Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 12(10): 966-74, 2011 Sep 04.
Article in English | MEDLINE | ID: mdl-21892173

ABSTRACT

Natural killer T cells (NKT cells) recognize glycolipid antigens presented by CD1d. These cells express an evolutionarily conserved, invariant T cell antigen receptor (TCR), but the forces that drive TCR conservation have remained uncertain. Here we show that NKT cells recognized diacylglycerol-containing glycolipids from Streptococcus pneumoniae, the leading cause of community-acquired pneumonia, and group B Streptococcus, which causes neonatal sepsis and meningitis. Furthermore, CD1d-dependent responses by NKT cells were required for activation and host protection. The glycolipid response was dependent on vaccenic acid, which is present in low concentrations in mammalian cells. Our results show how microbial lipids position the sugar for recognition by the invariant TCR and, most notably, extend the range of microbes recognized by this conserved TCR to several clinically important bacteria.


Subject(s)
Glycolipids/immunology , Gram-Positive Bacteria/immunology , Natural Killer T-Cells/immunology , Animals , Antigens, CD1d/chemistry , Antigens, CD1d/physiology , Cell Line , Glycolipids/chemistry , Humans , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/metabolism
2.
Emerg Infect Dis ; 28(1): 196-200, 2022 01.
Article in English | MEDLINE | ID: mdl-34647864

ABSTRACT

We report severe acute respiratory syndrome coronavirus 2 in semen by using quantitative reverse transcription PCR during the late convalescent phase. Virus was associated with adequate humoral and cell-mediated responses, suggesting possible seeding of the immune-privileged testes. We provide longitudinal semen quality data for 6 other men, including 3 who had oligozoospermia.


Subject(s)
COVID-19 , Oligospermia , Humans , Male , RNA, Viral/genetics , SARS-CoV-2 , Semen , Semen Analysis , Virus Shedding
3.
Am J Transplant ; 22(2): 649-653, 2022 02.
Article in English | MEDLINE | ID: mdl-34510730

ABSTRACT

Unlike immunocompetent hosts, the duration of viral persistence after infection with severe acute respiratory syndrome coronavirus 2 can be prolonged in immunosuppressed patients. Here, we present a case of viral persistence for over 19 weeks in a patient with a history of solid organ transplant and explore the clinical, virologic, and immunologic course. Our patient still demonstrated viral persistence at 138 days with low polymerase chain reaction cycle threshold values and evidence of continuing viral sequence evolution indicative of ongoing virus replication. These findings have important implications for infection prevention and control recommendations in immunosuppressed patients. Immune response, including neutralizing antibody titers, T cell activity, and cytokine levels, peaked around days 44-72 after diagnosis. Anti-S trimer antibodies were low at all time points, and T cell response was attenuated by day 119. As immune response waned and viral load increased, increased genetic diversity emerged, suggesting a mechanism for the development of viral variants.


Subject(s)
COVID-19 , Organ Transplantation , Antibodies, Viral , Humans , Organ Transplantation/adverse effects , SARS-CoV-2 , Viral Load
4.
J Infect Dis ; 221(2): 201-213, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31647546

ABSTRACT

BACKGROUND: For the purpose of studying functional human dendritic cells (DCs) in a humanized mouse model that mimics the human immune system (HIS), a model referred to as HIS mice was established. METHODS: Human immune system mice were made by engrafting NOD/SCID/IL2Rgammanull (NSG) mice with human hematopoietic stem cells (HSCs) following the transduction of genes encoding human cytokines and human leukocyte antigen (HLA)-A2.1 by adeno-associated virus serotype 9 (AAV9) vectors. RESULTS: Our results indicate that human DC subsets, such as CD141+CD11c+ and CD1c+CD11c+ myeloid DCs, distribute throughout several organs in HIS mice including blood, bone marrow, spleen, and draining lymph nodes. The CD141+CD11c+ and CD1c+CD11c+ human DCs isolated from HIS mice immunized with adenoviruses expressing malaria/human immunodeficiency virus (HIV) epitopes were able to induce the proliferation of malaria/HIV epitopes-specific human CD8+ T cells in vitro. Upregulation of CD1c was also observed in human CD141+ DCs 1 day after immunization with the adenovirus-based vaccines. CONCLUSIONS: Establishment of such a humanized mouse model that mounts functional human DCs enables preclinical assessment of the immunogenicity of human vaccines in vivo.


Subject(s)
Adenovirus Vaccines/immunology , Antigens, Surface/immunology , Dendritic Cells/immunology , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Thrombomodulin
5.
Chemistry ; 26(20): 4476-4479, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-31788876

ABSTRACT

α-Galactosylceramides are glycosphingolipids that show promise in cancer immunotherapy. After presentation by CD1d, they activate natural killer T cells (NKT), which results in the production of a variety of pro-inflammatory and immunomodulatory cytokines. Herein, we report the synthesis and biological evaluation of photochromic derivatives of KRN-7000, the activity of which can be modulated with light. Based on established structure-activity relationships, we designed photoswitchable analogues of this glycolipid that control the production of pro-inflammatory cytokines, such as IFN-γ. The azobenzene derivative α-GalACer-4 proved to be more potent than KRN-7000 itself when activated with 370 nm light. Photolipids of this type could improve our mechanistic understanding of cytokine production and could open new directions in photoimmunotherapy.


Subject(s)
Antigens, CD1d/metabolism , Cytokines/chemistry , Galactosylceramides/pharmacology , Glycolipids/chemistry , Killer Cells, Natural/drug effects , Antigens, CD1d/chemistry , Cytokines/metabolism , Galactosylceramides/chemistry , Killer Cells, Natural/chemistry , Natural Killer T-Cells , Structure-Activity Relationship
6.
J Virol ; 90(10): 5068-5074, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26962219

ABSTRACT

UNLABELLED: Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract disease, which causes high rates of morbidity and mortality in infants and the elderly. Models of human RSV pulmonary disease are needed to better understand RSV pathogenesis and to assess the efficacy of RSV vaccines. We assessed the RSV-specific human innate, humoral, and cellular immune responses in humanized mice (mice with a human immune system [HIS mice]) with functional human CD4(+) T and B cells. These mice were generated by introduction of HLA class II genes, various human cytokines, and human B cell activation factor into immunodeficient NOD scid gamma (NSG) mice by the use of an adeno-associated virus vector, followed by engraftment of human hematopoietic stem cells. During the first 3 days of infection, HIS mice lost more weight and cleared RSV faster than NSG mice. Human chemokine (C-C motif) ligand 3 (CCL3) and human interleukin-1ß (IL-1ß) expression was detected in the RSV-infected HIS mice. The pathological features induced by RSV infection in HIS mice included peribronchiolar inflammation, neutrophil predominance in the bronchioalveolar lavage fluid, and enhanced airway mucus production. Human anti-RSV IgG and RSV-neutralizing antibodies were detected in serum and human anti-RSV mucosal IgA was detected in bronchioalveolar lavage fluid for up to 6 weeks. RSV infection induced an RSV-specific human gamma interferon response in HIS mouse splenocytes. These results indicate that human immune cells can induce features of RSV lung disease, including mucus hyperplasia, in murine lungs and that HIS mice can be used to elicit human anti-RSV humoral and cellular immunity. IMPORTANCE: Infections with respiratory syncytial virus (RSV) are common and can cause severe lung disease in infants and the elderly. The lack of a suitable animal model with disease features similar to those in humans has hampered efforts to predict the efficacy of novel anti-RSV therapies and vaccines for use in humans. A murine model consisting of mice with a human immune system (HIS mice) could be useful for assessment of RSV disease and anti-RSV responses specific to humans. This study investigates an HIS mouse model to imitate human RSV disease and immune responses. We found that RSV lung infection in HIS mice results in an RSV-specific pathology that mimics RSV disease in humans and induces human anti-RSV immune responses. This model could be useful for better understanding of human RSV disease and for the development of RSV therapies.


Subject(s)
Lung Diseases/immunology , Lung/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/physiopathology , Respiratory Syncytial Viruses/immunology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/physiopathology , Animals , Antibodies, Neutralizing/analysis , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/analysis , Antibodies, Viral/blood , Antibodies, Viral/immunology , Bronchoalveolar Lavage Fluid/immunology , Chemokine CCL3/genetics , Disease Models, Animal , Humans , Immunoglobulin A/analysis , Immunoglobulin A/immunology , Interferon-gamma/metabolism , Interleukin-1beta/genetics , Lung/pathology , Lung Diseases/pathology , Lung Diseases/physiopathology , Lung Diseases/virology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/pathogenicity
7.
J Virol ; 90(15): 6918-6935, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27307565

ABSTRACT

UNLABELLED: Cleavage and polyadenylation specificity factor subunit 6 (CPSF6), a host factor that interacts with the HIV-1 capsid (CA) protein, is implicated in diverse functions during the early part of the HIV-1 life cycle, including uncoating, nuclear entry, and integration targeting. Preservation of CA binding to CPSF6 in vivo suggests that this interaction is fine-tuned for efficient HIV-1 replication in physiologically relevant settings. Nevertheless, this possibility has not been formally examined. To assess the requirement for optimal CPSF6-CA binding during infection of primary cells and in vivo, we utilized a novel CA mutation, A77V, that significantly reduced CA binding to CPSF6. The A77V mutation rendered HIV-1 largely independent from TNPO3, NUP358, and NUP153 for infection and altered the integration site preference of HIV-1 without any discernible effects during the late steps of the virus life cycle. Surprisingly, the A77V mutant virus maintained the ability to replicate in monocyte-derived macrophages, primary CD4(+) T cells, and humanized mice at a level comparable to that for the wild-type (WT) virus. Nonetheless, revertant viruses that restored the WT CA sequence and hence CA binding to CPSF6 emerged in three out of four A77V-infected animals. These results suggest that the optimal interaction of CA with CPSF6, though not absolutely essential for HIV-1 replication in physiologically relevant settings, confers a significant fitness advantage to the virus and thus is strictly conserved among naturally circulating HIV-1 strains. IMPORTANCE: CPSF6 interacts with the HIV-1 capsid (CA) protein and has been implicated in nuclear entry and integration targeting. Preservation of CPSF6-CA binding across various HIV-1 strains suggested that the optimal interaction between CA and CPSF6 is critical during HIV-1 replication in vivo Here, we identified a novel HIV-1 capsid mutant that reduces binding to CPSF6, is largely independent from the known cofactors for nuclear entry, and alters integration site preference. Despite these changes, virus carrying this mutation replicated in humanized mice at levels indistinguishable from those of the wild-type virus. However, in the majority of the animals, the mutant virus reverted back to the wild-type sequence, hence restoring the wild-type level of CA-CPSF6 interactions. These results suggest that optimal binding of CA to CPSF6 is not absolutely essential for HIV-1 replication in vivo but provides a fitness advantage that leads to the widespread usage of CPSF6 by HIV-1 in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Capsid Proteins/metabolism , HIV Infections/virology , HIV-1/physiology , Macrophages/virology , Virus Replication , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , HEK293 Cells , HIV Infections/metabolism , HeLa Cells , Humans , Macrophages/metabolism , Mice , Mice, Inbred NOD
8.
Org Biomol Chem ; 15(10): 2217-2225, 2017 Mar 08.
Article in English | MEDLINE | ID: mdl-28221388

ABSTRACT

α-GalCer analogues that combine known Th1 polarizing C6''-modifications with a C-glycosidic linkage were synthesized. We employed a protecting group strategy that allowed the preparation of both saturated and unsaturated derivatives with variable C6''-substituents. Selected analogues demonstrate promising activity in mice. Interestingly, the introduction of a 6''-O-pyridinylcarbamoyl substituent to α-C-GalCer restores its antigenicity in human iNKT cells.


Subject(s)
Galactosylceramides/chemical synthesis , Galactosylceramides/pharmacology , Natural Killer T-Cells/drug effects , Animals , Cell Line , Cytokines/biosynthesis , Cytokines/blood , Dose-Response Relationship, Drug , Galactosylceramides/chemistry , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Molecular Structure , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Structure-Activity Relationship
9.
J Immunol ; 195(6): 2710-21, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26254338

