Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Immunity ; 38(4): 818-30, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23562160

ABSTRACT

In comparison to murine dendritic cells (DCs), less is known about the function of human DCs in tissues. Here, we analyzed, by using lung tissues from humans and humanized mice, the role of human CD1c(+) and CD141(+) DCs in determining the type of CD8(+) T cell immunity generated to live-attenuated influenza virus (LAIV) vaccine. We found that both lung DC subsets acquired influenza antigens in vivo and expanded specific cytotoxic CD8(+) T cells in vitro. However, lung-tissue-resident CD1c(+) DCs, but not CD141(+) DCs, were able to drive CD103 expression on CD8(+) T cells and promoted CD8(+) T cell accumulation in lung epithelia in vitro and in vivo. CD1c(+) DCs induction of CD103 expression was dependent on membrane-bound cytokine TGF-ß1. Thus, CD1c(+) and CD141(+) DCs generate CD8(+) T cells with different properties, and CD1c(+) DCs specialize in the regulation of mucosal CD8(+) T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lung/immunology , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/metabolism , Antigens, CD1/metabolism , Antigens, Viral/immunology , Cell Differentiation , Cells, Cultured , Cytotoxicity, Immunologic , Glycoproteins/metabolism , Humans , Immunity, Mucosal , Immunologic Memory , Influenza Vaccines/immunology , Integrin alpha Chains/metabolism , Lung/virology , Lymphocyte Activation , Mice , Mice, SCID , Microarray Analysis
2.
J Inherit Metab Dis ; 39(2): 293-303, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26310963

ABSTRACT

Enzyme replacement therapy (ERT) is an effective treatment for several lysosomal storage disorders (LSDs). Intravenously infused enzymes are taken up by tissues through either the mannose 6-phosphate receptor (M6PR) or the mannose receptor (MR). It is generally believed that M6PR-mediated endocytosis is a key mechanism for ERT in treating LSDs that affect the non-macrophage cells of visceral organs. However, the therapeutic efficacy of MR-mediated delivery of mannose-terminated enzymes in these diseases has not been fully evaluated. We tested the effectiveness of a non-phosphorylated α-galactosidase A produced from moss (referred to as moss-aGal) in vitro and in a mouse model of Fabry disease. Endocytosis of moss-aGal was MR-dependent. Compared to agalsidase alfa, a phosphorylated form of α-galactosidase A, moss-aGal was more preferentially targeted to the kidney. Cellular localization of moss-aGal and agalsidase alfa in the heart and kidney was essentially identical. A single injection of moss-aGal led to clearance of accumulated substrate in the heart and kidney to an extent comparable to that achieved by agalsidase alfa. This study suggested that mannose-terminated enzymes may be sufficiently effective for some LSDs in which non-macrophage cells are affected, and that M6P residues may not always be a prerequisite for ERT as previously considered.


Subject(s)
Fabry Disease/enzymology , Fabry Disease/metabolism , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Mannosephosphates/metabolism , Receptors, Cell Surface/metabolism , alpha-Galactosidase/metabolism , Animals , Cell Line , Disease Models, Animal , Enzyme Replacement Therapy/methods , Female , Humans , Isoenzymes/metabolism , Kidney/metabolism , Lysosomal Storage Diseases/enzymology , Lysosomal Storage Diseases/metabolism , Male , Mannose Receptor , Mice , Mice, Inbred C57BL , Receptor, IGF Type 2/metabolism , Recombinant Proteins
3.
J Immunol ; 193(9): 4335-43, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25246496

ABSTRACT

Dendritic cells (DCs) play the central role in the priming of naive T cells and the differentiation of unique effector T cells. In this study, using lung tissues and blood from both humans and humanized mice, we analyzed the response of human CD1c(+) and CD141(+) DC subsets to live-attenuated influenza virus. Specifically, we analyzed the type of CD4(+) T cell immunity elicited by live-attenuated influenza virus-exposed DCs. Both DC subsets induce proliferation of allogeneic naive CD4(+) T cells with the capacity to secrete IFN-γ. However, CD141(+) DCs are uniquely able to induce the differentiation of IL-4- and IL-13-producing CD4(+) T cells. CD141(+) DCs induce IL-4- and IL-13-secreting CD4(+) T cells through OX40 ligand. Thus, CD141(+) DCs demonstrate remarkable plasticity in guiding adaptive immune responses.


Subject(s)
Antigens, Surface/metabolism , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Animals , Antigens, CD1/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/metabolism , Cell Differentiation , Cells, Cultured , Dendritic Cells/metabolism , Glycoproteins/metabolism , Humans , Immunophenotyping , Lung/immunology , Lung/metabolism , Lung/virology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , OX40 Ligand/metabolism , Orthomyxoviridae/immunology , Phenotype , Signal Transduction , Th2 Cells/immunology , Th2 Cells/metabolism , Thrombomodulin
4.
iScience ; 27(3): 109238, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38433905

ABSTRACT

Pre-clinical use of humanized mice transplanted with CD34+ hematopoietic stem and progenitor cells (HSPCs) is limited by insufficient engraftment with adult non-mobilized HSPCs. Here, we developed a novel immunodeficient mice based on NOD-SCID-Il2γc-/- (NSG) mice to support long-term engraftment with human adult HSPCs. As both Flt3L and IL-6 are critical for many aspects of hematopoiesis, we knock-out mouse Flt3 and knock-in human IL6 gene. The resulting mice showed an increase in the availability of mouse Flt3L to human cells and a dose-dependent production of human IL-6 upon activation. Upon transplantation with low number of human HSPCs from adult bone marrow, these humanized mice demonstrated a significantly higher engraftment with multilineage differentiation of human lymphoid and myeloid cells, and tissue colonization at one year after adult HSPC transplant. Thus, these mice enable studies of human hematopoiesis and tissue colonization over time and may facilitate building autologous models for immuno-oncology studies.

5.
STAR Protoc ; 5(3): 103155, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38935509

ABSTRACT

Humanized mice, defined as mice with human immune systems, have become an emerging model to study human hematopoiesis, infectious disease, and cancer. Here, we describe the techniques to generate humanized NSGF6 mice using adult human CD34+ hematopoietic stem and progenitor cells (HSPCs). We describe steps for constructing and monitoring the engraftment of humanized mice. We then detail procedures for tissue processing and immunophenotyping by flow cytometry to evaluate the multilineage hematopoietic differentiation. For complete details on the use and execution of this protocol, please refer to Yu et al.1.

6.
Trends Cancer ; 9(7): 578-590, 2023 07.
Article in English | MEDLINE | ID: mdl-37087398

ABSTRACT

Realizing the clinical promise of cancer immunotherapy is hindered by gaps in our knowledge of in vivo mechanisms underlying treatment response as well as treatment limiting toxicity. Preclinical in vivo model systems and technologies are required to address these knowledge gaps and to surmount the challenges faced in the clinical application of immunotherapy. Mice are commonly used for basic and translational research to support development and testing of immune interventions, including for cancer. Here, we discuss the advantages and the limitations of current models as well as future developments.


Subject(s)
Neoplasms , Animals , Mice , Neoplasms/drug therapy , Medical Oncology , Disease Models, Animal , Translational Research, Biomedical , Immunotherapy
7.
bioRxiv ; 2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37873457

ABSTRACT

Pre-clinical use of humanized mice transplanted with CD34 + hematopoietic progenitor cells (HPCs) is limited by insufficient engraftment with adult HPCs. Here, we developed a novel immunodeficient mice based in NOD-SCID- Il2γc -/- (NSG) mice to support long-term engraftment with human adult HPCs and tissue colonization with human myeloid cells. As both Flt3L and IL-6 are critical for many aspects of hematopoiesis, we knock-out mouse Flt3 and knock-in human IL6 gene. The resulting mice showed an increase in the availability of mouse Flt3L to human cells, and a dose-dependent production of human IL-6 upon activation. Upon transplantation with low number of human HPCs from adult bone marrow, these humanized mice demonstrated a significantly higher engraftment with multilineage differentiation of human lymphoid and myeloid cells. Furthermore, higher frequencies of human lymphoid and myeloid cells were detected in tissues at one year after adult HPC transplant. Thus, these mice enable studies of human hematopoiesis and tissue colonization over time. Summary: Pre-clinical use of humanized mice is limited by insufficient engraftment with adult hematopoietic progenitor cells (HPCs). Here, we developed a novel immunodeficient mice which support long-term engraftment with adult bone marrow HPCs and facilitate building autologous models for immuno-oncology studies.

8.
J Immunol ; 182(5): 2766-76, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234171

ABSTRACT

During viral infection, dendritic cells (DCs) capture infected cells and present viral Ags to CD8(+) T cells. However, activated DCs might potentially present cell-associated Ags derived from captured dead cells. In this study, we find that human DCs that captured dead cells containing the TLR3 agonist poly(I:C) produced cytokines and underwent maturation, but failed to elicit autologous CD8(+) T cell responses against Ags of dead cells. Accordingly, DCs that captured dead cells containing poly(I:C), or influenza virus, are unable to activate CD8(+) T cell clones specific to cell-associated Ags of captured dead cells. CD4(+) T cells are expanded with DCs that have captured poly(I:C)-containing dead cells, indicating the inhibition is specific for MHC class I-restricted cross-presentation. Furthermore, these DCs can expand naive allogeneic CD8(+) T cells. Finally, soluble or targeted Ag is presented when coloaded onto DCs that have captured poly(I:C)-containing dead cells, indicating the inhibition is specific for dead cell cargo that is accompanied by viral or poly(I:C) stimulus. Thus, DCs have a mechanism that prevents MHC class I-restricted cross-presentation of cell-associated Ag when they have captured dead infected cells.


Subject(s)
Cross-Priming/immunology , Dendritic Cells/virology , Growth Inhibitors/immunology , HLA-A2 Antigen/immunology , Immunosuppression Therapy , Influenza A virus/immunology , Melanoma/virology , Poly I-C/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/pathology , HLA-A2 Antigen/metabolism , Humans , Immunosuppression Therapy/methods , Lymphocyte Activation/immunology , Melanoma/immunology , Melanoma/pathology , Necrosis , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology
9.
J Immunol ; 182(11): 6815-23, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19454677

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are key regulators of antiviral immunity. They rapidly secrete IFN-alpha and cross-present viral Ags, thereby launching adaptive immunity. In this study, we show that activated human pDCs inhibit replication of cancer cells and kill them in a contact-dependent fashion. Expression of CD2 distinguishes two pDC subsets with distinct phenotype and function. Both subsets secrete IFN-alpha and express granzyme B and TRAIL. CD2(high) pDCs uniquely express lysozyme and can be found in tonsils and in tumors. Both subsets launch recall T cell responses. However, CD2(high) pDCs secrete higher levels of IL12p40, express higher levels of costimulatory molecule CD80, and are more efficient in triggering proliferation of naive allogeneic T cells. Thus, human blood pDCs are composed of subsets with specific phenotype and functions.


Subject(s)
CD2 Antigens , Dendritic Cells/cytology , B7-1 Antigen/analysis , Cell Proliferation , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Humans , Interleukin-12 Subunit p40/analysis , Neoplasms/immunology , Phenotype , T-Lymphocytes/cytology , T-Lymphocytes/immunology
10.
J Exp Med ; 218(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33857287

ABSTRACT

Metastasis of melanoma significantly worsens prognosis; thus, therapeutic interventions that prevent metastasis could improve patient outcomes. Here, we show using humanized mice that colonization of distant visceral organs with melanoma is dependent upon a human CD33+CD11b+CD117+ progenitor cell subset comprising <4% of the human CD45+ leukocytes. Metastatic tumor-infiltrating CD33+ cells from patients and humanized (h)NSG-SGM3 mice showed converging transcriptional profiles. Single-cell RNA-seq analysis identified a gene signature of a KIT/CD117-expressing CD33+ subset that correlated with decreased overall survival in a TCGA melanoma cohort. Thus, human CD33+CD11b+CD117+ myeloid cells represent a novel candidate biomarker as well as a therapeutic target for metastatic melanoma.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Proto-Oncogene Proteins c-kit/metabolism , Animals , Biomarkers/metabolism , CD11b Antigen/metabolism , Cell Line, Tumor , Cohort Studies , Humans , Leukocyte Common Antigens/metabolism , Leukocytes/metabolism , Leukocytes/pathology , Mice , Mice, Inbred NOD , Prognosis
11.
Blood ; 112(9): 3671-8, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18713944

ABSTRACT

The development of novel human vaccines would be greatly facilitated by the development of in vivo models that permit preclinical analysis of human immune responses. Here, we show that nonobese diabetic severe combined immunodeficiency (NOD/SCID) beta(2) microglobulin(-/-) mice, engrafted with human CD34+ hematopoietic progenitors and further reconstituted with T cells, can mount specific immune responses against influenza virus vaccines. Live attenuated trivalent influenza virus vaccine induces expansion of CD8+ T cells specific to influenza matrix protein (FluM1) and nonstructural protein 1 in blood, spleen, and lungs. On ex vivo exposure to influenza antigens, antigen-specific CD8+ T cells produce IFN-gamma and express cell-surface CD107a. FluM1-specific CD8+ T cells can be also expanded in mice vaccinated with inactivated trivalent influenza virus vaccine. Expansion of antigen-specific CD8+ T cells is dependent on reconstitution of the human myeloid compartment. Thus, this humanized mouse model permits preclinical testing of vaccines designed to induce cellular immunity, including those against influenza virus. Furthermore, this work sets the stage for systematic analysis of the in vivo functions of human DCs. This, in turn, will allow a new approach to the rational design and preclinical testing of vaccines that cannot be tested in human volunteers.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Influenza Vaccines/pharmacology , Adoptive Transfer , Animals , Dendritic Cells/immunology , Hematopoietic Stem Cell Transplantation , Humans , Immunity, Cellular , Influenza Vaccines/immunology , Lymphocyte Transfusion , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , T-Lymphocytes/transplantation , Tetanus Toxoid/immunology , Tetanus Toxoid/pharmacology , Transplantation, Heterologous , Viral Matrix Proteins/immunology , Viral Nonstructural Proteins/immunology , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
12.
J Biomed Biotechnol ; 2010: 548280, 2010.
Article in English | MEDLINE | ID: mdl-20625493

ABSTRACT

Chronic hepatitis C virus (HCV) infection remains a serious burden to public health worldwide. Currently, HCV-infected patients could undergo antiviral therapy by giving pegylated IFN-alpha with ribavirin. However, this therapy is only effective in around 50% of patients with HCV genotype 1, which accounts for more than 70% of all HCV infection, and it is not well tolerated for most patients. Moreover, there is no vaccine available. The efforts on identifying protective immunity against HCV have progressed recently. Neutralizing antibodies and robust T cell responses including both CD4(+) and CD8(+) have been shown to be related to the clearance of HCV, which have shed lights on the potential success of HCV vaccines. There are many vaccines developed and tested before entering clinical trials. Here, we would first discuss strategies of viral immune evasion and correlates of protective host immunity and finally review some prospective vaccine approaches against chronic HCV infection.


Subject(s)
Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/immunology , Viral Hepatitis Vaccines/immunology , Viral Hepatitis Vaccines/therapeutic use , Hepacivirus/immunology , Humans , Immune Evasion/immunology , Immunity/immunology
13.
Methods Enzymol ; 636: 351-368, 2020.
Article in English | MEDLINE | ID: mdl-32178826

ABSTRACT

Mouse models of human cancer have been used extensively to circumvent the complexity in human patients. However, murine models often inadequately recapitulate the human cancer-immune interface partly due to important differences between mouse and human immune systems. Immunodeficient mice when transplanted with CD34+ hematopoietic progenitor cells (HPCs) develop multilineage human immune cells. While there remain limitations, efforts have been made to improve the function of human immune system. Thus, humanized mice, defined as mice with human immune system, have become an emerging model to study human cancers. Humanized mouse models have been used for various areas of cancer research including adoptive transfer of chimeric antigen receptor (CAR)-modified T cells, neoantigen vaccination to increase T cell repertoire and reprograming tumor microenvironment. Here, we describe the essential techniques to generate humanized mouse models for immuno-oncology studies.


Subject(s)
Neoplasms , Animals , Disease Models, Animal , Humans , Mice , Neoplasms/therapy , T-Lymphocytes , Tumor Microenvironment , Xenograft Model Antitumor Assays
14.
Sci Immunol ; 2(13)2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28783704

ABSTRACT

Dendritic cells (DCs) are critical for the launching of protective T cell immunity in response to viral infection. Viruses can directly infect DCs, thereby compromising their viability and suppressing their ability to activate immune responses. How DC function is maintained in light of this paradox is not understood. By analyzing the susceptibility of primary human DC subsets to viral infections, we report that CD141+ DCs have an innate resistance to infection by a broad range of enveloped viruses, including HIV and influenza virus. In contrast, CD1c+ DCs are susceptible to infection, which enables viral antigen production but impairs their immune functions and survival. The ability of CD141+ DCs to resist infection is conferred by RAB15, a vesicle-trafficking protein constitutively expressed in this DC subset. We show that CD141+ DCs rely on viral antigens produced in bystander cells to launch cross-presentation-driven T cell responses. By dissociating viral infection from antigen presentation, this mechanism protects the functional capacity of DCs to launch adaptive immunity against viral infection.

15.
Vaccine ; 34(41): 4857-4865, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27595442

ABSTRACT

The targeting of vaccine antigens to antigen presenting cells (APC), such as dendritic cells (DCs), is a promising strategy for boosting vaccine immunogenicity and, in turn, protective and/or therapeutic efficacy. However, in vivo systems are needed to evaluate the potential of this approach for testing human vaccines. To this end, we examined human CD8(+) T-cell expansion to novel DC-targeting vaccines in vitro and in vivo in humanized mice. Vaccines incorporating the influenza matrix protein-1 (FluM1) antigen fused to human specific antibodies targeting different DC receptors, including DEC-205, DCIR, Dectin-1, and CD40, elicited human CD8(+) T-cell responses, as defined by the magnitude of specific CD8(+) T-cells to the targeted antigen. In vitro we observed differences in response to the different vaccines, particularly between the weakly immunogenic DEC-205-targeted and more strongly immunogenic CD40-targeted vaccines, consistent with previous studies. However, in humanized mice adoptively transferred (AT) with mature human T cells (HM-T), vaccines that performed weakly in vitro (i.e., DEC-205, DCIR, and Dectin-1) gave stronger responses in vivo, some resembling those of the strongly immunogenic CD40-targeted vaccine. These results demonstrate the utility of the humanized mouse model as a platform for studies of human vaccines.


Subject(s)
Adoptive Transfer , Antibodies, Monoclonal/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Influenza Vaccines/immunology , Viral Matrix Proteins/immunology , Animals , Antigen Presentation , Antigens, CD/immunology , CD40 Antigens/immunology , Cross-Priming , Epitopes/immunology , Humans , Immunity, Cellular , Lectins, C-Type/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Minor Histocompatibility Antigens/immunology , Receptors, Cell Surface/immunology , Recombinant Fusion Proteins/immunology
16.
Cancer Immunol Res ; 2(5): 487-500, 2014 May.
Article in English | MEDLINE | ID: mdl-24795361

ABSTRACT

Our studies showed that tumor-infiltrating dendritic cells (DC) in breast cancer drive inflammatory Th2 (iTh2) cells and protumor inflammation. Here, we show that intratumoral delivery of the ß-glucan curdlan, a ligand of dectin-1, blocks the generation of iTh2 cells and prevents breast cancer progression in vivo. Curdlan reprograms tumor-infiltrating DCs via the ligation of dectin-1, enabling the DCs to become resistant to cancer-derived thymic stromal lymphopoietin (TSLP), to produce IL-12p70, and to favor the generation of Th1 cells. DCs activated via dectin-1, but not those activated with TLR-7/8 ligand or poly I:C, induce CD8+ T cells to express CD103 (αE integrin), a ligand for cancer cells, E-cadherin. Generation of these mucosal CD8+ T cells is regulated by DC-derived integrin αvß8 and TGF-ß activation in a dectin-1-dependent fashion. These CD103+ CD8+ mucosal T cells accumulate in the tumors, thereby increasing cancer necrosis and inhibiting cancer progression in vivo in a humanized mouse model of breast cancer. Importantly, CD103+ CD8+ mucosal T cells elicited by reprogrammed DCs can reject established cancer. Thus, reprogramming tumor-infiltrating DCs represents a new strategy for cancer rejection.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Mucous Membrane/immunology , Animals , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Lectins, C-Type/metabolism , Mice , Mucous Membrane/metabolism , Signal Transduction , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Transforming Growth Factor beta/metabolism , beta-Glucans/immunology , beta-Glucans/pharmacology
17.
Nat Commun ; 5: 5283, 2014 Oct 22.
Article in English | MEDLINE | ID: mdl-25335753

ABSTRACT

The mechanisms by which microbial vaccines interact with human APCs remain elusive. Herein, we describe the transcriptional programs induced in human DCs by pathogens, innate receptor ligands and vaccines. Exposure of DCs to influenza, Salmonella enterica and Staphylococcus aureus allows us to build a modular framework containing 204 transcript clusters. We use this framework to characterize the responses of human monocytes, monocyte-derived DCs and blood DC subsets to 13 vaccines. Different vaccines induce distinct transcriptional programs based on pathogen type, adjuvant formulation and APC targeted. Fluzone, Pneumovax and Gardasil, respectively, activate monocyte-derived DCs, monocytes and CD1c+ blood DCs, highlighting APC specialization in response to vaccines. Finally, the blood signatures from individuals vaccinated with Fluzone or infected with influenza reveal a signature of adaptive immunity activation following vaccination and symptomatic infections, but not asymptomatic infections. These data, offered with a web interface, may guide the development of improved vaccines.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/microbiology , Transcription, Genetic , Vaccines/chemistry , Algorithms , Animals , Antigens, CD1/metabolism , Antigens, Surface/metabolism , Cluster Analysis , Cytokines/metabolism , Dendritic Cells/metabolism , Dogs , Gene Expression Profiling , Glycoproteins/metabolism , Humans , Influenza A Virus, H1N1 Subtype , Interleukin-4/metabolism , Madin Darby Canine Kidney Cells , Monocytes/cytology , Monocytes/metabolism , Principal Component Analysis , Salmonella enterica , Staphylococcus aureus , Thrombomodulin , Transcriptome
18.
Vaccine ; 30(7): 1305-12, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22214888

ABSTRACT

Enterovirus type 71 (EV71) is a virulent form of enteroviruses causing hospitalizations for children less than three years of age. Currently there are no anti-viral therapies or vaccines available for EV71. Due to the high risk of poliomyelitis-like paralysis and fatal encephalitis, an effective vaccine to EV71 could potentially prevent virus-induced morbidity and mortality. In this study, we first tested a potential EV71 vaccine candidate based on virus-like particles (VLP). We vaccinated macaque monkeys to validate the immunogenicity of the VLP vaccine to EV71. We detected the VLP or EV71-specific antibodies, neutralization titers, ELISPOT, and T cell response to find their immune responses to EV71. When the VLP vaccine adjuvanted with alum was given to macaque monkeys, these monkeys developed both specific humoral and cellular immune responses to EV71. Despite lower neutralizing antibodies to EV71 were found in sera of VLP-immunized monkeys than monkeys vaccinated with inactivated EV71, VLP-based vaccine generated a memory immune response to EV71. Hence, VLP-based EV71 vaccine is a potential vaccine against EV71 infection.


Subject(s)
Antibodies, Neutralizing/immunology , Enterovirus A, Human/immunology , Enterovirus Infections/prevention & control , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Antibodies, Viral/immunology , Child, Preschool , Enterovirus Infections/immunology , Enterovirus Infections/virology , Humans , Immunity, Cellular , Immunity, Humoral , Immunologic Memory , Macaca , Vaccination , Vaccines, Inactivated , Vaccines, Virus-Like Particle/genetics , Viral Vaccines/genetics
19.
Expert Opin Drug Discov ; 2(7): 949-60, 2007 Jul.
Article in English | MEDLINE | ID: mdl-23484815

ABSTRACT

Mouse models of human disease form a link between genetics and biology. However, mice and humans differ in many aspects of immune system biology. These differences might explain, in part, why many successful preclinical immunotherapy studies in mice turn out to be unsuccessful when used in clinical trials in humans. Pioneering studies in the late 1980s demonstrated the reconstitution of human lympho-hematopoietic cells in immunodeficient mice. Since this time, immunodeficient mice are being tested as hosts for human hematopoietic organs or cells in an effort to create an in vivo model of the complete human immune system. Such Humouse models could permit us to generate and test novel human vaccines.

20.
Fish Shellfish Immunol ; 14(4): 317-31, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12657535

ABSTRACT

Pacific white shrimp (Litopenaeus vannamei) were injected with Taura syndrome virus (TSV) to assess shrimp immune responses and survival. TSV-infected shrimp suffered high mortality, but mock-infected and untreated shrimp experienced no mortality. Moribund shrimp were a pale, reddish colour and were lethargic and soft-shelled. Their haemolymph was clear red and coagulated poorly. In TSV-infected shrimp, the total haemocyte count (THC), hyalinocyte and granulocyte counts, and total plasma protein decreased significantly to 21%, 24%, 17% and 56% of untreated control values, respectively. Haemocyanin decreased to 67%, and clottable proteins to 80% of control values (P< 0.01). Copper and calcium ions, haemocytic transglutaminase (TGase) activity and plasma growth inhibitory activity against Vibrio harveyi also decreased significantly. Generation of intrahaemocytic superoxide anion, O(-2), in TSV-infected shrimp was significantly greater (P< 0.05) than in both control groups, no matter whether glucan stimulated or unstimulated. But the relative increase of intrahaemocytic O(-2) generation in TSV-infected shrimp response to glucan stimulation was lower in both controls. Plasma phenoloxidase (PO) activity increased significantly in TSV-infected shrimp. The plasma bacterial agglutinin titre against E. coli and V. harveyi, growth inhibition of E. coli and the concentration of magnesium ions in TSV-infected shrimp did not change significantly. In conclusion, ten of thirteen haemolymph parameters changed significantly during the host-TSV interaction. These parameters might be valuable references of shrimp health status.


Subject(s)
Hemolymph/immunology , Penaeidae/virology , Picornaviridae Infections/veterinary , Picornaviridae/growth & development , Agglutination Tests/veterinary , Animals , Aquaculture , Calcium/blood , Copper/blood , Escherichia coli/growth & development , Hemocyanins/immunology , Hemocyanins/metabolism , Hemocytes/immunology , Hemocytes/virology , Hemolymph/virology , Magnesium/blood , Monophenol Monooxygenase/immunology , Monophenol Monooxygenase/metabolism , Penaeidae/immunology , Picornaviridae/immunology , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Superoxides/blood , Transglutaminases/blood , Vibrio/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL