells accompanied by neutralizing antibodies, while in patients with chronic hepatitis B (CHB), immune cells are dysfunctional and impaired. We describe a lipid nanoparticle (LNP)-formulated mRNA vaccine, optimized for the expression of HBV core, polymerase, and surface (preS2-S) antigens with the aim of inducing an effective immune response in patients with CHB. Prime and prime/boost vaccination with LNP-formulated mRNA encoding for core, pol, and/or preS2-S dosing strategies were compared in naive C57BL/6 and BALB/c mice. Immune responses were assessed by IFN-γ ELISpot, intracellular cytokine staining (ICS), and ELISA for antibody production, whereas anti-viral efficacy was evaluated in the AAV-HBV mouse model. The mRNA vaccine induced strong antigen-specific polyfunctional T cell responses in these mouse models, accompanied by the emergence of anti-HBs and anti-HBe antibodies. After three immunizations, the antigen-specific immune stimulation resulted in up to 1.7 log10 IU/mL reduction in systemic HBV surface antigen (HBsAg), accompanied by a transient drop in systemic HBeAg, and this was observed in 50% of the AAV-HBV-transduced mice in the absence of additional modalities such as adjuvants, HBsAg reducing agents, or checkpoint inhibitors. However, no treatment-related effect on viremia was observed in the liver. These results warrant further optimization and evaluation of this mRNA vaccine as a candidate in a multimodal therapeutic regimen for the treatment of chronic HBV infection.

4.
Viruses ; 16(3)2024 02 23.
Article in English | MEDLINE | ID: mdl-38543713

ABSTRACT

BACKGROUND AND AIMS: Treatment with siRNAs that target HBV has demonstrated robust declines in HBV antigens. This effect is also observed in the AAV-HBV mouse model, which was used to investigate if two cycles of GalNAc-HBV-siRNA treatment could induce deeper declines in HBsAg levels or prevent rebound, and to provide insights into the liver immune microenvironment. METHODS: C57Bl/6 mice were transduced with one of two different titers of AAV-HBV for 28 days, resulting in stable levels of HBsAg of about 103 or 105 IU/mL. Mice were treated for 12 weeks (four doses q3wk) per cycle with 3 mg/kg of siRNA-targeting HBV or an irrelevant sequence either once (single treatment) or twice (retreatment) with an 8-week treatment pause in between. Blood was collected to evaluate viral parameters. Nine weeks after the last treatment, liver samples were collected to perform phenotyping, bulk RNA-sequencing, and immunohistochemistry. RESULTS: Independent of HBsAg baseline levels, treatment with HBV-siRNA induced a rapid decline in HBsAg levels, which then plateaued before gradually rebounding 12 weeks after treatment stopped. A second cycle of HBV-siRNA treatment induced a further decline in HBsAg levels in serum and the liver, reaching undetectable levels and preventing rebound when baseline levels were 103 IU/mL. This was accompanied with a significant increase in inflammatory macrophages in the liver and significant upregulation of regulatory T-cells and T-cells expressing immune checkpoint receptors. CONCLUSIONS: Retreatment induced an additional decline in HBsAg levels, reaching undetectable levels when baseline HBsAg levels were 3log10 or less. This correlated with T-cell activation and upregulation of Trem2.


Subject(s)
Hepatitis B Surface Antigens , Hepatitis B virus , Animals , Mice , Hepatitis B virus/genetics , RNA, Small Interfering/genetics , Liver , Retreatment , DNA, Viral , Antiviral Agents/therapeutic use , Membrane Glycoproteins , Receptors, Immunologic
5.
Nat Commun ; 14(1): 8397, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38110433

ABSTRACT

The development of latency reversing agents that potently reactivate HIV without inducing global T cell activation would benefit the field of HIV reservoir research and could pave the way to a functional cure. Here, we explore the reactivation capacity of a lipid nanoparticle containing Tat mRNA (Tat-LNP) in CD4 T cells from people living with HIV undergoing antiretroviral therapy (ART). When combined with panobinostat, Tat-LNP induces latency reversal in a significantly higher proportion of latently infected cells compared to PMA/ionomycin (≈ 4-fold higher). We demonstrate that Tat-LNP does not alter the transcriptome of CD4 T cells, enabling the characterization of latently infected cells in their near-native state. Upon latency reversal, we identify transcriptomic differences between infected cells carrying an inducible provirus and non-infected cells (e.g. LINC02964, GZMA, CCL5). We confirm the transcriptomic differences at the protein level and provide evidence that the long non-coding RNA LINC02964 plays a role in active HIV infection. Furthermore, p24+ cells exhibit heightened PI3K/Akt signaling, along with downregulation of protein translation, suggesting that HIV-infected cells display distinct signatures facilitating their long-term persistence. Tat-LNP represents a valuable research tool for in vitro reservoir studies as it greatly facilitates the in-depth characterization of HIV reservoir cells' transcriptome and proteome profiles.


