Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
2.
Front Immunol ; 12: 726421, 2021.
Article in English | MEDLINE | ID: mdl-34526998

ABSTRACT

CD8 T cell infiltration of the central nervous system (CNS) is necessary for host protection but contributes to neuropathology. Antigen presenting cells (APCs) situated at CNS borders are thought to mediate T cell entry into the parenchyma during neuroinflammation. The identity of the CNS-resident APC that presents antigen via major histocompatibility complex (MHC) class I to CD8 T cells is unknown. Herein, we characterize MHC class I expression in the naïve and virally infected brain and identify microglia and macrophages (CNS-myeloid cells) as APCs that upregulate H-2Kb and H-2Db upon infection. Conditional ablation of H-2Kb and H-2Db from CNS-myeloid cells allowed us to determine that antigen presentation via H-2Db, but not H-2Kb, was required for CNS immune infiltration during Theiler's murine encephalomyelitis virus (TMEV) infection and drives brain atrophy as a consequence of infection. These results demonstrate that CNS-myeloid cells are key APCs mediating CD8 T cell brain infiltration.


Subject(s)
Antigen-Presenting Cells/pathology , Brain Diseases/virology , Brain/pathology , H-2 Antigens/immunology , Theilovirus/immunology , Animals , Antigen Presentation , Antigen-Presenting Cells/virology , Atrophy , Brain/immunology , Brain/virology , Brain Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Macrophages/pathology , Macrophages/virology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Microglia/pathology , Microglia/virology
3.
Front Immunol ; 12: 667478, 2021.
Article in English | MEDLINE | ID: mdl-34025669

ABSTRACT

Viral encephalitis is the most common cause of encephalitis. It is responsible for high morbidity rates, permanent neurological sequelae, and even high mortality rates. The host immune response plays a critical role in preventing or clearing invading pathogens, especially when effective antiviral treatment is lacking. However, due to blockade of the blood-brain barrier, it remains unclear how peripheral immune cells contribute to the fight against intracerebral viruses. Here, we report that peripheral injection of an antibody against human Tim-3, an immune checkpoint inhibitor widely expressed on immune cells, markedly attenuated vesicular stomatitis virus (VSV) encephalitis, marked by decreased mortality and improved neuroethology in mice. Peripheral injection of Tim-3 antibody enhanced the recruitment of immune cells to the brain, increased the expression of major histocompatibility complex-I (MHC-I) on macrophages, and as a result, promoted the activation of VSV-specific CD8+ T cells. Depletion of macrophages abolished the peripheral injection-mediated protection against VSV encephalitis. Notably, for the first time, we found a novel post-translational modification of MHC-I by Tim-3, wherein, by enhancing the expression of MARCH9, Tim-3 promoted the proteasome-dependent degradation of MHC-I via K48-linked ubiquitination in macrophages. These results provide insights into the immune response against intracranial infections; thus, manipulating the peripheral immune cells with Tim-3 antibody to fight viruses in the brain may have potential applications for combating viral encephalitis.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antigen-Presenting Cells/drug effects , Brain/drug effects , Encephalitis, Viral/prevention & control , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Macrophages/drug effects , Rhabdoviridae Infections/prevention & control , Vesiculovirus/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , Brain/immunology , Brain/metabolism , Brain/virology , Chlorocebus aethiops , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/metabolism , Encephalitis, Viral/virology , HEK293 Cells , Hepatitis A Virus Cellular Receptor 2/immunology , Histocompatibility Antigens Class I/metabolism , Host-Pathogen Interactions , Humans , Injections, Intraperitoneal , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RAW 264.7 Cells , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/metabolism , Rhabdoviridae Infections/virology , Ubiquitination , Vero Cells , Vesiculovirus/pathogenicity , Viral Load
4.
Cell Rep Med ; 2(3): 100220, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33763658

ABSTRACT

Hantaviruses are zoonotic RNA viruses that cause severe acute disease in humans. Infected individuals have strong inflammatory responses that likely cause immunopathology. Here, we studied the response of mucosal-associated invariant T (MAIT) cells in peripheral blood of individuals with hemorrhagic fever with renal syndrome (HFRS) caused by Puumala orthohantavirus, a hantavirus endemic in Europe. We show that MAIT cell levels decrease in the blood during HFRS and that residual MAIT cells are highly activated. This activation correlates with HFRS severity markers. In vitro activation of MAIT cells by hantavirus-exposed antigen-presenting cells is dependent on type I interferons (IFNs) and independent of interleukin-18 (IL-18). These findings highlight the role of type I IFNs in virus-driven MAIT cell activation and suggest a potential role of MAIT cells in the disease pathogenesis of viral infections.


Subject(s)
Antigen-Presenting Cells/immunology , Hantavirus Infections/immunology , Hemorrhagic Fever with Renal Syndrome/immunology , Lymphocyte Activation , Mucosal-Associated Invariant T Cells/immunology , Puumala virus/pathogenicity , Adult , Antibodies, Viral/blood , Antigen-Presenting Cells/virology , Biomarkers/metabolism , Case-Control Studies , Disease Progression , Endothelial Cells/immunology , Endothelial Cells/virology , Female , Gene Expression Regulation , Hantavirus Infections/genetics , Hantavirus Infections/pathology , Hantavirus Infections/virology , Hemorrhagic Fever with Renal Syndrome/genetics , Hemorrhagic Fever with Renal Syndrome/pathology , Hemorrhagic Fever with Renal Syndrome/virology , Humans , Immunophenotyping , Interferon Type I/genetics , Interferon Type I/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Male , Middle Aged , Monocytes/immunology , Monocytes/virology , Mucosal-Associated Invariant T Cells/virology , Puumala virus/immunology , Severity of Illness Index
5.
Immunity ; 49(4): 678-694.e5, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30314757

ABSTRACT

CD8+ T cell exhaustion impedes control of chronic viral infection; yet how new T cell responses are mounted during chronic infection is unclear. Unlike T cells primed at the onset of infection that rapidly differentiate into effectors and exhaust, we demonstrate that virus-specific CD8+ T cells primed after establishment of chronic LCMV infection preferentially generate memory-like transcription factor TCF1+ cells that were transcriptionally and proteomically distinct, less exhausted, and more responsive to immunotherapy. Mechanistically, adaptations of antigen-presenting cells and diminished T cell signaling intensity promoted differentiation of the memory-like subset at the expense of rapid effector cell differentiation, which was now highly dependent on IL-21-mediated CD4+ T cell help for its functional generation. Chronic viral infection similarly redirected de novo differentiation of tumor-specific CD8+ T cells, ultimately preventing cancer control. Thus, targeting these T cell stimulatory pathways could enable strategies to control chronic infection, tumors, and enhance immunotherapeutic efficacy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Immunity/immunology , Immunologic Memory/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Chronic Disease , Gene Expression Profiling/methods , Immunity/genetics , Immunologic Memory/genetics , Immunotherapy , Lymphocytic Choriomeningitis/therapy , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Proteomics/methods , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/immunology , T Cell Transcription Factor 1/metabolism
6.
Virus Res ; 258: 73-80, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30316802

ABSTRACT

African swine fever (ASF) is a devastating viral disease of domestic pigs and wild boar for which there is no vaccine available. The aetiological agent ASF virus (ASFV) has a predilection for cells of the myeloid lineage. Macrophages provide a first line defence against pathogens and are the main target of ASFV, thus several studies analysed their response to infection in terms of cytokine/chemokine expression and modulation of functionality. These studies have typically used macrophages differentiated in vitro from blood or bone marrow progenitors and few studies have focused on responses of polarized macrophages (M1, M2) or functional macrophage subsets isolated from different tissues. ASFV can also infect dendritic cells (DC), but regardless of their central role in the induction of adaptive immune responses, their role in ASFV infection was only partially analysed. Future studies on ASFV-DC interaction are needed, which should take into consideration the heterogeneity within this family, composed of different subsets whose phenotype is also organ specific. Other porcine immune cells such as γδ-T cells, NK cells and fibrocytes, can act as 'non-conventional' antigen-presenting cells (APCs). In particular, γδ-T cells from ASFV immune pigs were shown to present viral antigens to T cells, but no studies have further explored the interaction of ASFV with this cell type or other non-conventional APCs. In this review we will provide an overview of the interaction of APCs with ASFV, describing the differences between virulent and attenuated strains, and suggesting areas for possible future studies.


Subject(s)
African Swine Fever Virus/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , African Swine Fever/virology , African Swine Fever Virus/pathogenicity , African Swine Fever Virus/physiology , Animals , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Histocompatibility Antigens Class I/genetics , Killer Cells, Natural/immunology , Macrophages/immunology , Macrophages/virology , Swine , Viral Proteins/genetics , Virulence , Virus Replication
7.
Viruses ; 10(2)2018 02 06.
Article in English | MEDLINE | ID: mdl-29415518

ABSTRACT

Myeloid cells such as monocytes, dendritic cells (DC) and macrophages (MΦ) are key components of the innate immune system contributing to the maintenance of tissue homeostasis and the development/resolution of immune responses to pathogens. Monocytes and DC, circulating in the blood or infiltrating various lymphoid and non-lymphoid tissues, are derived from distinct bone marrow precursors and are typically short lived. Conversely, recent studies revealed that subsets of tissue resident MΦ are long-lived as they originate from embryonic/fetal precursors that have the ability to self-renew during the life of an individual. Pathogens such as the human immunodeficiency virus type 1 (HIV-1) highjack the functions of myeloid cells for viral replication (e.g., MΦ) or distal dissemination and cell-to-cell transmission (e.g., DC). Although the long-term persistence of HIV reservoirs in CD4+ T-cells during viral suppressive antiretroviral therapy (ART) is well documented, the ability of myeloid cells to harbor replication competent viral reservoirs is still a matter of debate. This review summarizes the current knowledge on the biology of monocytes and DC during homeostasis and in the context of HIV-1 infection and highlights the importance of future studies on long-lived resident MΦ to HIV persistence in ART-treated patients.


Subject(s)
Dendritic Cells/immunology , HIV Infections/etiology , HIV Infections/metabolism , HIV-1/immunology , Monocytes/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , Dendritic Cells/metabolism , Dendritic Cells/virology , HIV Infections/drug therapy , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Monocytes/metabolism , Monocytes/virology
8.
J Leukoc Biol ; 103(4): 731-738, 2018 04.
Article in English | MEDLINE | ID: mdl-29345359

ABSTRACT

In this work, we identified the expression, regulation, and viral targeting of Scribble and Dlg1 in antigen-presenting cells. Scribble and Dlg1 belong to the family of PDZ (postsynaptic density (PSD95), disc large (Dlg), and zonula occludens (ZO-1)) proteins involved in cell polarity. The relevance of PDZ proteins in cellular functions is reinforced by the fact that many viruses interfere with host PDZ-dependent interactions affecting cellular mechanisms thus favoring viral replication. The functions of Scribble and Dlg have been widely studied in polarized cells such as epithelial and neuron cells. However, within the cells of the immune system, their functions have been described only in T and B lymphocytes. Here we demonstrated that Scribble and Dlg1 are differentially expressed during antigen-presenting cell differentiation and dendritic cell maturation. While both Scribble and Dlg1 seem to participate in distinct dendritic cell functions, both are targeted by the viral protein NS1 of influenza A in a PDZ-dependent manner in dendritic cells. Our findings suggest that these proteins might be involved in the mechanisms of innate immunity and/or antigen processing and presentation that can be hijacked by viral pathogens.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigen-Presenting Cells/immunology , Host-Pathogen Interactions , Influenza A virus/pathogenicity , Influenza, Human/virology , Membrane Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Discs Large Homolog 1 Protein , Humans , Influenza, Human/immunology , Influenza, Human/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Membrane Proteins/genetics , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , PDZ Domains , Tumor Suppressor Proteins/genetics , Viral Nonstructural Proteins/genetics
9.
Microb Pathog ; 110: 325-334, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28710013

ABSTRACT

Rotavirus (RV) infection causes acute, watery dehydrating diarrhea and even death in infants and other young animals, resulting in a severe economic burden; however, little is known about the innate immune mechanisms associated with RV infection. Dendritic cells (DCs), which are professional antigen-presenting cells (APCs), serve as a bridge connecting the innate and adaptive immune system. In this study, the interaction between murine bone marrow-derived DCs (BMDCs) and porcine rotavirus (PRV) was investigated in vitro. Upon stimulation, the expression levels of MHC-II, CD40, CD80, CD86 and CD83 in BMDCs increased in a time-dependent manner, indicating activation and maturation by PRV. In addition, up-regulated Toll-like receptor 2 (TLR2), TLR3 and NF-κB increased the production of interleukin-12 and interferon-γ. The PRV-stimulated BMDCs also showed increased stimulatory capacity in mixed lymphocyte reactions and promoted the Th1 subtype response.