ABSTRACT

A CD1d-binding glycolipid, α-Galactosylceramide (αGalCer), activates invariant NK T cells and acts as an adjuvant. We previously identified a fluorinated phenyl ring-modified αGalCer analog, 7DW8-5, displaying nearly 100-fold stronger CD1d binding affinity. In the current study, 7DW8-5 was found to exert a more potent adjuvant effect than αGalCer for a vaccine based on radiation-attenuated sporozoites of a rodent malaria parasite, Plasmodium yoelii, also referred to as irradiated P. yoelii sporozoites (IrPySpz). 7DW8-5 had a superb adjuvant effect only when the glycolipid and IrPySpz were conjointly administered i.m. Therefore, we evaluated the effect of distinctly different biodistribution patterns of αGalCer and 7DW8-5 on their respective adjuvant activities. Although both glycolipids induce a similar cytokine response in sera of mice injected i.v., after i.m. injection, αGalCer induces a systemic cytokine response, whereas 7DW8-5 is locally trapped by CD1d expressed by dendritic cells (DCs) in draining lymph nodes (dLNs). Moreover, the i.m. coadministration of 7DW8-5 with IrPySpz results in the recruitment of DCs to dLNs and the activation and maturation of DCs. These events cause the potent adjuvant effect of 7DW8-5, resulting in the enhancement of the CD8(+) T cell response induced by IrPySpz and, ultimately, improved protection against malaria. Our study is the first to show that the colocalization of a CD1d-binding invariant NK T cell-stimulatory glycolipid and a vaccine, like radiation-attenuated sporozoites, in dLN-resident DCs upon i.m. conjoint administration governs the potency of the adjuvant effect of the glycolipid.


Subject(s)
Antigens, CD1d/immunology , Galactosylceramides/pharmacology , Malaria Vaccines/immunology , Malaria/immunology , Adjuvants, Immunologic/pharmacology , Animals , CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Female , Galactosylceramides/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Malaria/parasitology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Natural Killer T-Cells/immunology , Plasmodium yoelii/immunology , Protein Binding/immunology , Sporozoites/immunology
10.
Semin Immunol ; 25(2): 146-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23757291

ABSTRACT

Carbohydrates as T cell-activating antigens have been generating significant interest. For many years, carbohydrates were thought of as T-independent antigens, however, more recent research had demonstrated that mono- or oligosaccharides glycosidically linked to peptides can be recognized by T cells. T cell recognition of these glycopeptides depends on the structure of both peptide and glycan portions of the antigen. Subsequently, it was discovered that natural killer T cells recognized glycolipids when presented by the antigen presenting molecule CD1d. A transformative insight into glycan-recognition by T cells occurred when zwitterionic polysaccharides were discovered to bind to and be presented by MHCII to CD4+ T cells. Based on this latter observation, the role that carbohydrate epitopes generated from glycoconjugate vaccines had in activating helper T cells was explored and it was found that these epitopes are presented to specific carbohydrate recognizing T cells through a unique mechanism. Here we review the key interactions between carbohydrate antigens and the adaptive immune system at the molecular, cellular and systems levels exploring the significant biological implications in health and disease.


Subject(s)
Carbohydrates/immunology , T-Lymphocytes/immunology , Vaccines, Conjugate , Adaptive Immunity , Animals , Antigen Presentation , Antigens, CD1d/metabolism , Humans , Lymphocyte Activation
11.
Proc Natl Acad Sci U S A ; 111(34): 12474-9, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25118276

ABSTRACT

Immunological memory has been regarded as a unique feature of the adaptive immune response mediated in an antigen-specific manner by T and B lymphocytes. However, natural killer (NK) cells and γδT cells, which traditionally are classified as innate immune cells, have been shown in recent studies to have hallmark features of memory cells. Invariant NKT cell (iNKT cell)-mediated antitumor effects indicate that iNKT cells are activated in vivo by vaccination with iNKT cell ligand-loaded CD1d(+) cells, but not by vaccination with unbound NKT cell ligand. In such models, it previously was thought that the numbers of IFN-γ-producing cells in the spleen returned to the basal level around 1 wk after the vaccination. In the current study, we demonstrate the surprising presence of effector memory-like iNKT cells in the lung. We found long-term antitumor activity in the lungs of mice was enhanced after vaccination with iNKT cell ligand-loaded dendritic cells. Further analyses showed that the KLRG1(+) (Killer cell lectin-like receptor subfamily G, member 1-positive) iNKT cells coexpressing CD49d and granzyme A persisted for several months and displayed a potent secondary response to cognate antigen. Finally, analyses of CDR3ß by RNA deep sequencing demonstrated that some particular KLRG1(+) iNKT-cell clones accumulated, suggesting the selection of certain T-cell receptor repertoires by an antigen. The current findings identifying effector memory-like KLRG1(+) iNKT cells in the lung could result in a paradigm shift regarding the basis of newly developed extrathymic iNKT cells and could contribute to the future development of antitumor immunotherapy by uniquely energizing iNKT cells.