Subject(s)
Gene Products, tat , HIV-1 , Nanoparticles , RNA, Viral , Virus Latency , Virus Latency/drug effects , Virus Latency/genetics , Gene Products, tat/genetics , Gene Products, tat/metabolism , RNA, Viral/administration & dosage , RNA, Viral/genetics , RNA, Viral/metabolism , Nanoparticles/administration & dosage , Nanoparticles/chemistry , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/virology , Panobinostat/pharmacology , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , CD4 Antigens/genetics , CD4 Antigens/metabolism , HIV-1/drug effects , HIV-1/genetics , Proviruses/drug effects , Proviruses/genetics , Single-Cell Gene Expression Analysis , HIV Core Protein p24/genetics , HIV Core Protein p24/metabolism , RNA, Long Noncoding/metabolism , Cells, Cultured , Humans , Ionomycin/pharmacology
6.
Vaccines (Basel) ; 11(12)2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38140229

ABSTRACT

BACKGROUND: Suppression of HBV DNA, inhibition of HBV surface (HBsAg) production and therapeutic vaccination to reverse HBV-specific T-cell exhaustion in chronic HBV patients are likely required to achieve a functional cure. In the AAV-HBV mouse model, therapeutic vaccination can be effective in clearing HBV when HBsAg levels are low. Using a single-cell approach, we investigated the liver immune environment with different levels of HBsAg and sustained HBsAg loss through treatment with a GalNAc-HBV-siRNA followed by therapeutic vaccination. METHODS: AAV-HBV-transduced C57BL/6 mice were treated with GalNAc-HBV-siRNA to lower HBsAg levels and then vaccinated using a DNA vaccine. We used single-cell RNA and V(D)J sequencing to understand liver immune microenvironment changes. RESULTS: GalNAc-HBV-siRNA, followed by therapeutic vaccination, achieved sustained HBsAg loss in all mice. This was accompanied by CD4 follicular helper T-cell induction, polyclonal activation of CD8 T cells and clonal expansion of plasma cells that were responsible for antibody production. CONCLUSIONS: This study provides novel insights into liver immune changes at the single-cell level, highlighting the correlation between induced reduction of HBsAg levels and clonal expansion of CD4, CD8 T cells and plasma cells in the liver upon HBV siRNA and subsequent therapeutic vaccination.

7.
Antimicrob Agents Chemother ; 67(11): e0041723, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37874295

ABSTRACT

A major barrier to HIV-1 cure is caused by the pool of latently infected CD4 T-cells that persist under combination antiretroviral therapy (cART). This latent reservoir is capable of producing replication-competent infectious viruses once prolonged suppressive cART is withdrawn. Inducing the reactivation of HIV-1 gene expression in T-cells harboring a latent provirus in people living with HIV-1 under cART may result in depletion of this latent reservoir due to cytopathic effects or immune clearance. Studies have investigated molecules that reactivate HIV-1 gene expression, but to date, no latency reversal agent has been identified to eliminate latently infected cells harboring replication-competent HIV in cART-treated individuals. Stochastic fluctuations in HIV-1 tat gene expression have been described and hypothesized to allow the progression into proviral latency. We hypothesized that exposing latently infected CD4+ T-cells to Tat would result in effective latency reversal. Our results indicate the capacity of a truncated Tat protein and mRNA to reactivate HIV-1 in latently infected T-cells ex vivo to a similar degree as the protein kinase C agonist: phorbol 12-myristate 13-acetate, without T-cell activation or any significant transcriptome perturbation.


