Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Pediatr Infect Dis J ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38920375

ABSTRACT

A multicenter cross-sectional diagnostic study was carried out including 45 children with nontuberculous mycobacterial cervicofacial lymphadenitis and controls. The tested immunoassay, detecting M. avium-specific anti-glycopeptidolipid-core immunoglobulin A antibodies, had inadequate diagnostic performance in the studied population and seems to be of no additional value in detecting cases of nontuberculous mycobacterial cervicofacial lymphadenitis.

2.
Front Med (Lausanne) ; 10: 1146457, 2023.
Article in English | MEDLINE | ID: mdl-37261119

ABSTRACT

Background: Toll-like receptor (TLR) agonists have been investigated due to their potential dual effects as latency reverting agents and immune modulatory compounds in people living with HIV (PLWH). Here, we investigated whether co-stimulation of TLR7/8 agonists with RIG-I-like receptor (RLR) agonists enhances antiviral immunity. Methods: Peripheral blood mononuclear cells (PBMCs) and monocyte-derived dendritic cells (DCs) were incubated with TLR and RLR-agonists for 24 h and innate and adaptive immune responses were determined (maturation markers, cytokines in supernatant, ISG expression). Results: Both TLR7 and TLR8 agonists induced pro-inflammatory cytokines in DCs as well as PBMCs. TLR8 agonists were more potent in inducing cytokine responses and had a stronger effect on DC-induced immunity. Notably, while all compounds induced IL-12p70, co-stimulation with TLR8 agonists and RLR agonist polyI: C induced significantly higher levels of IL-12p70 in PBMCs. Moreover, crosstalk between TLR8 and RLR agonists induced a strong type I Interferon (IFN) response as different antiviral IFN-stimulated genes were upregulated by the combination compared to the agonists alone. Conclusion: Our data strongly suggest that TLR crosstalk with RLRs leads to strong antiviral immunity as shown by induction of IL-12 and type I IFN responses in contrast to TLRs alone. Thus, co-stimulation of TLRs and RLRs might be a powerful strategy to induce reactivation of latent reservoir as well as antiviral immunity that eliminates the reactivated cells.

3.
Nat Immunol ; 23(12): 1735-1748, 2022 12.
Article in English | MEDLINE | ID: mdl-36456734

ABSTRACT

The non-pathogenic TH17 subset of helper T cells clears fungal infections, whereas pathogenic TH17 cells cause inflammation and tissue damage; however, the mechanisms controlling these distinct responses remain unclear. Here we found that fungi sensing by the C-type lectin dectin-1 in human dendritic cells (DCs) directed the polarization of non-pathogenic TH17 cells. Dectin-1 signaling triggered transient and intermediate expression of interferon (IFN)-ß in DCs, which was mediated by the opposed activities of transcription factors IRF1 and IRF5. IFN-ß-induced signaling led to integrin αvß8 expression directly and to the release of the active form of the cytokine transforming growth factor (TGF)-ß indirectly. Uncontrolled IFN-ß responses as a result of IRF1 deficiency induced high expression of the IFN-stimulated gene BST2 in DCs and restrained TGF-ß activation. Active TGF-ß was required for polarization of non-pathogenic TH17 cells, whereas pathogenic TH17 cells developed in the absence of active TGF-ß. Thus, dectin-1-mediated modulation of type I IFN responses allowed TGF-ß activation and non-pathogenic TH17 cell development during fungal infections in humans.


Subject(s)
Dendritic Cells , Interferon Type I , Mycoses , Humans , Cytokines/metabolism , Dendritic Cells/metabolism , Interferon Type I/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Th17 Cells/metabolism , Transforming Growth Factor beta/metabolism , Mycoses/immunology
4.
mBio ; 13(6): e0255822, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36326251

ABSTRACT

New SARS-CoV-2 variants of concern and waning immunity demonstrate the need for a quick and simple prophylactic agent to prevent infection. Low molecular weight heparins (LMWH) are potent inhibitors of SARS-CoV-2 binding and infection in vitro. The airways are a major route for infection and therefore inhaled LMWH could be a prophylactic treatment against SARS-CoV-2. We investigated the efficacy of in vivo inhalation of LMWH in humans to prevent SARS-CoV-2 attachment to nasal epithelial cells in a single-center, open-label intervention study. Volunteers received enoxaparin in the right and a placebo (NaCl 0.9%) in the left nostril using a nebulizer. After application, nasal epithelial cells were retrieved with a brush for ex-vivo exposure to either SARS-CoV-2 pseudovirus or an authentic SARS-CoV-2 isolate and virus attachment as determined. LMWH inhalation significantly reduced attachment of SARS-CoV-2 pseudovirus as well as authentic SARS-CoV-2 to human nasal cells. Moreover, in vivo inhalation was as efficient as in vitro LMWH application. Cell phenotyping revealed no differences between placebo and treatment groups and no adverse events were observed in the study participants. Our data strongly suggested that inhalation of LMWH was effective to prevent SARS-CoV-2 attachment and subsequent infection. LMWH is ubiquitously available, affordable, and easy to apply, making them suitable candidates for prophylactic treatment against SARS-CoV-2. IMPORTANCE New SARS-CoV-2 variants of concern and waning immunity demonstrate the need for a quick and simple agent to prevent infection. Low molecular weight heparins (LMWH) have been shown to inhibit SARS-CoV-2 in experimental settings. The airways are a major route for SARS-CoV-2 infection and inhaled LMWH could be a prophylactic treatment. We investigated the efficacy of inhalation of the LMWH enoxaparin in humans to prevent SARS-CoV-2 attachment because this is a prerequisite for infection. Volunteers received enoxaparin in the right and a placebo in the left nostril using a nebulizer. Subsequently, nasal epithelial cells were retrieved with a brush and exposed to SARS-CoV-2. LMWH inhalation significantly reduced the binding of SARS-Cov-2 to human nasal cells. Cell phenotyping revealed no differences between placebo and treatment groups and no adverse events were observed in the participants. Our data indicated that LMWH can be used to block SARS-CoV-2 attachment to nasal cells. LMWH was ubiquitously available, affordable, and easily applicable, making them excellent candidates for prophylactic treatment against SARS-CoV-2.


Subject(s)
COVID-19 , Heparin, Low-Molecular-Weight , Humans , Heparin, Low-Molecular-Weight/adverse effects , SARS-CoV-2 , Enoxaparin/therapeutic use
5.
EMBO J ; 40(20): e106765, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34510494

ABSTRACT

The current pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and outbreaks of new variants highlight the need for preventive treatments. Here, we identified heparan sulfate proteoglycans as attachment receptors for SARS-CoV-2. Notably, neutralizing antibodies against SARS-CoV-2 isolated from COVID-19 patients interfered with SARS-CoV-2 binding to heparan sulfate proteoglycans, which might be an additional mechanism of antibodies to neutralize infection. SARS-CoV-2 binding to and infection of epithelial cells was blocked by low molecular weight heparins (LMWH). Although dendritic cells (DCs) and mucosal Langerhans cells (LCs) were not infected by SARS-CoV-2, both DC subsets efficiently captured SARS-CoV-2 via heparan sulfate proteoglycans and transmitted the virus to ACE2-positive cells. Notably, human primary nasal cells were infected by SARS-CoV-2, and infection was blocked by pre-treatment with LMWH. These data strongly suggest that heparan sulfate proteoglycans are important attachment receptors facilitating infection and transmission, and support the use of LMWH as prophylaxis against SARS-CoV-2 infection.


Subject(s)
COVID-19/transmission , Heparan Sulfate Proteoglycans/metabolism , Heparin, Low-Molecular-Weight/pharmacology , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/immunology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Neutralizing/pharmacology , Chlorocebus aethiops , Dendritic Cells/metabolism , Dendritic Cells/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Host-Pathogen Interactions , Humans , Mucous Membrane/cytology , Mucous Membrane/virology , SARS-CoV-2/metabolism , Syndecan-1/metabolism , Syndecan-4/metabolism , Vero Cells , COVID-19 Drug Treatment
6.
Mucosal Immunol ; 14(3): 743-750, 2021 05.
Article in English | MEDLINE | ID: mdl-33568786

ABSTRACT

Semen is important in determining HIV-1 susceptibility but it is unclear how it affects virus transmission during sexual contact. Mucosal Langerhans cells (LCs) are the first immune cells to encounter HIV-1 during sexual contact and have a barrier function as LCs are restrictive to HIV-1. As semen from people living with HIV-1 contains complement-opsonized HIV-1, we investigated the effect of complement on HIV-1 dissemination by human LCs in vitro and ex vivo. Notably, pre-treatment of HIV-1 with semen enhanced LC infection compared to untreated HIV-1 in the ex vivo explant model. Infection of LCs and transmission to target cells by opsonized HIV-1 was efficiently inhibited by blocking complement receptors CR3 and CR4. Complement opsonization of HIV-1 enhanced uptake, fusion, and integration by LCs leading to an increased transmission of HIV-1 to target cells. However, in the absence of both CR3 and CR4, C-type lectin receptor langerin was able to restrict infection of complement-opsonized HIV-1. These data suggest that complement enhances HIV-1 infection of LCs by binding CR3 and CR4, thereby bypassing langerin and changing the restrictive nature of LCs into virus-disseminating cells. Targeting complement factors might be effective in preventing HIV-1 transmission.


Subject(s)
HIV Infections/immunology , HIV-1/physiology , Langerhans Cells/immunology , Semen/immunology , Antibodies, Blocking/metabolism , Antigens, CD/metabolism , Cell Line , Complement Activation , Disease Transmission, Infectious , HIV Infections/transmission , HIV-1/pathogenicity , Host-Parasite Interactions , Humans , Immune Evasion , Integrin alphaXbeta2/metabolism , Lectins, C-Type/metabolism , Macrophage-1 Antigen/metabolism , Mannose-Binding Lectins/metabolism , Opsonization , Semen/virology
7.
Front Immunol ; 11: 503, 2020.
Article in English | MEDLINE | ID: mdl-32292405

ABSTRACT

Sexually transmitted Hepatitis C virus (HCV) infections and high reinfections are a major concern amongst men who have sex with men (MSM) living with HIV-1 and HIV-negative MSM. Immune activation and/or HIV-1 coinfection enhance HCV susceptibility via sexual contact, suggesting that changes in immune cells or external factors are involved in increased susceptibility. Activation of anal mucosal Langerhans cells (LCs) has been implicated in increased HCV susceptibility as activated but not immature LCs efficiently retain and transmit HCV to other cells. However, the underlying molecular mechanism of transmission remains unclear. Here we identified the Heparan Sulfate Proteoglycan Syndecan 4 as the molecular switch, controlling HCV transmission by LCs. Syndecan 4 was highly upregulated upon activation of LCs and interference with Heparan Sulfate Proteoglycans or silencing of Syndecan 4 abrogated HCV transmission. These data strongly suggest that Syndecan 4 mediates HCV transmission by activated LCs. Notably, our data also identified the C-type lectin receptor langerin as a restriction factor for HCV infection and transmission. Langerin expression abrogated HCV infection in HCV permissive cells, whereas langerin expression on the Syndecan 4 expressing cell line strongly decreased HCV transmission to a target hepatoma cell line. These data suggest that the balanced interplay between langerin restriction and Syndecan 4 transmission determines HCV dissemination. Silencing of langerin enhanced HCV transmission whereas silencing Syndecan 4 on activated LCs decreased transmission. Blocking Heparan Sulfate Proteoglycans abrogated HCV transmission by LCs ex vivo identifying Heparan Sulfate Proteoglycans and Syndecan 4 as potential targets to prevent sexual transmission of HCV. Thus, our data strongly suggest that the interplay between receptors promotes or restricts transmission and further indicate that Syndecan 4 is the molecular switch controlling HCV susceptibility after sexual contact.


Subject(s)
Antigens, CD/metabolism , HIV Infections/metabolism , HIV-1/physiology , Hepacivirus/physiology , Hepatitis C/metabolism , Langerhans Cells/physiology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Sexually Transmitted Diseases/metabolism , Syndecan-4/metabolism , Antigens, CD/genetics , Cell Differentiation , Cell Line , Coinfection , Disease Transmission, Infectious , Homosexuality, Male , Humans , Lectins, C-Type/genetics , Male , Mannose-Binding Lectins/genetics , RNA, Small Interfering/genetics , Syndecan-4/genetics , Up-Regulation
8.
Front Immunol ; 11: 8, 2020.
Article in English | MEDLINE | ID: mdl-32038656

ABSTRACT

Strong innate and adaptive immune responses are paramount in combating viral infections. Dendritic cells (DCs) detect viral infections via cytosolic RIG-I like receptors (RLRs) RIG-I and MDA5 leading to MAVS-induced immunity. The DEAD-box RNA helicase DDX3 senses abortive human immunodeficiency virus 1 (HIV-1) transcripts and induces MAVS-dependent type I interferon (IFN) responses, suggesting that abortive HIV-1 RNA transcripts induce antiviral immunity. Little is known about the induction of antiviral immunity by DDX3-ligand abortive HIV-1 RNA. Here we synthesized a 58 nucleotide-long capped RNA (HIV-1 Cap-RNA58) that mimics abortive HIV-1 RNA transcripts. HIV-1 Cap-RNA58 induced potent type I IFN responses in monocyte-derived DCs, monocytes, macrophages and primary CD1c+ DCs. Compared with RLR agonist poly-I:C, HIV-1 Cap-RNA58 induced comparable levels of type I IFN responses, identifying HIV-1 Cap-RNA58 as a potent trigger of antiviral immunity. In monocyte-derived DCs, HIV-1 Cap-RNA58 activated the transcription factors IRF3 and NF-κB. Moreover, HIV-1 Cap-RNA58 induced DC maturation and the expression of pro-inflammatory cytokines. HIV-1 Cap-RNA58-stimulated DCs induced proliferation of CD4+ and CD8+ T cells and differentiated naïve T helper (TH) cells toward a TH2 phenotype. Importantly, treatment of DCs with HIV-1 Cap-RNA58 resulted in an efficient antiviral innate immune response that reduced ongoing HIV-1 replication in DCs. Our data strongly suggest that HIV-1 Cap-RNA58 induces potent innate and adaptive immune responses, making it an interesting addition in vaccine design strategies.


Subject(s)
Adaptive Immunity , HIV Infections/immunology , HIV-1/genetics , Host Microbial Interactions/immunology , Immunity, Innate , RNA, Viral/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/virology , HIV Infections/virology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Macrophages/immunology , Macrophages/virology , Monocytes/immunology , Monocytes/virology , NF-kappa B/metabolism , RNA, Viral/chemical synthesis , RNA, Viral/immunology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transcription, Genetic
9.
J Immunol ; 204(2): 386-393, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31818980

ABSTRACT

The spirochete Borrelia miyamotoi has recently been shown to cause relapsing fever. Like the Lyme disease agent, Borrelia burgdorferi, B. miyamotoi is transmitted through the bite of infected ticks; however, little is known about the response of the immune system upon infection. Dendritic cells (DCs) play a central role in the early immune response against B. burgdorferi We investigated the response of DCs to two different strains of B. miyamotoi using in vitro and ex vivo models and compared this to the response elicited by B. burgdorferi. Our findings show that B. miyamotoi is phagocytosed by monocyte-derived DCs, causing upregulation of activation markers and production of proinflammatory cytokines in a similar manner to B. burgdorferi. Recognition of B. miyamotoi was demonstrated to be partially mediated by TLR2. DCs migrated out of human skin explants upon inoculation of the skin with B. miyamotoi. Finally, we showed that B. miyamotoi-stimulated DCs induced proliferation of naive CD4+ and CD8+ T cells to a larger extent than B. burgdorferi. In conclusion, we show in this study that DCs respond to and mount an immune response against B. miyamotoi that is similar to the response to B. burgdorferi and is able to induce T cell proliferation.


Subject(s)
Borrelia/physiology , Dendritic Cells/immunology , Insect Bites and Stings/immunology , Relapsing Fever/immunology , Skin/pathology , T-Lymphocytes/immunology , Ticks/immunology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation , Phagocytosis , Ticks/microbiology , Toll-Like Receptor 2/metabolism
10.
PLoS One ; 14(12): e0226651, 2019.
Article in English | MEDLINE | ID: mdl-31856198

ABSTRACT

A single HIV-1 variant establishes infection of the host after sexual contact. Identifying the phenotypic characteristics of these Transmitted Founder (T/F) viruses is important to understand the restriction mechanisms during transmission. Langerhans cells (LCs) are the mucosal dendritic cell subset that has been shown to have a protective role in HIV-1 transmission. Immature LCs efficiently capture and degrade HIV-1 via langerin-mediated restriction. Here we have investigated the capacity of T/F HIV-1 strains to infect mucosal Langerhans cells (LCs). Notably, most T/F variants efficiently infected immature LCs derived from skin and vaginal tissue in contrast to chronic HIV-1 laboratory strains. Next we screened a panel of T/F viruses and their matched 6-month consensus sequence viruses. Interestingly most T/F variants infected immature LCs whereas donor-matched 6-month consensus sequence viruses had lost the ability to infect LCs. However, we also identified 6-month consensus sequence viruses that had retained an ability to infect LCs similar to that of the donor-matched T/F virus. Moreover, some T/F viruses and 6-month consensus sequence viruses were unable to infect immature LCs. Further analyses indicated that T/F viruses are less sensitive to langerin-mediated restriction. These data suggest that T/F HIV-1 variants have the ability to infect immature LCs, which will facilitate transmission.


Subject(s)
HIV-1/pathogenicity , Host-Pathogen Interactions , Langerhans Cells/virology , Antigens, CD/metabolism , Cells, Cultured , HEK293 Cells , Humans , Langerhans Cells/immunology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism
11.
PLoS Pathog ; 13(11): e1006738, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29186193

ABSTRACT

Follicular T helper cells (TFH) are fundamental in orchestrating effective antibody-mediated responses critical for immunity against viral infections and effective vaccines. However, it is unclear how virus infection leads to TFH induction. We here show that dengue virus (DENV) infection of human dendritic cells (DCs) drives TFH formation via crosstalk of RIG-I-like receptor (RLR) RIG-I and MDA5 with type I Interferon (IFN) signaling. DENV infection leads to RLR-dependent IKKε activation, which phosphorylates IFNα/ß receptor-induced STAT1 to drive IL-27 production via the transcriptional complex ISGF3. Inhibiting RLR activation as well as neutralizing antibodies against IL-27 prevented TFH formation. DENV-induced CXCR5+PD-1+Bcl-6+ TFH cells secreted IL-21 and activated B cells to produce IgM and IgG. Notably, RLR activation by synthetic ligands also induced IL-27 secretion and TFH polarization. These results identify an innate mechanism by which antibodies develop during viral disease and identify RLR ligands as potent adjuvants for TFH-promoting vaccination strategies.


Subject(s)
Antibodies, Viral/immunology , Dengue Virus/physiology , Dengue/immunology , T-Lymphocytes, Helper-Inducer/immunology , Antibody Formation , B-Lymphocytes/immunology , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Dendritic Cells/immunology , Dengue/genetics , Dengue/virology , Humans , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Interleukin-27/genetics , Interleukin-27/immunology , Interleukins/genetics , Interleukins/immunology , Lymphocyte Activation , Receptors, Immunologic
12.
PLoS One ; 12(10): e0185580, 2017.
Article in English | MEDLINE | ID: mdl-28976999

ABSTRACT

Microbial DNA is highly immunostimulatory and is sensed by endosomal pattern recognition receptors after release from internalized microbes. It is unclear how extracellular DNA released from dead microbes is delivered to endosomal PRRs to induce immune responses. Here we have investigated the ability of DCs to bind and internalize extracellular E.coli DNA as well as synthetic DNA. DCs internalized E.coli and synthetic DNA, which was dependent on the C-type lectin receptor DC-SIGN. Notably, endosomal uptake of DNA by DCs enhanced TLR9-dependent responses of B cells against DNA. Hence, we have identified DC-SIGN as a cell surface receptor for DNA that facilitates immune responses directed against DNA.


