Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Stem Cell Res Ther ; 14(1): 259, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37726837

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) have broad potential as a cell therapy including for the treatment of drug-resistant inflammatory conditions with abnormal T cell proliferation such as graft-versus-host disease (GVHD). Clinical success, however, has been complicated by the heterogeneity of culture-expanded MSCs as well as donor variability. Here, we devise culture conditions that promote expansion of MSCs with enhanced immunomodulatory functions both in vitro and in animal models of GVHD. METHODS: Human bone marrow-derived MSCs were expanded at high-confluency (MSCHC) and low-confluency state (MSCLC). Their immunomodulatory properties were evaluated with in vitro co-culture assays based on suppression of activated T cell proliferation and secretion of pro-inflammatory cytokines from activated T cells. Metabolic state of these cells was determined, while RNA sequencing was performed to explore transcriptome of these MSCs. Ex vivo expanded MSCHC or MSCLC was injected into human peripheral blood mononuclear cells (PBMC)-induced GVHD mouse model to determine their in vivo therapeutic efficacy based on clinical grade scoring, human CD45+ blood count and histopathological examination. RESULTS: As compared to MSCLC, MSCHC significantly reduced both the proliferation of anti-CD3/CD28-activated T cells and secretion of pro-inflammatory cytokines upon MSCHC co-culture across several donors even in the absence of cytokine priming. Mechanistically, metabolic analysis of MSCHC prior to co-culture with activated T cells showed increased glycolytic metabolism and lactate secretion compared to MSCLC, consistent with their ability to inhibit T cell proliferation. Transcriptome analysis further revealed differential expression of immunomodulatory genes including TRIM29, BPIFB4, MMP3 and SPP1 in MSCHC as well as enriched pathways including cytokine-cytokine receptor interactions, cell adhesion and PI3K-AKT signalling. Lastly, we demonstrate in a human PBMC-induced GVHD mouse model that delivery of MSCHC showed greater suppression of inflammation and improved outcomes compared to MSCLC and saline controls. CONCLUSION: Our study provides evidence that ex vivo expansion of MSCs at high confluency alters the metabolic and transcriptomic states of these cells. Importantly, this approach maximizes the production of MSCs with enhanced immunomodulatory functions without priming, thus providing a non-invasive and generalizable strategy for improving the use of MSCs for the treatment of inflammatory diseases.


Subject(s)
Leukocytes, Mononuclear , Mesenchymal Stem Cells , Animals , Mice , Humans , Bone Marrow , Phosphatidylinositol 3-Kinases , Cytokines , Disease Models, Animal , DNA-Binding Proteins , Transcription Factors , Intercellular Signaling Peptides and Proteins
2.
J Allergy Clin Immunol ; 150(6): 1415-1426.e9, 2022 12.
Article in English | MEDLINE | ID: mdl-35917932

ABSTRACT

BACKGROUND: Patients with asthma often suffer from frequent respiratory viral infections and reduced virus clearance. Lung resident memory T cells provide rapid protection against viral reinfections. OBJECTIVE: Because the development of resident memory T cells relies on the lung microenvironment, we investigated the impact of allergen sensitization on the development of virus-specific lung resident memory T cells and viral clearance. METHODS: Mice were sensitized with house dust mite extract followed by priming with X47 and a subsequent secondary influenza infection. Antiviral memory T-cell response and protection to viral infection was assessed before and after secondary influenza infection, respectively. Gene set variation analysis was performed on data sets from the U-BIOPRED asthma cohort using an IFN-γ-induced epithelial cell signature and a tissue resident memory T-cell signature. RESULTS: Viral loads were higher in lungs of sensitized compared with nonsensitized mice after secondary infection, indicating reduced virus clearance. X47 priming induced fewer antiviral lung resident memory CD8 T cells and resulted in lower pulmonary IFN-γ levels in the lungs of sensitized as compared with nonsensitized mice. Using data from the U-BIOPRED cohort, we found that patients with enrichment of epithelial IFN-γ-induced genes in nasal brushings and bronchial biopsies were also enriched in resident memory T-cell-associated genes, had more epithelial CD8 T cells, and reported significantly fewer exacerbations. CONCLUSIONS: The allergen-sensitized lung microenvironment interferes with the formation of antiviral resident memory CD8 T cells in lungs and virus clearance. Defective antiviral memory response might contribute to increased susceptibility of patients with asthma to viral exacerbations.


Subject(s)
Influenza, Human , Memory T Cells , Mice , Animals , Humans , Lung , CD8-Positive T-Lymphocytes , Allergens
3.
Mucosal Immunol ; 15(4): 717-729, 2022 04.
Article in English | MEDLINE | ID: mdl-35260804

ABSTRACT

The fate of tissue-resident memory CD4 T cells (Trm) has been incompletely investigated. Here we show that intranasal, but not parenteral, immunization with CTA1-3M2e-DD stimulated M2e-specific Th17 Trm cells, which conferred strong protection against influenza virus infection in the lung. These cells rapidly expanded upon infection and effectively restricted virus replication as determined by CD4 T cell depletion studies. Single-cell RNAseq transcriptomic and TCR VDJ-analysis of M2e-tetramer-sorted CD4 T cells on day 3 and 8 post infection revealed complete Th17-lineage dominance (no Th1 or Tregs) with extensive functional diversity and expression of gene markers signifying mature resident Trm cells (Cd69, Nfkbid, Brd2, FosB). Unexpectedly, the same TCR clonotype hosted cells with different Th17 subcluster functions (IL-17, IL-22), regulatory and cytotoxic cells, suggesting a tissue and context-dependent differentiation of reactivated Th17 Trm cells. A gene set enrichment analysis demonstrated up-regulation of regulatory genes (Lag3, Tigit, Ctla4, Pdcd1) in M2e-specific Trm cells on day 8, indicating a tissue damage preventing function. Thus, contrary to current thinking, lung M2e-specific Th17 Trm cells are sufficient for controlling infection and for protecting against tissue injury. These findings will have strong implications for vaccine development against respiratory virus infections and influenza virus infections, in particular.


Subject(s)
Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Humans , Immunologic Memory , Lung , Receptors, Antigen, T-Cell , Th17 Cells
4.
Front Immunol ; 12: 749325, 2021.
Article in English | MEDLINE | ID: mdl-34659250

ABSTRACT

Previous work showed that interferon-λ (IFN-λ) can trigger the synthesis of thymic stromal lymphopoietin (TSLP) by specialized epithelial cells in the upper airways of mice, thereby improving the performance of intranasally administered influenza vaccines. Here we demonstrate that protein-only influenza vaccines containing either IFN-λ or TSLP boosted antigen-specific IgG1 and IgA responses and enhanced the resistance of mice to influenza virus challenge, irrespective of whether the vaccines were applied via the intranasal or the rectal route. TSLP receptor deficiency negatively influenced vaccine-induced antiviral immunity by impairing the migration of dendritic cells from the airways to the draining lymph nodes of immunized mice, thereby restraining follicular helper T cell and germinal center B cell responses. As previously observed during intranasal vaccination, the adjuvant effect of IFN-λ on a rectally administered influenza vaccine was no longer observed when TSLP receptor-deficient mice were used for immunization, highlighting the central role of the IFN-λ/TSLP axis for vaccine-induced antiviral immunity in the mucosa.


Subject(s)
Cytokines/administration & dosage , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Influenza Vaccines/administration & dosage , Interferons/administration & dosage , Orthomyxoviridae Infections/prevention & control , Vaccines, Subunit/administration & dosage , Administration, Intranasal , Administration, Rectal , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Bronchoalveolar Lavage Fluid/immunology , Female , Immunoglobulins/genetics , Influenza A virus , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytokine/genetics , Thymic Stromal Lymphopoietin
5.
J Gen Virol ; 102(3)2021 03.
Article in English | MEDLINE | ID: mdl-33410734

ABSTRACT

The complement alternative pathway (AP) is tightly regulated and changes in two important AP components, factor B (FB) and factor H (FH) are linked to severe dengue in humans. Here, a mouse model of dengue was investigated to define the changes in FB and FH and assess the utility of this model to study the role of the AP in severe dengue. Throughout the period of viremia in the AG129 IFN signalling-deficient mouse, an increase in FB and a decrease in FH was observed following dengue virus (DENV) infection, with the former only seen in a model of more severe disease associated with antibody-dependent enhancement (ADE). Terminal disease was associated with a decrease in FB and FH, with greater changes during ADE, and accompanied by increased C3 degradation consistent with complement activation. In silico analysis of NFκΒ, signal transducer and activator of transcription (STAT) and IFN-driven FB and FH promoter elements to reflect the likely impact of the lack of IFN-responses in AG129 mice, demonstrated that these elements differed markedly between human and mouse, notably with mouse FH lacking NFκΒ and key IFN-stimulated response elements (ISRE), and FB with many more NFκΒ and STAT-responsive elements than human FB. Thus, the AG129 mouse offers utility in demonstrating changes in FB and FH that, similar to humans, are associated with severe disease, but lack predicted important human-specific and IFN-dependent responses of FB and FH to DENV-infection that are likely to regulate the subtleties of the overall AP response during dengue disease in humans.


Subject(s)
Complement Factor B/metabolism , Complement Factor H/metabolism , Complement Pathway, Alternative , Dengue/immunology , Severe Dengue/immunology , Animals , Antibody-Dependent Enhancement , Complement Factor B/genetics , Complement Factor H/genetics , Dengue/virology , Dengue Virus/immunology , Dengue Virus/physiology , Disease Models, Animal , Humans , Interferons/metabolism , Mice , Promoter Regions, Genetic , Severe Dengue/virology , Viremia
6.
Mucosal Immunol ; 14(2): 523-536, 2021 03.
Article in English | MEDLINE | ID: mdl-32807838

ABSTRACT

This is a proof-of-principle study demonstrating that the combination of a cholera toxin derived adjuvant, CTA1-DD, and lipid nanoparticles (LNP) can significantly improve the immunogenicity and protective capacity of an intranasal vaccine. We explored the self-adjuvanted universal influenza vaccine candidate, CTA1-3M2e-DD (FPM2e), linked to LNPs. We found that the combined vector greatly enhanced survival against a highly virulent PR8 strain of influenza virus as compared to when mice were immunized with FPM2e alone. The combined vaccine vector enhanced early endosomal processing and peptide presentation in dendritic cells and upregulated co-stimulation. The augmenting effect was CTA1-enzyme dependent. Whereas systemic anti-M2e antibody and CD4+ T-cell responses were comparable to those of the soluble protein, the local respiratory tract IgA and the specific Th1 and Th17 responses were strongly enhanced. Surprisingly, the lung tissue did not exhibit gross pathology upon recovery from infection and M2e-specific lung resident CD4+ T cells were threefold higher than in FPM2e-immunized mice. This study conveys optimism as to the protective ability of a combination vaccine based on LNPs and various forms of the CTA1-DD adjuvant platform, in general, and, more specifically, an important way forward to develop a universal vaccine against influenza.


Subject(s)
Cholera Toxin/immunology , Influenza A virus/physiology , Influenza Vaccines/immunology , Influenza, Human/immunology , Liposomes/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Recombinant Fusion Proteins/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Administration, Intranasal , Animals , Antigen Presentation , Cells, Cultured , Cholera Toxin/metabolism , Humans , Immunogenicity, Vaccine , Immunoglobulin A/metabolism , Influenza Vaccines/metabolism , Liposomes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/metabolism , Peptides, Cyclic , Recombinant Fusion Proteins/metabolism , Vaccination
7.
J Exp Med ; 217(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32584412

ABSTRACT

Dengue is a major public health concern in the tropical and subtropical world, with no effective treatment. The controversial live attenuated virus vaccine Dengvaxia has boosted the pursuit of subunit vaccine approaches, and nonstructural protein 1 (NS1) has recently emerged as a promising candidate. However, we found that NS1 immunization or passive transfer of NS1 antibodies failed to confer protection in symptomatic dengue mouse models using two non-mouse-adapted DENV2 strains that are highly virulent. Exogenous administration of purified NS1 also failed to worsen in vivo vascular leakage in sublethally infected mice. Neither method of NS1 immune neutralization changed the disease outcome of a chimeric strain expressing a vascular leak-potent NS1. Instead, virus chimerization involving the prME structural region indicated that these proteins play a critical role in driving in vivo fitness and virulence of the virus, through induction of key proinflammatory cytokines. This work highlights that the pathogenic role of NS1 is DENV strain dependent, which warrants reevaluation of NS1 as a universal dengue vaccine candidate.


Subject(s)
Dengue Virus/pathogenicity , Viral Nonstructural Proteins/metabolism , Animals , Antibodies, Viral/immunology , Antibody-Dependent Enhancement/immunology , Capillary Permeability , Chimera , Cytokines/metabolism , Dengue/blood , Dengue/immunology , Dengue/virology , Dengue Virus/immunology , Dengue Virus/isolation & purification , Disease Models, Animal , Immunity , Immunization , Immunoglobulin G/blood , Mice , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/metabolism , Severity of Illness Index , Viremia/immunology , Virulence
8.
J Gen Virol ; 101(1): 79-85, 2020 01.
Article in English | MEDLINE | ID: mdl-31774391

ABSTRACT

Dengue virus (DENV) infection is associated with clinical ocular presentations and here DENV infection of the eye was assessed in mice. In an AG129 mouse model of antibody-dependent enhancement of DENV infection, DENV RNA was detected in the eye and vascular changes were present in the retinae. Intraocular CD8 and IFN-γ mRNA were increased in mice born to DENV-naïve, but not DENV-immune mothers, while TNF-α mRNA was induced and significantly higher in mice born to DENV-immune than DENV-naïve mothers. DENV RNA was detected in the eye following intracranial DENV infection and CD8 mRNA but not IFN-γ nor TNF-α were induced. In all models, viperin was increased following DENV infection. Thus, DENV in the circulation or the brain can infect the eye and stimulate innate immune responses, with induction of viperin as one response that consistently occurs in multiple DENV eye-infection models in both an IFN-dependent and independent manner.


Subject(s)
Dengue Virus/immunology , Dengue/immunology , Eye Infections, Viral/immunology , Eye Infections, Viral/virology , Inflammation/immunology , Inflammation/virology , Animals , Antibody-Dependent Enhancement/immunology , Dengue/virology , Disease Models, Animal , Eye/immunology , Eye/virology , Immunity, Innate/immunology , Interferon-gamma/immunology , Mice , Tumor Necrosis Factor-alpha/immunology
9.
J Virol ; 93(23)2019 12 01.
Article in English | MEDLINE | ID: mdl-31511392

ABSTRACT

Type I and type III interferons (IFNs) can promote adaptive immune responses in mice and improve vaccine-induced resistance to viral infections. The adjuvant effect of type III IFN (IFN-λ) specifically boosts mucosal immunity by an indirect mechanism, involving IFN-λ-induced production of thymic stromal lymphopoietin (TSLP), a cytokine that activates immune cells. To date, it remained unclear whether the previously described adjuvant effect of type I IFN (IFN-α/ß) would also depend on TSLP and whether type I IFN stimulates different antibody subtypes. Here, we show that after infection with a live attenuated influenza virus, mice lacking functional type I IFN receptors failed to produce normal amounts of virus-specific IgG2c and IgA antibodies. In contrast, mice lacking functional IFN-λ receptors contained normal levels of virus-specific IgG2c but had reduced IgG1 and IgA antibody levels. When applied together with protein antigen, IFN-α stimulated the production of antigen-specific IgA and IgG2c to a greater extent than IgG1, irrespective of whether the mice expressed functional TSLP receptors and irrespective of whether the vaccine was applied by the intranasal or the intraperitoneal route. Taken together, these results demonstrate that the adjuvant activities of type I and type III IFNs are mechanistically distinct.IMPORTANCE Interferons can shape antiviral immune responses, but it is not well understood how they influence vaccine efficacy. We find that type I IFN preferentially promotes the production of antigen-specific IgG2c and IgA antibodies after infection with a live attenuated influenza virus or after immunization with influenza subunit vaccines. In contrast, type III IFN specifically enhances influenza virus-specific IgG1 and IgA production. The adjuvant effect of type I IFN was not dependent on TSLP, which is essential for the adjuvant effect of type III IFN. Type I IFN boosted vaccine-induced antibody production after immunization by the intranasal or the intraperitoneal route, whereas type III IFN exhibited its adjuvant activity only when the vaccine was delivered by the mucosal route. Our findings demonstrate that type I and type III IFNs trigger distinct pathways to enhance the efficacy of vaccines. This knowledge might be used to design more efficient vaccines against infectious diseases.


Subject(s)
Adaptive Immunity/immunology , Adjuvants, Immunologic , Influenza Vaccines/immunology , Interferons/immunology , Animals , Antibody Formation/immunology , Cytokines , Disease Models, Animal , Female , Immunity, Mucosal/immunology , Immunization , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulins/genetics , Interferon Type I , Interferons/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/virology , Receptors, Cytokine/genetics , Vaccination , Interferon Lambda , Thymic Stromal Lymphopoietin
10.
PLoS Negl Trop Dis ; 12(2): e0006209, 2018 02.
Article in English | MEDLINE | ID: mdl-29425203

ABSTRACT

Dengue virus (DENV) infection imposes enormous health and economic burden worldwide with no approved treatment. Several small molecules, including lovastatin, celgosivir, balapiravir and chloroquine have been tested for potential anti-dengue activity in clinical trials; none of these have demonstrated a protective effect. Recently, based on identification and characterization of cross-serotype neutralizing antibodies, there is increasing attention on the potential for dengue immunotherapy. Here, we tested the ability of VIS513, an engineered cross-neutralizing humanized antibody targeting the DENV E protein domain III, to overcome antibody-enhanced infection and high but brief viremia, which are commonly encountered in dengue patients, in various in vitro and in vivo models. We observed that VIS513 efficiently neutralizes DENV at clinically relevant viral loads or in the presence of enhancing levels of DENV immune sera. Single therapeutic administration of VIS513 in mouse models of primary infection or lethal secondary antibody-enhanced infection, reduces DENV titers and protects from lethal infection. Finally, VIS513 administration does not readily lead to resistance, either in cell culture systems or in animal models of dengue infection. The findings suggest that rapid viral reduction during acute DENV infection with a monoclonal antibody is feasible.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dengue Virus/immunology , Dengue/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/administration & dosage , Antibodies, Viral/blood , Antibodies, Viral/genetics , Antigens, Viral/chemistry , Antigens, Viral/genetics , Cell Line , Chlorocebus aethiops , Cross Reactions/immunology , Dengue Virus/genetics , Dengue Virus/pathogenicity , Disease Models, Animal , Epitopes , Female , Humans , Immune Sera , Immunotherapy , In Vitro Techniques , Mice , Models, Structural , Mutation , Neutralization Tests , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Serogroup , THP-1 Cells , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Plaque Assay
11.
PLoS Negl Trop Dis ; 10(6): e0004805, 2016 06.
Article in English | MEDLINE | ID: mdl-27341339

ABSTRACT

Dengue virus (DENV) causes a spectrum of diseases ranging from self-limiting dengue fever to severe conditions such as haemorrhagic fever and dengue shock syndrome. Antibody-dependent enhancement (ADE) is thought to explain the occurrence of severe dengue whereby pre-existing binding but non-neutralising antibodies enhance DENV infection. The ADE phenomenon is supported by epidemiological findings that infants that born to dengue immune mothers are at greater risk to develop severe dengue upon primary infection. The role of maternally acquired dengue-specific antibodies in disease enhancement was recently recapitulated in a mouse model where mice born to DENV1-immune mothers experienced enhanced disease severity upon DENV2 infection. Here, this study investigates the relative contribution of maternal dengue-specific antibodies acquired during gestation and breastfeeding in dengue disease. Using a surrogate breastfeeding mother experimental approach, we showed that majority of the maternal dengue-specific antibodies were acquired during breastfeeding and conferred an extended enhancement window. On the other hand, in the context of homologous infection, breastfeeding conferred protection. Furthermore, measurement of dengue-specific antibody titres over time in mice born to dengue immune mothers revealed a biphasic pattern of antibody decay as reported in humans. Our work provides evidence of the potential contribution of breast milk-acquired dengue-specific IgG antibodies in enhancement and protection against dengue. Should such contribution be established in humans as well, it may have important implications for the development of guidelines to dengue-immune breastfeeding mothers.


Subject(s)
Antibodies, Viral/immunology , Dengue Virus/immunology , Dengue/immunology , Immunoglobulin G/immunology , Interferon Type I/metabolism , Pregnancy Complications, Infectious/immunology , Aging , Animals , Breast Feeding , Cell Line , Cricetinae , Dengue/pathology , Dengue/prevention & control , Female , Interferon Type I/genetics , Lactation , Mice , Mice, Knockout , Pregnancy , Pregnancy Complications, Infectious/pathology , Pregnancy Complications, Infectious/prevention & control , Viral Plaque Assay
12.
PLoS Negl Trop Dis ; 10(3): e0004536, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27007501

ABSTRACT

Epidemiological studies have reported that most of the severe dengue cases occur upon a secondary heterologous infection. Furthermore, babies born to dengue immune mothers are at greater risk of developing severe disease upon primary infection with a heterologous or homologous dengue virus (DENV) serotype when maternal antibodies reach sub-neutralizing concentrations. These observations have been explained by the antibody mediated disease enhancement (ADE) phenomenon whereby heterologous antibodies or sub-neutralizing homologous antibodies bind to but fail to neutralize DENV particles, allowing Fc-receptor mediated entry of the virus-antibody complexes into host cells. This eventually results in enhanced viral replication and heightened inflammatory responses. In an attempt to replicate this ADE phenomenon in a mouse model, we previously reported that upon DENV2 infection 5-week old type I and II interferon (IFN) receptors-deficient mice (AG129) born to DENV1-immune mothers displayed enhancement of disease severity characterized by increased virus titers and extensive vascular leakage which eventually led to the animals' death. However, as dengue occurs in immune competent individuals, we sought to reproduce this mouse model in a less immunocompromised background. Here, we report an ADE model that is mediated by maternal antibodies in type I IFN receptor-deficient A129 mice. We show that 5-week old A129 mice born to DENV1-immune mothers succumbed to a DENV2 infection within 4 days that was sub-lethal in mice born to naïve mothers. Clinical manifestations included extensive hepatocyte vacuolation, moderate vascular leakage, lymphopenia, and thrombocytopenia. Anti-TNFα therapy totally protected the mice and correlated with healthy hepatocytes. In contrast, blocking IL-6 did not impact the virus titers or disease outcome. This A129 mouse model of ADE may help dissecting the mechanisms involved in dengue pathogenesis and evaluate the efficacy of vaccine and therapeutic candidates.


Subject(s)
Antibodies, Viral/blood , Dengue/immunology , Interferon Type I/metabolism , Liver Diseases/etiology , Animals , Cytokines/metabolism , Dengue/genetics , Dengue/mortality , Dengue Virus/classification , Female , Gene Deletion , Immunoglobulin G/blood , Interferon Type I/genetics , Mice , Receptors, Interferon/genetics , Receptors, Interferon/metabolism
13.
Expert Rev Vaccines ; 15(4): 483-95, 2016.
Article in English | MEDLINE | ID: mdl-26508565

ABSTRACT

Despite 70 years of research that has intensified in the past decade, a safe and efficacious dengue vaccine has yet to be available. In addition to the expected challenges such as identifying immune correlates of protection, the dengue vaccine field has faced additional hurdles including the necessity to design a tetravalent formulation and the risk of antibody-mediated disease enhancement. Nevertheless, tetravalent live attenuated vaccine candidates have reached efficacy trials and demonstrated some benefit, despite imbalanced immunogenicity and incomplete protection against the four serotypes. Meanwhile, the development of sub-unit dengue vaccines has gained momentum. As the target of most of the neutralizing antibodies so far reported, the virus envelope E protein has been the focus of much effort and represents the leading dengue sub-unit vaccine candidate. However, its notorious poor immunogenicity has prompted the development of innovative approaches to make E-derived constructs part of the second generation dengue vaccines portfolio.


Subject(s)
Dengue Vaccines/immunology , Dengue Vaccines/isolation & purification , Drug Discovery/methods , Drug Discovery/trends , Humans , Vaccines, Attenuated/immunology , Vaccines, Attenuated/isolation & purification , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification , Viral Envelope Proteins/immunology
14.
Cell ; 162(3): 493-504, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26189681

ABSTRACT

Dengue is the most common vector-borne viral disease, causing nearly 400 million infections yearly. Currently there are no approved therapies. Antibody epitopes that elicit weak humoral responses may not be accessible by conventional B cell panning methods. To demonstrate an alternative strategy to generating a therapeutic antibody, we employed a non-immunodominant, but functionally relevant, epitope in domain III of the E protein, and engineered by structure-guided methods an antibody directed to it. The resulting antibody, Ab513, exhibits high-affinity binding to, and broadly neutralizes, multiple genotypes within all four serotypes. To assess therapeutic relevance of Ab513, activity against important human clinical features of dengue was investigated. Ab513 mitigates thrombocytopenia in a humanized mouse model, resolves vascular leakage, reduces viremia to nearly undetectable levels, and protects mice in a maternal transfer model of lethal antibody-mediated enhancement. The results demonstrate that Ab513 may reduce the public health burden from dengue.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/chemistry , Dengue Virus/physiology , Dengue/therapy , Immunodominant Epitopes/chemistry , Amino Acid Sequence , Animals , Dengue/immunology , Dengue/virology , Dengue Virus/immunology , Disease Models, Animal , Mice , Models, Molecular , Molecular Sequence Data , Phagocytosis , Protein Engineering , Receptors, Fc/immunology , Sequence Alignment
15.
Biomaterials ; 35(9): 2987-98, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24412082

ABSTRACT

Autofluorescence, photodamage and photobleaching are often encountered when using downconverting fluorophores and fluorescent proteins for bacteria labeling. These caveats represent a serious limitation when trying to map bacteria dissemination for prolonged periods. Upconversion nanoparticles (UCNs), which are able to convert low energy near-infrared (NIR) excitation light into higher energy visible or NIR light, can address these limitations. These particles' unique optical properties translate into attractive advantages of minimal autofluorescence, reduced photodamage, deeper tissue penetration and prolonged photostability. Here, we report a UCN-based bacteria labeling strategy using Escherichia coli as prototypic bacteria. A comparative analysis highlighted the superior photostability of UCN-labeled bacteria over green fluorescent protein-expressing bacteria. Infection study of UCN-labeled bacteria in dendritic cells indicated co-localization of the UCN signal with bacterial position for up to 6 h post-infection. Furthermore, long-term monitoring of the same infected cells demonstrated the potential to utilize photostable UCN-based imaging for bacterial trafficking purposes.


Subject(s)
Escherichia coli/metabolism , Fluorescent Dyes/metabolism , Nanoparticles/chemistry , Optical Imaging/methods , Animals , Antibodies/metabolism , Cell Line , Citrates/chemistry , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Escherichia coli/ultrastructure , Green Fluorescent Proteins/metabolism , Mice , Nanoparticles/ultrastructure , Solutions , Staining and Labeling
16.
Nanomedicine (Lond) ; 6(7): 1273-88, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21929461

ABSTRACT

Upconversion nanoparticles (UCNs) are an emerging class of luminescent nanomaterials, exhibiting many advantages over conventional fluorophores, such as high signal-to-noise ratio and superior photostability. The near-infrared excitation wavelengths of these particles offer additional advantages such as deep tissue penetration and low photodamage to biological samples. In the last 5 years, with the advances in nanoparticles synthesis and modification technology, much research has been performed to exploit UCNs' advantages and integrate them into various biological applications. This review focuses on the recent developments of UCNs as imaging, detection and therapeutic tools, highlighting the respective strategies adopted.


Subject(s)
Diagnostic Imaging/methods , Luminescent Agents/therapeutic use , Nanomedicine/methods , Nanoparticles/therapeutic use , Animals , Humans , Luminescent Agents/chemistry , Nanoparticles/chemistry
17.
Luminescence ; 25(4): 290-3, 2010.
Article in English | MEDLINE | ID: mdl-20737581

ABSTRACT

Upconversion nanoparticles (UCNs), in the recent times have attracted attention due to their unique properties, which makes them ideal fluorophores for use in biological applications. There have been various reports on their use for targeted cell imaging, drug and gene delivery and also for diffuse optical tomography. Here we give a brief introduction on what are UCNs and the mechanism of upconversion, followed by a discussion on the biological applications of UCNs and further on what the future holds for UCNs.


Subject(s)
Fluorescence , Nanoparticles/chemistry , Animals , Cell Line , Humans , Luminescent Measurements , Photons , Quantum Dots , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...