Subject(s)
Bone Marrow/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Rotavirus Infections/immunology , Rotavirus/immunology , Th1 Cells/immunology , Adaptive Immunity , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD40 Antigens/metabolism , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Regulation , Genes, MHC Class II , Host-Pathogen Interactions/immunology , Immunity, Innate , Immunity, Mucosal , Immunoglobulins/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Rotavirus/pathogenicity , Swine , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Up-Regulation , CD83 Antigen
10.
Nanomedicine (Lond) ; 11(14): 1759-67, 2016 07.
Article in English | MEDLINE | ID: mdl-27389189

ABSTRACT

AIM: To engineer a novel nanoimmunotherapy comprising Prussian blue nanoparticles (PBNPs) conjugated to antigen-specific cytotoxic T lymphocytes (CTL), which leverages PBNPs for their photothermal therapy (PTT) capabilities and Epstein-Barr virus (EBV) antigen-specific CTL for their ability to traffic to and destroy EBV antigen-expressing target cells. MATERIALS & METHODS: PBNPs and CTL were independently biofunctionalized. Subsequently, PBNPs were conjugated onto CTL using avidin-biotin interactions. The resultant cell-nanoparticle construct (CTL:PBNPs) were analyzed for their physical, phenotypic and functional properties. RESULTS: Both PBNPs and CTL maintained their intrinsic physical, phenotypic and functional properties within the CTL:PBNPs. CONCLUSION: This study highlights the potential of our CTL:PBNPs nanoimmunotherapy as a novel therapeutic for treating virus-associated malignancies such as EBV+ cancers.


Subject(s)
Coloring Agents/therapeutic use , Epstein-Barr Virus Infections/therapy , Ferrocyanides/therapeutic use , Herpesvirus 4, Human/immunology , Nanoparticles/therapeutic use , Neoplasms/therapy , Neoplasms/virology , T-Lymphocytes, Cytotoxic/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , Coculture Techniques , Coloring Agents/chemistry , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/immunology , Ferrocyanides/chemistry , Humans , Immunotherapy , Jurkat Cells , Lymphocyte Activation , Nanomedicine , Nanoparticles/chemistry , Neoplasms/immunology , Phototherapy , T-Lymphocytes, Cytotoxic/chemistry
11.
Sci Rep ; 6: 29447, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27385120

ABSTRACT

The innate and adaptive immune systems fail to control HCV infection in the majority of infected individuals. HCV is an ssRNA virus, which suggests a role for Toll-like receptors (TLRs) 7 and 8 in initiating the anti-viral response. Here we demonstrate that HCV genomic RNA harbours specific sequences that initiate an anti-HCV immune response through TLR7 and TLR8 in various antigen presenting cells. Conversely, HCV particles are detected by macrophages, but not by monocytes and DCs, through a TLR7/8 dependent mechanism; this leads to chloroquine sensitive production of pro-inflammatory cytokines including IL-1ß, while the antiviral type I Interferon response is not triggered in these cells. Antibodies to DC-SIGN, a c-type lectin selectively expressed by macrophages but not pDCs or mDCs, block the production of cytokines. Novel anti-HCV vaccination strategies should target the induction of TLR7/8 stimulation in APCs in order to establish potent immune responses against HCV.


Subject(s)
Antigen-Presenting Cells/virology , Hepacivirus/genetics , Macrophages/virology , RNA, Viral/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , Antigen-Presenting Cells/immunology , Cell Line , Chloroquine/pharmacology , HEK293 Cells , Hepacivirus/immunology , Hepatitis C/immunology , Humans , Interleukin-1beta/metabolism , Macrophages/metabolism
12.
Papillomavirus Res ; 2: 21-30, 2016 12.
Article in English | MEDLINE | ID: mdl-27182559

ABSTRACT

Human papillomavirus type 16 (HPV16) infections are intra-epithelial, and thus, HPV16 is known to interact with Langerhans cells (LCs), the resident epithelial antigen-presenting cells (APCs). The current paradigm for APC-mediated induction of T cell anergy is through delivery of T cell receptor signals via peptides on MHC molecules (signal 1), but without costimulation (signal 2). We previously demonstrated that LCs exposed to HPV16 in vitro present HPV antigens to T cells without costimulation, but it remained uncertain if such T cells would remain ignorant, become anergic, or in the case of CD4+ T cells, differentiate into Tregs. Here we demonstrate that Tregs were not induced by LCs presenting only signal 1, and through a series of in vitro immunizations show that CD8+ T cells receiving signal 1 + 2 from LCs weeks after consistently receiving signal 1 are capable of robust effector functions. Importantly, this indicates that T cells are not tolerized but instead remain ignorant to HPV, and are activated given the proper signals.


Subject(s)
Antigen Presentation , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Human papillomavirus 16/immunology , Immune Tolerance , Langerhans Cells/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , CD8-Positive T-Lymphocytes/immunology , Cellular Senescence/immunology , Chemokines/immunology , Costimulatory and Inhibitory T-Cell Receptors/immunology , Cytokines/immunology , Humans , Interleukin-2/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology
13.
PLoS Negl Trop Dis ; 10(5): e0004709, 2016 05.
Article in English | MEDLINE | ID: mdl-27191161

ABSTRACT

Humans infected with yellow fever virus (YFV), a mosquito-borne flavivirus, can develop illness ranging from a mild febrile disease to hemorrhagic fever and death. The 17D vaccine strain of YFV was developed in the 1930s, has been used continuously since development and has proven very effective. Genetic differences between vaccine and wild-type viruses are few, yet viral or host mechanisms associated with protection or disease are not fully understood. Over the past 20 years, a number of cases of vaccine-associated disease have been identified following vaccination with 17D; these cases have been correlated with reduced immune status at the time of vaccination. Recently, several studies have evaluated T cell responses to vaccination in both humans and non-human primates, but none have evaluated the response to wild-type virus infection. In the studies described here, monocyte-derived macrophages (MDM) and dendritic cells (MoDC) from both humans and rhesus macaques were evaluated for their ability to support infection with either wild-type Asibi virus or the 17D vaccine strain and the host cytokine and chemokine response characterized. Human MoDC and MDM were also evaluated for their ability to stimulate CD4+ T cells. It was found that MoDC and MDM supported viral replication and that there were differential cytokine responses to infection with either wild-type or vaccine viruses. Additionally, MoDCs infected with live 17D virus were able to stimulate IFN-γ and IL-2 production in CD4+ T cells, while cells infected with Asibi virus were not. These data demonstrate that wild-type and vaccine YFV stimulate different responses in target antigen presenting cells and that wild-type YFV can inhibit MoDC activation of CD4+ T cells, a critical component in development of protective immunity. These data provide initial, but critical insight into regulatory capabilities of wild-type YFV in development of disease.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , CD4-Positive T-Lymphocytes/immunology , Yellow Fever Vaccine/immunology , Yellow fever virus/immunology , Animals , Antigen Presentation , Chemokines/analysis , Chemokines/immunology , Cytokines/analysis , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Host-Pathogen Interactions , Humans , Interferon-gamma/analysis , Interferon-gamma/immunology , Interleukin-2/analysis , Interleukin-2/immunology , Lymphocyte Activation , Macaca mulatta , Macrophages/immunology , Macrophages/virology , Vaccines, Attenuated/immunology , Virus Replication , Yellow fever virus/physiology
14.
PLoS One ; 10(7): e0132942, 2015.
Article in English | MEDLINE | ID: mdl-26172445

ABSTRACT

Studies have shown that the transmission of HIV is most likely to occur via rectal or vaginal routes, and rarely through oral exposure. However, the mechanisms of virus entry at mucosal surfaces remain incompletely understood. Prophylactic strategies against HIV infection may be attainable once gaps in current knowledge are filled. To address these gaps, we evaluated essentially normal epithelial surfaces and mapped the periluminal distribution of CD4+ HIV target cells, including T cells and antigen-presenting cells, and an HIV-binding molecule gp340 that can be expressed by epithelial cells in secreted and cell-associated forms. Immunohistochemistry for CD4, CD16, CD3, CD1a and gp340 in human oral, rectal/sigmoid and cervical mucosal samples from HIV-negative subjects demonstrated that periluminal HIV target cells were more prevalent at rectal/sigmoid and endocervical surfaces lined by simple columnar epithelium, than at oral and ectocervical surfaces covered by multilayered stratified squamous epithelium (p<0.001). gp340 expression patterns at these sites were also distinct and strong in oral minor salivary gland acini and ducts, including ductal saliva, in individual rectum/sigmoid and endocervix periluminar columnar cells, and in ectocervix squamous cells. Only weak expression was noted in the oral non-ductal squamous epithelium. We conclude that periluminal HIV target cells, together with periluminal epithelial cell-associated gp340 appear to be most accessible for HIV transmission at rectal/sigmoid and endocervical surfaces. Our data help define vulnerable structural features of mucosal sites exposed to HIV.


Subject(s)
Cervix Uteri/virology , Colon, Sigmoid/virology , HIV Infections/metabolism , HIV/metabolism , Mucous Membrane/virology , Receptors, Cell Surface/metabolism , Rectum/virology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Calcium-Binding Proteins , Cervix Uteri/metabolism , Colon, Sigmoid/metabolism , DNA-Binding Proteins , Epithelial Cells/metabolism , Epithelial Cells/virology , Epithelium/metabolism , Epithelium/virology , Female , HIV Infections/transmission , Humans , Mucous Membrane/metabolism , Rectum/metabolism , Tumor Suppressor Proteins , Vagina/metabolism , Vagina/virology
15.
Immunology ; 146(2): 312-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26173587

ABSTRACT

Self-amplifying mRNAs (SAM(®) ) are a novel class of nucleic acid vaccines, delivered by a non-viral delivery system. They are effective at eliciting potent and protective immune responses and are being developed as a platform technology with potential to be used for a broad range of targets. However, their mechanism of action has not been fully elucidated. To date, no evidence of in vivo transduction of professional antigen-presenting cells (APCs) by SAM vector has been reported, while the antigen expression has been shown to occur mostly in the muscle fibres. Here we show that bone-marrow-derived APCs rather than muscle cells are responsible for induction of MHC class-I restricted CD8 T cells in vivo, but direct transfection of APCs by SAM vectors is not required. Based on all our in vivo and in vitro data we propose that upon SAM vaccination the antigen is expressed within muscle cells and then transferred to APCs, suggesting cross-priming as the prevalent mechanism for priming the CD8 T-cell response by SAM vaccines.


Subject(s)
Antigen-Presenting Cells/immunology , Bone Marrow Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Muscle Fibers, Skeletal/immunology , RNA, Messenger/immunology , RNA, Viral/immunology , RNA-Binding Proteins/immunology , Viral Core Proteins/immunology , Animals , Antigen-Presenting Cells/virology , Bone Marrow Cells/virology , Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/virology , Cell Communication , Cell Line , Cricetinae , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Influenza A Virus, H1N1 Subtype/genetics , Influenza Vaccines/genetics , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Muscle Fibers, Skeletal/virology , Nucleocapsid Proteins , RNA, Messenger/genetics , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Transfection , Transplantation Chimera , Viral Core Proteins/genetics
16.
J Control Release ; 207: 143-53, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25886704

ABSTRACT

Chronic hepatitis B is a serious liver disease and puts people at high risk of death from cirrhosis and liver cancer. Although DNA vaccination has been emerged as a potential immunotherapeutic strategy for the treatment of chronic hepatitis B, the efficiencies were not adequate in clinical trials. Here we describe the design, synthesis, and evaluation of mannosylated phenylalanine grafted chitosan (Man-CS-Phe) as a DNA delivery vector for direct transfection of antigen presenting cells to improve cellular and humoral immunity to plasmid-coded antigen. The cationic Man-CS-Phe micelles condense plasmid DNA into nanoscale polyplexes and provide efficient protection of complexed DNA from nuclease degradation. The mannose receptor-mediated enhanced cell uptake and high in vitro transfection efficiency of the polyplexes were demonstrated in RAW 264.7 and DC 2.4 cells using GFP-expressing plasmid DNA. Furthermore, intradermal immunization of BALB/c mice indicated that hepatitis B DNA vaccine/Man-CS-Phe polyplexes not only induced multi-fold higher serum antibody titer in comparison to all other formulations including FuGENE HD, but also significantly stimulated T-cell proliferation and skewed T helper toward Th1 polarization. These results illustrate that the Man-CS-Phe can serve as a promising DNA delivery vector to harness both cellular and humoral arms of immune system.


Subject(s)
Antigen-Presenting Cells/immunology , Chitosan/chemistry , Drug Carriers , Hepatitis B Surface Antigens/administration & dosage , Hepatitis B Vaccines/administration & dosage , Mannose/chemistry , Phenylalanine/chemistry , Administration, Cutaneous , Animals , Antigen-Presenting Cells/virology , Biomarkers/blood , Cell Proliferation , Chemistry, Pharmaceutical , Chitosan/analogs & derivatives , Chitosan/toxicity , Female , Hepatitis B Antibodies/blood , Hepatitis B Surface Antigens/biosynthesis , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/biosynthesis , Hepatitis B Vaccines/genetics , Hepatitis B Vaccines/immunology , Immunity, Cellular , Immunity, Humoral , Immunization , Lymphocyte Activation , Mannose/analogs & derivatives , Mannose/toxicity , Mice , Mice, Inbred BALB C , Micelles , Phenylalanine/analogs & derivatives , Phenylalanine/toxicity , RAW 264.7 Cells , T-Lymphocytes, Helper-Inducer/immunology , Transfection , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
17.
J Virol ; 89(3): 1867-78, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25428872

ABSTRACT

UNLABELLED: Intracerebral infection with Theiler's murine encephalomyelitis virus (TMEV) induces immune-mediated demyelinating disease in susceptible SJL/J mice but not in resistant C57BL/6 mice. Previous studies have indicated that the major histocompatibility complex (MHC) genes play the most prominent role in the development of TMEV-induced demyelinating disease. In this study, we used C57BL/6.S (B6.S) congenic mice, which carry H-2(s) MHC genes instead of H-2(b) MHC genes in conjunction with the C57BL/6 (B6) background genes. Our data show that virus-infected B6.S mice are free from disease and have significantly lower viral loads than susceptible SJL mice, particularly in the spinal cord. A strong protective Th1-type T helper response with virtually no pathogenic Th17 response was detected in B6.S mice, in contrast to the reduced Th1- and robust Th17-type responses in SJL mice. Notably, lower levels of viral infectivity in B6.S antigen-presenting cells (APCs) correlated with the disease resistance and T-cell-type response. In vitro studies using APCs from B6.S and SJL mice show that TLR2, -3, -4, and -7, but not TLR9, signaling can replace viral infection and augment the effect of viral infection in the differentiation of the pathogenic Th17 cell type. Taken together, these results strongly suggest that the viral replication levels in APCs critically affect the induction of protective versus pathogenic Th cell types via the signaling of pattern recognition receptors for innate immune responses. Our current findings further imply that the levels of viral infectivity/replication and TLR-mediated signaling play critical roles in the pathogenesis of chronic viral diseases. IMPORTANCE: This study indicates that innate immune cytokines produced in antigen-presenting cells stimulating the T cell immune responses during early viral infection play a critical role in determining the susceptibility of mice to the development of demyelinating disease. The level of innate immune cytokines reflects the level of initial viral infection in the antigen-presenting cells, and the level determines the development of T cell types, which are either protective or pathogenic. The level of initial viral infection to the cells is controlled by a gene or genes that are not associated with the major histocompatibility antigen complex genes. This finding has an important implication in controlling not only chronic viral infections but also infection-induced autoimmune-like diseases, which are closely associated with the pathogenic type of T cell responses.


Subject(s)
Antigen-Presenting Cells/virology , Cardiovirus Infections/pathology , Cardiovirus Infections/virology , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Theilovirus/isolation & purification , Viral Load , Animals , Antigen-Presenting Cells/immunology , Cardiovirus Infections/immunology , Demyelinating Diseases/immunology , Female , Genes, MHC Class I , Mice, Inbred C57BL , Th1 Cells/immunology , Th17 Cells/immunology , Theilovirus/immunology , Toll-Like Receptors/immunology
18.
Comp Immunol Microbiol Infect Dis ; 37(5-6): 321-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25456193

ABSTRACT

The mucosal surfaces are important sites of entry for a majority of pathogens, and viruses in particular. The migration of antigen presenting cells (APCs) from the apical side of the mucosal epithelium to the lymph node is a key event in the development of mucosal immunity during viral infections. However, the mechanism by which viruses utilize the transmigration of these cells to invade the mucosa is largely unexplored. Here, we establish an ex vivo explant model of monocytic cell transmigration across the nasal mucosal epithelium and lamina propria. Equine nasal mucosal CD172a(+) cells (nmCD172a(+) cells), blood-derived monocytes and monocyte-derived DCs (moDCs) were labeled with a fluorescent dye and transferred to the apical part of a polarized mucosal explant. Confocal imaging was used to monitor the migration patterns of monocytic cells and the effect of equine herpesvirus type 1 (EHV-1) on their transmigration. We observed that 16-26% of mock-inoculated nmCD172a(+) cells and moDCs moved into the nasal epithelia, and 1-7% moved further in the lamina propria. The migration of EHV-1 inoculated monocytic cells was not increased in these tissues compared to the mock-inoculated monocytic cells. Immediate early protein positive (IEP(+)) cells were observed beneath the basement membrane (BM) 48 hours post addition (hpa) of moDCs and nmCD172a(+) cells, but not blood-derived monocytes. Together, our finding demonstrate that monocytic cells may become infected with EHV-1 in the respiratory mucosa and transport the virus from the apical side of the epithelium to the lamina propria en route to the lymph and blood circulation.


Subject(s)
Antigen-Presenting Cells/immunology , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/immunology , Horse Diseases/immunology , Horses/immunology , Immunity, Mucosal , Monocytes/immunology , Animals , Antigen-Presenting Cells/pathology , Antigen-Presenting Cells/virology , Cell Movement/immunology , Cell Polarity , Female , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Herpesvirus 1, Equid/pathogenicity , Horse Diseases/pathology , Horse Diseases/virology , Horses/virology , Male , Monocytes/pathology , Monocytes/virology , Nasal Mucosa/immunology , Nasal Mucosa/pathology , Nasal Mucosa/virology , Tissue Culture Techniques , Virus Replication/physiology
19.
Curr Opin HIV AIDS ; 9(5): 478-84, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25093528

ABSTRACT

PURPOSE OF REVIEW: The purpose of this study is to describe the alterations that HIV-1 induces in antigen-presenting cells (APCs), in vitro, ex vivo and in vivo. RECENT FINDINGS: HIV-1 disarms several arms of the immune system including APCs. We summarize here recent findings on the impact of the virus on APC. SUMMARY: HIV-1 can invade APC and overall reduce their capacity to present antigens effectively, mostly by reducing their numbers and inducing permanent hyperactivation. This occurs via a combination of alterations; however, the host can counteract, at least in part, some of these defects via restriction factors, autophagy, the production of type I interferon, antiviral cytokines, among others. However, these specific mechanisms of viral evasion from APCs' control lead to a chronic hyperactivation of the immune system implicated in AIDS-related and non-AIDS related pathogenesis. Unfortunately, the current regimens of antiretroviral therapy are unable to dampen sufficiently APC-driven viral-induced immune hyperactivation. Understanding how HIV alters APC will help to tune appropriately both intrinsic immunity and innate immunity, as well as achieve efficient antigen presentation to the adaptive immune system, without inducing a detrimental pervasive hyperactivation of the immune system.


Subject(s)
Antigen-Presenting Cells , HIV Infections , Anti-Retroviral Agents , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/physiology , Antigen-Presenting Cells/virology , HIV Infections/immunology , HIV Infections/physiopathology , HIV Infections/virology , HIV-1/immunology , Humans , Interferons
20.
J Immunol ; 193(3): 1223-32, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24951814

ABSTRACT

Adenoviral vectors have long been forerunners in the development of effective CD8 T cell-based vaccines; therefore, it is imperative that we understand the factors controlling the induction of robust and long-lasting transgene-specific immune responses by these vectors. In this study, we investigated the organ sites, molecules, and cell subsets that play a critical role in the priming of transgene-specific CD8 T cells after vaccination with a replication-deficient adenoviral vector. Using a human adenovirus serotype 5 (Ad5) vector and genetically engineered mice, we found that CD8(+) and/or CD103(+) dendritic cells in the draining lymph node played a critical role in the priming of Ad5-induced CD8 T cell responses. Moreover, we found that CD80/86, but not CD28, was essential for efficient generation of both primary effectors and memory CD8 T cells. Interestingly, the lack of CD28 expression resulted in a delayed primary response, whereas memory CD8 T cells generated in CD28-deficient mice appeared almost normal in terms of both phenotype and effector cytokine profile, but they exhibited a significantly reduced proliferative capacity upon secondary challenge while retaining immediate in vivo effector capabilities: in vivo cytotoxicity and short-term in vivo protective capacity. Overall, our data point to an absolute requirement for professional APCs and the expression of the costimulatory molecules CD80/86 for efficient CD8 T cell priming by adenoviral vectors. Additionally, our results suggest the existence of an alternative receptor for CD80/86, which may substitute, in part, for CD28.


Subject(s)
Adenovirus Infections, Human/prevention & control , Adenoviruses, Human/immunology , B7-1 Antigen/deficiency , B7-2 Antigen/deficiency , CD28 Antigens/deficiency , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Genetic Engineering , Adenovirus Infections, Human/genetics , Adenoviruses, Human/genetics , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/virology , B7-1 Antigen/genetics , B7-2 Antigen/genetics , CD28 Antigens/genetics , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Dendritic Cells/metabolism , Dendritic Cells/virology , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Humans , Ligands , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...