Subject(s)
HIV Infections , HIV-1 , Virus Activation , tat Gene Products, Human Immunodeficiency Virus , Humans , CD4-Positive T-Lymphocytes , HIV Infections/genetics , HIV Infections/metabolism , Proviruses/genetics , Virus Latency , Virus Replication , tat Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , HIV-1/genetics , HIV-1/metabolism
8.
Vaccines (Basel) ; 9(9)2021 Aug 29.
Article in English | MEDLINE | ID: mdl-34579206

ABSTRACT

Despite the availability of an effective prophylactic vaccine for more than 30 years, nearly 300 million people worldwide are chronically infected with the hepatitis B virus (HBV), leading to 1 death every 30 s mainly from viral hepatitis-related cirrhosis and liver cancer. Chronic HBV patients exhibit weak, transient, or dysfunctional CD8+ T-cell responses to HBV, which contrasts with high CD8+ T-cell responses seen for resolvers of acute HBV infection. Therefore, a therapeutic DNA vaccine was designed, expressing both HBV core and polymerase proteins, and was sequence optimized to ensure high protein expression and secretion. Although the vaccine, administered intramuscularly via electroporation, had no effect on plasma viral parameters in a mouse model of persistent HBV infection, it did induce robust HBV-specific immune responses in healthy and adeno-associated hepatitis B virus (AAV-HBV) infected mice as well as in healthy non-human primates.

9.
Pharmaceutics ; 11(10)2019 Oct 03.
Article in English | MEDLINE | ID: mdl-31623333

ABSTRACT

Currently, there is an increasing interest to apply pre-fusion (pre-F) protein of respiratory syncytial virus (RSV) as antigen for the development of a subunit vaccine. A pre-F-containing powder would increase the flexibility regarding the route of administration. For instance, a pre-F-containing powder could be incorporated into a single-injection system releasing a primer, and after a lag time, a booster. The most challenging aspect, obtaining the booster after a lag time, may be achieved by incorporating the powder into a core encapsulated by a nonporous poly(dl-lactic-co-glycolic acid) (PLGA) shell. We intended to develop a stable freeze-dried pre-F-containing powder. Furthermore, we investigated whether incorporation of this powder into the core-shell implant was feasible and whether this system would induce a delayed RSV virus-neutralizing antibody (VNA) response in mice. The developed pre-F-containing powder, consisting of pre-F in a matrix of inulin, HEPES, sodium chloride, and Tween 80, was stable during freeze-drying and storage for at least 28 days at 60 °C. Incorporation of this powder into the core-shell implant was feasible and the core-shell production process did not affect the stability of pre-F. An in vitro release study showed that pre-F was incompletely released from the core-shell implant after a lag time of 4 weeks. The incomplete release may be the result of pre-F instability within the core-shell implant during the lag time and requires further research. Mice subcutaneously immunized with a pre-F-containing core-shell implant showed a delayed RSV VNA response that corresponded with pre-F release from the core-shell implant after a lag time of approximately 4 weeks. Moreover, pre-F-containing core-shell implants were able to boost RSV VNA titers of primed mice after a lag time of 4 weeks. These findings could contribute to the development of a single-injection pre-F-based vaccine containing a primer and a booster.

10.
PLoS One ; 13(8): e0202961, 2018.
Article in English | MEDLINE | ID: mdl-30161264

ABSTRACT

A single-injection vaccine formulation that provides for both a prime and a boost immunization would have various advantages over a multiple-injection regime. For such a vaccine formulation, it is essential that the booster dose is released after a certain, preferably adjustable, lag time. In this study we investigated whether a core-shell based implant, containing ovalbumin as core material and poly(DL-lactic-co-glycolic acid) of various monomer ratios as shell material can be used to obtain such a booster release. An in vitro release study showed that the lag time after which the ovalbumin was released from the core-shell implant increased with increasing lactic to glycolic acid ratio of the polymer and ranged from 3-6 weeks. Fluorescence spectroscopy showed minimal differences between native ovalbumin and ovalbumin from core-shell implants that were incubated until just before the observed in vitro release. In addition, mice immunized with a subcutaneous inserted core-shell implant containing ovalbumin showed an ovalbumin-specific IgG1 antibody response after a lag time of 4 or 6-8 weeks. Moreover, delayed release of ovalbumin caused higher IgG1 antibody titers than conventional subcutaneous vaccination with ovalbumin dissolved in PBS. Collectively, these findings could contribute to the further development of a single-injection vaccine, making multiple injections of the vaccine superfluous.


Subject(s)
Immunization , Immunoglobulin G/metabolism , Immunologic Factors/administration & dosage , Ovalbumin/administration & dosage , Animals , Drug Implants , Female , Immunologic Factors/pharmacokinetics , In Vitro Techniques , Mice, Inbred BALB C , Ovalbumin/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Time Factors
11.
J Virol ; 87(20): 11304-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23864630

ABSTRACT

Antigen-presenting cells are a heterogeneous group of cells that are characterized by their functional specialization. Consequently, targeting specific antigen-presenting cell subsets offers opportunities to induce distinct T cell responses. Here we report on the generation and use of nanobodies (Nbs) to target lentivectors specifically to human lymph node-resident myeloid dendritic cells, demonstrating that Nbs represent a powerful tool to redirect lentivectors to human antigen-presenting cell subsets.


Subject(s)
Antigen-Presenting Cells/immunology , Genetic Vectors/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Single-Domain Antibodies/metabolism , Animals , Humans , Lymph Nodes/immunology
12.
Virol J ; 10: 192, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23758776

ABSTRACT

BACKGROUND: Human polyomaviruses (HPyV) infections cause mostly unapparent or mild primary infections, followed by lifelong nonpathogenic persistence. HPyV, and specifically JCPyV, are known to co-diverge with their host, implying a slow rate of viral evolution and a large timescale of virus/host co-existence. Recent bio-informatic reports showed a large level of peptide homology between JCPyV and the human proteome. In this study, the antibody response to PyV peptides is evaluated. METHODS: The in-silico analysis of the HPyV proteome was followed by peptide microarray serology. A HPyV-peptide microarray containing 4,284 peptides was designed and covered 10 polyomavirus proteomes. Plasma samples from 49 healthy subjects were tested against these peptides. RESULTS: In-silico analysis of all possible HPyV 5-mer amino acid sequences were compared to the human proteome, and 1,609 unique motifs are presented. Assuming a linear epitope being as small as a pentapeptide, on average 9.3% of the polyomavirus proteome is unique and could be recognized by the host as non-self. Small t Ag (stAg) contains a significantly higher percentage of unique pentapeptides. Experimental evidence for the presence of antibodies against HPyV 15-mer peptides in healthy subjects resulted in the following observations: i) antibody responses against stAg were significantly elevated, and against viral protein 2 (VP2) significantly reduced; and ii) there was a significant correlation between the increasing number of embedded unique HPyV penta-peptides and the increase in microarray fluorescent signal. CONCLUSION: The anti-peptide HPyV-antibodies in healthy subjects are preferably directed against the penta-peptide derived unique fraction of the viral proteome.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Polyomavirus/immunology , Adult , Antigens, Viral/genetics , DNA, Viral/chemistry , DNA, Viral/genetics , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Polyomavirus/genetics , Protein Array Analysis , Sequence Analysis, DNA , Seroepidemiologic Studies , Viral Proteins/genetics , Viral Proteins/immunology , Young Adult
13.
Mol Ther ; 21(1): 251-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23011030

ABSTRACT

The use of DNA and viral vector-based vaccines for the induction of cellular immune responses is increasingly gaining interest. However, concerns have been raised regarding the safety of these immunization strategies. Due to the lack of their genome integration, mRNA-based vaccines have emerged as a promising alternative. In this study, we evaluated the potency of antigen-encoding mRNA complexed with the cationic lipid 1,2-dioleoyl-3trimethylammonium-propane/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOTAP/DOPE ) as a novel vaccination approach. We demonstrate that subcutaneous immunization of mice with mRNA encoding the HIV-1 antigen Gag complexed with DOTAP/DOPE elicits antigen-specific, functional T cell responses resulting in specific killing of Gag peptide-pulsed cells and the induction of humoral responses. In addition, we show that DOTAP/DOPE complexed antigen-encoding mRNA displays immune-activating properties characterized by secretion of type I interferon (IFN) and the recruitment of proinflammatory monocytes to the draining lymph nodes. Finally, we demonstrate that type I IFN inhibit the expression of DOTAP/DOPE complexed antigen-encoding mRNA and the subsequent induction of antigen-specific immune responses. These results are of high relevance as they will stimulate the design and development of improved mRNA-based vaccination approaches.


Subject(s)
Antigens/immunology , Immunity, Cellular/drug effects , Interferon Type I/pharmacology , RNA, Messenger/immunology , Animals , Mice , RNA, Messenger/administration & dosage
14.
Nanomedicine (Lond) ; 8(1): 77-87, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22891862

ABSTRACT

AIM: Cationic lipids (Lipofectamine™ [Invitrogen, Merelbeke, Belgium] and 1,2-dioleoyl-3-trimethylammonium-propane/1,2-dioleoyl-sn-glycero-3-phosphoethanolamine) and polymers (jetPEI™ and in vivo-jetPEI™ [Polyplus-transfection, Illkirch, France]) were evaluated for their potential to deliver mRNA to monocyte-derived dendritic cells. MATERIALS & METHODS: Lipoplexes and polyplexes, containing mRNA encoding GFP or Gag protein, were incubated with human monocyte-derived dendritic cells and transfection efficiencies were assessed by flow cytometry. RESULTS: Lipofectamine was by far the most efficient in mRNA delivery, therefore it was used in further experiments. Incubation of monocyte-derived dendritic cells isolated from HIV-1-positive donors with mRNA encoding Gag protein complexed to Lipofectamine resulted in 50% transfection. Importantly, coculture of these Gag-transfected dendritic cells with autologous T cells induced an over tenfold expansion of IFN-γ- and IL-2-secreting CD4(+) and CD8(+) T cells. CONCLUSION: Cationic lipid-mediated mRNA delivery may be a useful tool for therapeutic vaccination against HIV-1. This approach can be applied to develop vaccination strategies for other infectious diseases and cancer.


Subject(s)
Dendritic Cells/immunology , Gene Products, gag/genetics , HIV/immunology , RNA, Messenger/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunophenotyping , Microscopy, Fluorescence
15.
Retrovirology ; 9: 72, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22958464

ABSTRACT

Immunotherapy aims to assist the natural immune system in achieving control over viral infection. Various immunotherapy formats have been evaluated in either therapy-naive or therapy-experienced HIV-infected patients over the last 20 years. These formats included non-antigen specific strategies such as cytokines that stimulate immunity or suppress the viral replication, as well as antibodies that block negative regulatory pathways. A number of HIV-specific therapeutic vaccinations have also been proposed, using in vivo injection of inactivated virus, plasmid DNA encoding HIV antigens, or recombinant viral vectors containing HIV genes. A specific format of therapeutic vaccines consists of ex vivo loading of autologous dendritic cells with one of the above mentioned antigenic formats or mRNA encoding HIV antigens.This review provides an extensive overview of the background and rationale of these different therapeutic attempts and discusses the results of trials in the SIV macaque model and in patients. To date success has been limited, which could be explained by insufficient quality or strength of the induced immune responses, incomplete coverage of HIV variability and/or inappropriate immune activation, with ensuing increased susceptibility of target cells.Future attempts at therapeutic vaccination should ideally be performed under the protection of highly active antiretroviral drugs in patients with a recovered immune system. Risks for immune escape should be limited by a better coverage of the HIV variability, using either conserved or mosaic sequences. Appropriate molecular adjuvants should be included to enhance the quality and strength of the responses, without inducing inappropriate immune activation. Finally, to achieve a long-lasting effect on viral control (i.e. a "functional cure") it is likely that these immune interventions should be combined with anti-latency drugs and/or gene therapy.


Subject(s)
HIV Infections/therapy , HIV-1/immunology , Immunotherapy/methods , AIDS Vaccines/administration & dosage , Animals , Anti-Retroviral Agents/administration & dosage , Clinical Trials as Topic , Disease Models, Animal , HIV Infections/virology , Humans , Macaca , Treatment Outcome
16.
Clin Dev Immunol ; 2012: 184979, 2012.
Article in English | MEDLINE | ID: mdl-22844321

ABSTRACT

A variety of immune-based therapies has been developed in order to boost or induce protective CD8(+) T cell responses in order to control HIV replication. Since dendritic cells (DCs) are professional antigen-presenting cells (APCs) with the unique capability to stimulate naïve T cells into effector T cells, their use for the induction of HIV-specific immune responses has been studied intensively. In the present study we investigated whether modulation of the activation state of DCs electroporated with consensus codon-optimized HxB2 gag mRNA enhances their capacity to induce HIV gag-specific T cell responses. To this end, mature DCs were (i) co-electroporated with mRNA encoding interleukin (IL)-12p70 mRNA, or (ii) activated with a cytokine cocktail consisting of R848 and interferon (IFN)-γ. Our results confirm the ability of HxB2 gag-expressing DCs to expand functional HIV-specific CD8(+) T cells. However, although most of the patients had detectable gag-specific CD8(+) T cell responses, no significant differences in the level of expansion of functional CD8(+) T cells could be demonstrated when comparing conventional or immune-modulated DCs expressing IL-12p70. This result which goes against expectation may lead to a re-evaluation of the need for IL-12 expression by DCs in order to improve T-cell responses in HIV-1-infected individuals.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , HIV Infections/immunology , HIV-1 , Interleukin-12/metabolism , gag Gene Products, Human Immunodeficiency Virus/immunology , Antigen Presentation , Dendritic Cells/metabolism , Electroporation , Gene Transfer Techniques , HIV Infections/metabolism , HIV Infections/therapy , HIV-1/immunology , Humans , Imidazoles/pharmacology , Interferon-gamma/pharmacology , Interleukin-12/genetics , Lymphocyte Activation , RNA, Messenger/genetics , RNA, Messenger/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics
17.
PLoS One ; 7(5): e37792, 2012.
Article in English | MEDLINE | ID: mdl-22666392

ABSTRACT

Upon interruption of antiretroviral therapy, HIV-infected patients usually show viral load rebound to pre-treatment levels. Four patients, hereafter referred to as secondary controllers (SC), were identified who initiated therapy during chronic infection and, after stopping treatment, could control virus replication at undetectable levels for more than six months. In the present study we set out to unravel possible viral and immune parameters or mechanisms of this phenomenon by comparing secondary controllers with elite controllers and non-controllers, including patients under HAART. As candidate correlates of protection, virus growth kinetics, levels of intracellular viral markers, several aspects of HIV-specific CD4+ and CD8+ T cell function and HIV neutralizing antibodies were investigated. As expected all intracellular viral markers were lower in aviremic as compared to viremic subjects, but in addition both elite and secondary controllers had lower levels of viral unspliced RNA in PBMC as compared to patients on HAART. Ex vivo cultivation of the virus from CD4+ T cells of SC consistently failed in one patient and showed delayed kinetics in the three others. Formal in vitro replication studies of these three viruses showed low to absent growth in two cases and a virus with normal fitness in the third case. T cell responses toward HIV peptides, evaluated in IFN-γ ELISPOT, revealed no significant differences in breadth, magnitude or avidity between SC and all other patient groups. Neither was there a difference in polyfunctionality of CD4+ or CD8+ T cells, as evaluated with intracellular cytokine staining. However, secondary and elite controllers showed higher proliferative responses to Gag and Pol peptides. SC also showed the highest level of autologous neutralizing antibodies. These data suggest that higher T cell proliferative responses and lower replication kinetics might be instrumental in secondary viral control in the absence of treatment.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/drug effects , HIV-1/pathogenicity , Withholding Treatment , Adult , Aged , Anti-HIV Agents/therapeutic use , Antibodies, Neutralizing/immunology , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Female , Follow-Up Studies , HIV Infections/pathology , HIV Infections/virology , HIV-1/immunology , HIV-1/physiology , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Intracellular Space/virology , Male , Middle Aged , Viral Proteins/metabolism , Virus Replication/drug effects
18.
PLoS One ; 7(6): e38491, 2012.
Article in English | MEDLINE | ID: mdl-22675567

ABSTRACT

Cord blood hematopoietic progenitor cells (CB-HPCs) transplanted immunodeficient NOD/LtsZ-scidIL2Rγ(null) (NSG) and NOD/SCID/IL2Rγ(null) (NOG) mice need efficient human cell engraftment for long-term HIV-1 replication studies. Total body irradiation (TBI) is a classical myeloablation regimen used to improve engraftment levels of human cells in these humanized mice. Some recent reports suggest the use of busulfan as a myeloablation regimen to transplant HPCs in neonatal and adult NSG mice. In the present study, we further ameliorated the busulfan myeloablation regimen with fresh CB-CD34+cell transplantation in 3-4 week old NSG mice. In this CB-CD34+transplanted NSG mice engraftment efficiency of human CD45+cell is over 90% in peripheral blood. Optimal engraftment promoted early and increased CD3+T cell levels, with better lymphoid tissue development and prolonged human cell chimerism over 300 days. These humanized NSG mice have shown long-lasting viremia after HIV-1JRCSF and HIV-1Bal inoculation through intravenous and rectal routes. We also saw a gradual decline of the CD4+T cell count, widespread immune activation, up-regulation of inflammation marker and microbial translocation after HIV-1 infection. Humanized NSG mice reconstituted according to our new protocol produced, moderate cellular and humoral immune responses to HIV-1 postinfection. We believe that NSG mice reconstituted according to our easy to use protocol will provide a better in vivo model for HIV-1 replication and anti-HIV-1 therapy trials.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , HIV/immunology , HIV/physiology , Immunity/immunology , Interleukin Receptor Common gamma Subunit/deficiency , Virus Replication/immunology , Animals , Antigens, CD34/metabolism , Cell Differentiation/immunology , Cell Lineage , Cell Proliferation , Disease Progression , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , Hematopoietic Stem Cells/metabolism , Humans , Interleukin Receptor Common gamma Subunit/immunology , Leukocyte Common Antigens/metabolism , Lymphoid Tissue/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Antigen, T-Cell/metabolism , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/virology
19.
Eur J Immunol ; 42(8): 2019-30, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22585548

ABSTRACT

The pivotal role of DCs in initiating immune responses led to their use as vaccine vectors. However, the relationship between DC subsets involved in antigen presentation and the type of elicited immune responses underlined the need for the characterization of the DCs generated in vitro. The phenotypes of tissue-derived APCs from a cynomolgus macaque model for human vaccine development were compared with ex vivo-derived DCs. Monocyte/macrophages predominated in bone marrow (BM) and blood. Myeloid DCs (mDCs) were present in all tested tissues and were more highly represented than plasmacytoid DCs (pDCs). As in human skin, Langerhans cells (LCs) resided exclusively in the macaque epidermis, expressing CD11c, high levels of CD1a and langerin (CD207). Most DC subsets were endowed with tissue-specific combinations of PRRs. DCs generated from CD34(+) BM cells (CD34-DCs) were heterogeneous in phenotype. CD34-DCs shared properties (differentiation and PRR) of dermal and epidermal DCs. After injection into macaques, CD34-DCs expressing HIV-Gag induced Gag-specific CD4(+) and CD8(+) T cells producing IFN-γ, TNF-α, MIP-1ß, or IL-2. In high responding animals, the numbers of polyfunctional CD8(+) T cells increased with the number of booster injections. This DC-based vaccine strategy elicited immune responses relevant to the DC subsets generated in vitro.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lymphocyte Activation , gag Gene Products, Human Immunodeficiency Virus/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Antigen Presentation , Antigens, CD/biosynthesis , Antigens, CD1/biosynthesis , Antigens, CD34/genetics , Bone Marrow Cells , CD11c Antigen/biosynthesis , Cell Differentiation , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lectins, C-Type/biosynthesis , Macaca fascicularis/immunology , Macrophages , Male , Mannose-Binding Lectins/biosynthesis , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/biosynthesis
20.
Biomaterials ; 33(3): 962-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22018388

ABSTRACT

Despite many advances in modern medicine, human immunodeficiency virus (HIV) still affects the health of millions of people world-wide and much effort is put in developing methods to either prevent infection or to eradicate the virus after infection has occurred. Here, we describe the potential use of electrospun cellulose acetate phthalate (CAP) fibers as a tool to prevent HIV transmission. During the electrospinning process, anti-viral drugs can easily be incorporated in CAP fibers. Interestingly, as a result of the pH-dependent solubility of CAP, the fibers are stable in vaginal fluid (the healthy vaginal flora has a pH of below 4.5), whereas the addition of small amounts of human semen (pH between 7.4 and 8.4) immediately dissolves the fibers which results in the release of the encapsulated drugs. The pH-dependent release properties have been carefully studied and we show that the released anti-viral drugs, together with the CAP which has been reported to have intrinsic antimicrobial activity, efficiently neutralize HIV in vitro.


Subject(s)
Anti-HIV Agents/administration & dosage , Cellulose/analogs & derivatives , Semen/chemistry , Administration, Intravaginal , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Cell Line , Cellulose/chemistry , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...