Subject(s)
Natural Killer T-Cells/immunology , Receptors, Immunologic/metabolism , Animals , Cell Survival/immunology , Complementarity Determining Regions/genetics , Dendritic Cells/immunology , Galactosylceramides/administration & dosage , Galactosylceramides/immunology , Granzymes/metabolism , Immunologic Memory , Integrin alpha4/metabolism , Interferon-gamma/biosynthesis , Lectins, C-Type , Lung/cytology , Lung/immunology , Lymphocyte Activation , Mice , Natural Killer T-Cells/classification , Natural Killer T-Cells/cytology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism
12.
J Biol Chem ; 290(28): 17206-17, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26018083

ABSTRACT

The ability of different glycosphingolipids (GSLs) to activate type I natural killer T cells (NKT cells) has been known for 2 decades. The possible therapeutic use of these GSLs has been studied in many ways; however, studies are needed in which the efficacy of promising GSLs is compared under identical conditions. Here, we compare five unique GSLs structurally derived from α-galactosylceramide. We employed biophysical and biological assays, as well as x-ray crystallography to study the impact of the chemical modifications of the antigen on type I NKT cell activation. Although all glycolipids are bound by the T cell receptor of type I NKT cells in real time binding assays with high affinity, only a few activate type I NKT cells in in vivo or in vitro experiments. The differences in biological responses are likely a result of different pharmacokinetic properties of each lipid, which carry modifications at different parts of the molecule. Our results indicate a need to perform a variety of assays to ascertain the therapeutic potential of type I NKT cell GSL activators.


Subject(s)
Galactosylceramides/chemistry , Galactosylceramides/immunology , Natural Killer T-Cells/immunology , Animals , Antigen Presentation , Antigens, CD1d/chemistry , Antigens, CD1d/metabolism , Binding Sites , Cell Line , Crystallography, X-Ray , Galactosylceramides/metabolism , Glycosphingolipids/chemistry , Glycosphingolipids/immunology , Glycosphingolipids/metabolism , Humans , Kinetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Natural Killer T-Cells/classification , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Surface Plasmon Resonance
13.
Clin Immunol ; 168: 6-15, 2016 07.
Article in English | MEDLINE | ID: mdl-27132023

ABSTRACT

In the present study, the combined adjuvant effect of 7DW8-5, a potent α-GalCer-analog, and monophosphoryl lipid A (MPLA), a TLR4 agonist, on the induction of vaccine-induced CD8(+) T-cell responses and protective immunity was evaluated. Mice were immunized with peptides corresponding to the CD8(+) T-cell epitopes of a malaria antigen, a circumsporozoite protein of Plasmodium yoelii, and a tumor antigen, a Wilms Tumor antigen-1 (WT-1), together with 7DW8-5 and MPLA, as an adjuvant. These immunization regimens were able to induce higher levels of CD8(+) T-cell responses and, ultimately, enhanced levels of protection against malaria and tumor challenges compared to the levels induced by immunization with peptides mixed with 7DW8-5 or MPLA alone. Co-administration of 7DW8-5 and MPLA induces activation of memory-like effector natural killer T (NKT) cells, i.e. CD44(+)CD62L(-)NKT cells. Our study indicates that 7DW8-5 greatly enhances important synergistic pathways associated to memory immune responses when co-administered with MPLA, thus rendering this combination of adjuvants a novel vaccine adjuvant formulation.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Galactosylceramides/pharmacology , Killer Cells, Natural/drug effects , Lipid A/analogs & derivatives , Toll-Like Receptor 4/agonists , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Drug Synergism , Epitopes, T-Lymphocyte/immunology , Galactosylceramides/administration & dosage , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Immunization/methods , Immunologic Memory/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lipid A/administration & dosage , Lipid A/pharmacology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Immunological , Peptides/immunology , Plasmodium yoelii/immunology , Plasmodium yoelii/physiology , Protective Agents/administration & dosage , Protective Agents/pharmacology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Toll-Like Receptor 4/metabolism , WT1 Proteins/genetics , WT1 Proteins/immunology
14.
Malar J ; 15: 201, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27068454

ABSTRACT

BACKGROUND: Plasmodium circumsporozoite protein (CSP) is a major surface antigen present in the sporozoite (Spz) stage of a malaria parasite. RTS, S vaccine, the most clinically advanced malaria vaccine, consists of a large portion of Plasmodium falciparum CSP (PfCSP). A highly infectious, recombinant rodent malaria, Plasmodium yoelii parasite bearing a full-length PfCSP, PfCSP/Py Spz, was needed as a tool to evaluate the role of PfCSP in mediating, protective, anti-malaria immunity in a mouse model. METHODS: A transgenic parasite, PfCSP/Py Spz, was generated by inserting a construct expressing the PfCSP at the locus of the P. yoelii CSP gene by double cross-over homologous recombination. Then the biological and protective properties of PfCSP/Py Spz were determined. RESULTS: This PfCSP/Py parasite produced up to 30,000 Spz in mosquito salivary glands, which is equal or even higher than the number of Spz produced by wild-type P. yoelii parasites. Five bites of PfCSP/Py-infected mosquitoes could induce blood infection in BALB/c mice. CONCLUSIONS: The current study has demonstrated a successful establishment of a transgenic P. yoelii parasite clone that is able to express a full-length PfCSP, PfCSP/Py parasite. Importantly, this PfCSP/Py parasite can be as infectious as the wild-type P. yoelii parasite both in mosquito vector and in mouse, a mammalian host. A new transgenic parasite that expresses a full-length PfCSP may become a useful tool for researchers to investigate immunity against PfCSP in a mouse model.


Subject(s)
Culicidae/parasitology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/blood , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/immunology , Plasmodium falciparum/genetics , Salivary Glands/parasitology , T-Lymphocytes/parasitology , Vaccines, Synthetic/immunology
16.
Infect Immun ; 82(2): 808-17, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478094

ABSTRACT

Evidence from clinical trials of malaria vaccine candidates suggests that both cell-mediated and humoral immunity to pre-erythrocytic parasite stages can provide protection against infection. Novel pre-erythrocytic antibody (Ab) targets could be key to improving vaccine formulations, which are currently based on targeting antigens such as the circumsporozoite protein (CSP). However, methods to assess the effects of sporozoite-specific Abs on pre-erythrocytic infection in vivo remain underdeveloped. Here, we combined passive transfer of monoclonal Abs (MAbs) or immune serum with a luciferase-expressing Plasmodium yoelii sporozoite challenge to assess Ab-mediated inhibition of liver infection in mice. Passive transfer of a P. yoelii CSP MAb showed inhibition of liver infection when mice were challenged with sporozoites either intravenously or by infectious mosquito bite. However, inhibition was most potent for the mosquito bite challenge, leading to a more significant reduction of liver-stage burden and even a lack of progression to blood-stage parasitemia. This suggests that Abs provide effective protection against a natural infection. Inhibition of liver infection was also achieved by passive transfer of immune serum from whole-parasite-immunized mice. Furthermore, we demonstrated that passive transfer of a MAb against P. falciparum CSP inhibited liver-stage infection in a humanized mouse/P. falciparum challenge model. Together, these models constitute unique and sensitive in vivo methods to assess serum-transferable protection against Plasmodium sporozoite challenge.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Immune Sera/immunology , Malaria/prevention & control , Plasmodium falciparum/immunology , Plasmodium yoelii/immunology , Animals , Disease Models, Animal , Female , Humans , Immunization, Passive , Liver/immunology , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Mice , Mice, Inbred BALB C , Mice, SCID
17.
Planta Med ; 80(4): 283-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24549928

ABSTRACT

Juzen-taiho-to is an immunostimulatory herbal formulation that is clinically used in East Asia for cancer patients undergoing chemotherapy and radiation. The formulation stimulates various leukocytes, including T, B, and NK cells and macrophages. Although Juzen-taiho-to is known to contain numerous compounds with various pharmacological activities, it is not clear which compounds are responsible for the stimulation of individual cell types. Here, we conducted what we call "biomarker-guided screening" to purify compounds responsible for the macrophages stimulatory activity. To this end, gene expression was analyzed by a DNA array for macrophages treated with Juzen-taiho-to and DMSO (vehicle control), which identified intercellular adhesion molecule 1 as a biomarker of macrophage stimulation by Juzen-taiho-to. A quantitative reverse transcription polymerase chain reaction assay of intercellular adhesion molecule 1 was then used to guide the purification of active compounds. The screening resulted in the purification of a glycolipid mixture, containing ß-glucosylceramides. The glycolipid mixture potently stimulated intercellular adhesion molecule 1 expression in primary dendritic cells as well as in primary CD14+ (macrophages) cells. The identification of this glycolipid mixture opens up an opportunity for further studies to understand how plant-derived glycolipids stimulate macrophages and dendritic cells in a safe and effective manner as demonstrated by Juzen-taiho-to.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/drug effects , Drugs, Chinese Herbal/pharmacology , Glucosylceramides/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Macrophages/drug effects , Magnoliopsida/chemistry , Adjuvants, Immunologic/analysis , Antineoplastic Agents, Phytogenic/analysis , Antineoplastic Agents, Phytogenic/pharmacology , Dendritic Cells/metabolism , Drugs, Chinese Herbal/chemistry , Glucosylceramides/isolation & purification , Humans , Macrophages/metabolism
18.
Semin Immunol ; 22(2): 87-96, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19962909

ABSTRACT

NKT cells are known to play a role against certain microbial infections, including malaria and HIV, two major global infectious diseases. NKT cells exhibit either protective or pathogenic role against malaria. They are depleted by HIV infection and have a direct pathogenic role against many opportunistic infections common in end-stage AIDS. This review discusses the various features of the interaction between NKT cells and malaria parasites and HIV, and the potential to harness this interaction for therapeutic and vaccine strategies.


Subject(s)
Glycolipids/immunology , HIV Infections/immunology , Malaria/immunology , Natural Killer T-Cells/immunology , Adaptive Immunity , Animals , Antigens, CD1d/immunology , Antiretroviral Therapy, Highly Active , Apoptosis , Glycolipids/therapeutic use , HIV Infections/drug therapy , Humans , Immunity, Innate , Malaria/drug therapy , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/microbiology , Natural Killer T-Cells/virology , Vaccines
19.
Mediators Inflamm ; 2014: 728939, 2014.
Article in English | MEDLINE | ID: mdl-25132735

ABSTRACT

Although the roles of CD8+ T cells and a major preerythrocytic antigen, the circumsporozoite (CS) protein, in contributing protective antimalaria immunity induced by radiation-attenuated sporozoites, have been shown by a number of studies, the extent to which these players contribute to antimalaria immunity is still unknown. To address this question, we have generated C57BL/6 (B6) transgenic (Tg) mice, expressing K(d) molecules under the MHC-I promoter, called MHC-I-K(d)-Tg mice. In this study, we first determined that a single immunizing dose of IrPySpz induced a significant level of antimalaria protective immunity in MHC-I-K(d)-Tg mice but not in B6 mice. Then, by depleting various T-cell subsets in vivo, we determined that CD8+ T cells are the main mediator of the protective immunity induced by IrPySpz. Furthermore, when we immunized (MHC-I-K(d)-Tg × CS-Tg) F1 mice with IrPySpz after crossing MHC-I-K(d)-Tg mice with PyCS-transgenic mice (CS-Tg), which are unable to mount PyCS-specific immunity, we found that IrPySpz immunization failed to induce protective antimalaria immunity in (MHC-I-K(d)-Tg × CS-Tg) F1 mice, thus indicating the absence of PyCS antigen-dependent immunity in these mice. These results indicate that protective antimalaria immunity induced by IrPySpz in MHC-I-K(d)-Tg mice is mediated by CS protein-specific, K(d)-restricted CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/metabolism , Sporozoites/immunology , Sporozoites/metabolism , Animals , Anopheles , Antimalarials/immunology , Female , Histocompatibility Antigens Class I/genetics , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Plasmodium yoelii/immunology , Plasmodium yoelii/metabolism
20.
Vaccines (Basel) ; 12(3)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38543973

ABSTRACT

The COVID-19 outbreak was a global pandemic with wide-ranging healthcare implications. Although several mRNA-based vaccines delivered using lipid nanoparticles (LNP) have been approved and demonstrated efficacy at reducing the severity and spread of infection, continued rapid viral evolution and disadvantages currently associated with LNP delivery vehicles (such as toxicity) are driving the design of next-generation SARS-CoV-2 vaccines. Herein, we describe the development of a trimethylated chitosan-based nanoparticle layer-by-layer (LbL) delivery platform for multiple antigens as a scalable and safe COVID-19 vaccine, known as, "LbL-CoV19". These vaccine candidates have been demonstrated to be biocompatible, safe, and effective at stimulating both humoral and cellular responses for protection in preclinical studies. Preliminary results also indicate that LbL-CoV19 can potentially achieve rapid, long-lasting, and broad protection against the SARS-CoV-2 challenge. The "plug-and-play" platform technology is well suited to preparedness for future pandemics and disease outbreaks.

SELECTION OF CITATIONS
SEARCH DETAIL