Subject(s)
B-Lymphocytes/immunology , Cell Adhesion Molecules/physiology , DNA, Bacterial/immunology , Dendritic Cells/immunology , Escherichia coli/genetics , Lectins, C-Type/physiology , Receptors, Cell Surface/physiology , Cytokines/biosynthesis , Humans , Interferon Type I/biosynthesis
13.
J Immunol ; 198(12): 4764-4771, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28507028

ABSTRACT

Dengue virus (DENV) causes 400 million infections annually and is one of several viruses that can cause viral hemorrhagic fever, which is characterized by uncontrolled immune activation resulting in high fever and internal bleeding. Although the underlying mechanisms are unknown, massive cytokine secretion is thought to be involved. Dendritic cells (DCs) are the main target cells of DENV, and we investigated their role in DENV-induced cytokine production and adaptive immune responses. DENV infection induced DC maturation and secretion of IL-1ß, IL-6, and TNF. Inhibition of DENV RNA replication abrogated these responses. Notably, silencing of RNA sensors RIG-I or MDA5 abrogated DC maturation, as well as cytokine responses by DENV-infected DCs. DC maturation was induced by type I IFN responses because inhibition of IFN-α/ß receptor signaling abrogated DENV-induced DC maturation. Moreover, DENV infection of DCs resulted in CCL2, CCL3, and CCL4 expression, which was abrogated after RIG-I and MDA5 silencing. DCs play an essential role in TH cell differentiation, and we show that RIG-I and MDA5 triggering by DENV leads to TH1 polarization, which is characterized by high levels of IFN-γ. Notably, cytokines IL-6, TNF, and IFN-γ and chemokines CCL2, CCL3, and CCL4 have been associated with disease severity, endothelial dysfunction, and vasodilation. Therefore, we identified RIG-I and MDA5 as critical players in innate and adaptive immune responses against DENV, and targeting these receptors has the potential to decrease hemorrhagic fever in patients.


Subject(s)
DEAD Box Protein 58/immunology , Dendritic Cells/immunology , Dengue Virus/immunology , Th1 Cells/immunology , Cell Differentiation , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL3/genetics , Chemokine CCL3/immunology , Chemokine CCL4/genetics , Chemokine CCL4/immunology , DEAD Box Protein 58/deficiency , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Dendritic Cells/virology , Humans , Interferon-Induced Helicase, IFIH1/deficiency , Interferon-Induced Helicase, IFIH1/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Receptors, Immunologic , Th1 Cells/physiology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
15.
Nat Immunol ; 18(2): 225-235, 2017 02.
Article in English | MEDLINE | ID: mdl-28024153

ABSTRACT

The mechanisms by which human immunodeficiency virus 1 (HIV-1) avoids immune surveillance by dendritic cells (DCs), and thereby prevents protective adaptive immune responses, remain poorly understood. Here we showed that HIV-1 actively arrested antiviral immune responses by DCs, which contributed to efficient HIV-1 replication in infected individuals. We identified the RNA helicase DDX3 as an HIV-1 sensor that bound abortive HIV-1 RNA after HIV-1 infection and induced DC maturation and type I interferon responses via the signaling adaptor MAVS. Notably, HIV-1 recognition by the C-type lectin receptor DC-SIGN activated the mitotic kinase PLK1, which suppressed signaling downstream of MAVS, thereby interfering with intrinsic host defense during HIV-1 infection. Finally, we showed that PLK1-mediated suppression of DDX3-MAVS signaling was a viral strategy that accelerated HIV-1 replication in infected individuals.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dendritic Cells/virology , HIV Infections/immunology , HIV-1/physiology , Immune Evasion , Immunity , Macrophages/virology , Adaptor Proteins, Signal Transducing/genetics , Cell Extracts , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cohort Studies , DEAD-box RNA Helicases/metabolism , Dendritic Cells/immunology , Gene Expression Regulation, Viral , HEK293 Cells , HIV Infections/virology , Host-Pathogen Interactions/genetics , Humans , Interferon-beta/blood , Macrophages/immunology , Polymorphism, Single Nucleotide , RNA, Viral/immunology , RNA, Viral/metabolism , Receptors, Pattern Recognition/metabolism , Signal Transduction , Viral Load/genetics
16.
Front Pharmacol ; 6: 64, 2015.
Article in English | MEDLINE | ID: mdl-25870561

ABSTRACT

Traditional medicines that stimulate or modulate the immune system can be used as innovative approaches to treat immunological diseases. The herbal medicine IMOD has been shown to strongly modulate immune responses in several animal studies as well as in clinical trials. However, little is known about the mechanisms of IMOD to modulate immunity. Here we have investigated whether IMOD modulates the immunological function of human dendritic cells (DCs). IMOD alone did not induce DC maturation nor production of cytokines. Notably, IMOD decreased the production of pro-inflammatory cytokines IL-6, IL-12 p70, and TNFα by LPS-activated DCs at both mRNA and protein levels in a dose dependent manner. In contrast, treatment with IMOD did not affect LPS induced-production of the anti-inflammatory cytokine IL-10. Furthermore, IMOD inhibited T cell activation/proliferation by LPS-treated DCs and skewed T-cells responses toward the T helper type 2 polarization. These data strongly indicate that IMOD has a potent immunomodulatory ability that affects TLR signaling and thereby modulates DC function. Insight into the immunomodulatory effect of herbal medicine IMOD may provide innovative strategies to affect the immune system and to help combat various diseases.

17.
Nat Commun ; 5: 5074, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25278262

ABSTRACT

Dendritic cells (DCs) orchestrate antibody-mediated responses to combat extracellular pathogens including parasites by initiating T helper cell differentiation. Here we demonstrate that carbohydrate-specific signalling by DC-SIGN drives follicular T helper cell (TFH) differentiation via IL-27 expression. Fucose, but not mannose, engagement of DC-SIGN results in activation of IKKε, which collaborates with type I IFNR signalling to induce formation and activation of transcription factor ISGF3. Notably, ISGF3 induces expression of IL-27 subunit p28, and subsequent IL-27 secreted by DC-SIGN-primed DCs is pivotal for the induction of Bcl-6(+)CXCR5(+)PD-1(hi)Foxp1(lo) TFH cells, IL-21 secretion by TFH cells and T-cell-dependent IgG production by B cells. Thus, we have identified an essential role for DC-SIGN-induced ISGF3 by fucose-based PAMPs in driving IL-27 and subsequent TFH polarization, which might be harnessed for vaccination design.


Subject(s)
Cell Adhesion Molecules/metabolism , Dendritic Cells/cytology , Fucose/chemistry , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interleukin-27/metabolism , Lectins, C-Type/metabolism , Receptors, Cell Surface/metabolism , T-Lymphocytes, Helper-Inducer/cytology , Amino Acid Motifs , B-Lymphocytes/cytology , Cell Differentiation , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Flow Cytometry , Humans , Immunoglobulin G/chemistry , Interferon Regulatory Factor-7/metabolism , Leukocytes, Mononuclear/cytology , Lymphocyte Activation/immunology , Mannose/chemistry , Proto-Oncogene Proteins c-bcl-6 , RNA Interference , Signal Transduction
18.
Cell Host Microbe ; 16(1): 31-42, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-25011106

ABSTRACT

Dendritic cells (DCs) are targets of measles virus (MV) and play central roles in viral dissemination. However, DCs express the RIG-I-like receptors (RLRs) RIG-I and Mda5 that sense MV and induce type I interferon (IFN) production. Given the potency of this antiviral response, RLRs are tightly regulated at various steps, including dephosphorylation by PP1 phosphatases, which induces their activation. We demonstrate that MV suppresses RIG-I and Mda5 by activating the C-type lectin DC-SIGN and inducing signaling that prevents RLR dephosphorylation. MV binding to DC-SIGN leads to activation of the kinase Raf-1, which induces the association of PP1 inhibitor I-1 with GADD34-PP1 holoenzymes, thereby inhibiting phosphatase activity. Consequently, GADD34-PP1 holoenzymes are unable to dephosphorylate RIG-I and Mda5, hence suppressing type I IFN responses and enhancing MV replication. Blocking DC-SIGN signaling allows RLR activation and suppresses MV infection of DCs. Thus, MV subverts DC-SIGN to control RLR activation and escape antiviral responses.


Subject(s)
Cell Adhesion Molecules/metabolism , DEAD-box RNA Helicases/metabolism , Dendritic Cells/immunology , Host-Pathogen Interactions , Lectins, C-Type/metabolism , Measles virus/immunology , Protein Phosphatase 1/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Cell Line , DEAD Box Protein 58 , Dendritic Cells/virology , Humans , Immune Evasion , Measles virus/physiology , Receptors, Immunologic
19.
Nat Commun ; 5: 3898, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24867235

ABSTRACT

Carbohydrate-specific signalling through DC-SIGN provides dendritic cells with plasticity to tailor immunity to the nature of invading microbes. Here we demonstrate that recognition of fucose-expressing extracellular pathogens like Schistosoma mansoni and Helicobacter pylori by DC-SIGN favors T helper cell type-2 (TH2) responses via activation of atypical NF-κB family member Bcl3. Crosstalk between TLR and DC-SIGN signalling results in TLR-induced MK2-mediated phosphorylation of LSP1, associated with DC-SIGN, upon fucose binding. Subsequently, IKKε and CYLD are recruited to phosphorylated LSP1. IKKε activation is pivotal for suppression of CYLD deubiquitinase activity and subsequent nuclear translocation of ubiquitinated Bcl3. Bcl3 activation represses TLR-induced proinflammatory cytokine expression, while enhancing interleukin-10 (IL-10) and TH2-attracting chemokine expression, shifting TH differentiation from TH1 to TH2 polarization. Thus, DC-SIGN directs adaptive TH2 immunity to fucose-expressing pathogens via an IKKε-CYLD-dependent signalling pathway leading to Bcl3 activation, which might be targeted in vaccination strategies or to prevent aberrant inflammation and allergy.


Subject(s)
Cell Adhesion Molecules/metabolism , Fucose/metabolism , I-kappa B Kinase/metabolism , Lectins, C-Type/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Th2 Cells/immunology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , B-Cell Lymphoma 3 Protein , Cell Differentiation/drug effects , Cell Polarity/drug effects , Chemokines/genetics , Chemokines/metabolism , Deubiquitinating Enzyme CYLD , Down-Regulation/drug effects , Enzyme Activation/drug effects , Helicobacter pylori/immunology , Humans , Inflammation Mediators/metabolism , Lewis X Antigen/metabolism , Lipopolysaccharides/pharmacology , Microfilament Proteins/metabolism , Models, Biological , NF-kappa B/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schistosoma mansoni/immunology , Signal Transduction/drug effects , Th2 Cells/drug effects , Up-Regulation/drug effects
20.
Cell Host Microbe ; 15(4): 494-505, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24721577

ABSTRACT

Recognition of fungal pathogens by C-type lectin receptor (CLR) dectin-1 on human dendritic cells is essential for triggering protective antifungal TH1 and TH17 immune responses. We show that Fonsecaea monophora, a causative agent of chromoblastomycosis, a chronic fungal skin infection, evades these antifungal responses by engaging CLR mincle and suppressing IL-12, which drives TH1 differentiation. Dectin-1 triggering by F. monophora activates transcription factor IRF1, which is crucial for IL12A transcription via nucleosome remodeling. However, simultaneous F. monophora binding to mincle induces an E3 ubiquitin ligase Mdm2-dependent degradation pathway, via Syk-CARD9-mediated PKB signaling, that leads to loss of nuclear IRF1 activity, hence blocking IL12A transcription. The absence of IL-12 leads to impaired TH1 responses and promotes TH2 polarization. Notably, mincle is similarly exploited by other chromoblastomycosis-associated fungi to redirect TH responses. Thus, mincle is a fungal receptor that can suppress antifungal immunity and, as such, is a potential therapeutic target.


Subject(s)
Interleukin-12 Subunit p35/biosynthesis , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , Saccharomycetales/immunology , CARD Signaling Adaptor Proteins/immunology , Cell Differentiation/immunology , Cells, Cultured , Chromoblastomycosis/immunology , Dendritic Cells/immunology , Humans , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-1/genetics , Interleukin-12 Subunit p35/genetics , Interleukin-12 Subunit p35/immunology , Intracellular Signaling Peptides and Proteins/immunology , Protein-Tyrosine Kinases/immunology , Proto-Oncogene Proteins c-mdm2/immunology , RNA Interference , RNA, Small Interfering , Syk Kinase , Th1 Cells/immunology , Th17 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL