75 results on '"Geijtenbeek, TBH"'
Search Results
2. A6.9 Distinct expression of T-cell homing molecules in human autoimmune lymph node stromal cells upon TLR-3 triggering
- Author
-
Hähnlein, J, Ramwadhdoebe, TH, Semmelink, JF, Choi, IY, Smits, NAM, Berger, FH, Maas, M, Gerlag, DM, Geijtenbeek, TBH, Tak, PP, and van Baarsen, LBM
- Published
- 2015
- Full Text
- View/download PDF
3. A1.19 Altered distribution of innate lymphoid cell populations in human LYMPH node biopsies obtained during the earliest phases of systemic autoimmunity
- Author
-
Hähnlein, JS, primary, Rodriguez-Carrio, J, additional, Ramwadhdoebe, TH, additional, Semmelink, JF, additional, Choi, IY, additional, van Lienden, KP, additional, Maas, M, additional, Gerlag, DM, additional, Tak, PP, additional, Geijtenbeek, TBH, additional, and van Baarsen, LGM, additional
- Published
- 2016
- Full Text
- View/download PDF
4. A3.06 Distinct expression pattern of peripheral tissue-restricted antigens in human LYMPH node stromal cells during the earliest phases of rheumatoid arthritis
- Author
-
Hähnlein, J, primary, Ramwadhdoebe, TH, additional, Semmelink, JF, additional, Safy, M, additional, van Lienden, KP, additional, Maas, M, additional, Gerlag, DM, additional, Tak, PP, additional, Geijtenbeek, TBH, additional, and van Baarsen, LGM, additional
- Published
- 2016
- Full Text
- View/download PDF
5. O13.4 Cervicovaginal microbiome dysbiosis is associated with proteome changes related to alterations of the cervicovaginal mucosal barrier
- Author
-
Borgdorff, H, primary, Gautam, R, additional, Armstrong, SD, additional, Xia, D, additional, Ndayisaba, GF, additional, van Teijlingen, NH, additional, Geijtenbeek, TBH, additional, Wastling, JM, additional, and van de Wijgert, JHHM, additional
- Published
- 2015
- Full Text
- View/download PDF
6. Pathogen-recognition receptors as targets for pathogens to modulate immune function of antigen presenting cells
- Author
-
Engering, A., van Vliet, SJ, Koppel, E.A., Geijtenbeek, TBH, van Kooyk, Y, Molecular cell biology and Immunology, AGEM - Digestive immunity, AGEM - Re-generation and cancer of the digestive system, AII - Inflammatory diseases, AII - Cancer immunology, and CCA - Cancer biology and immunology
- Published
- 2006
7. Diminished transmission of drug resistant HIV-1 variants with reduced replication capacity in a human transmission model
- Author
-
Pingen, Marieke, Sarrami-Forooshani, R, Wensing, AMJ, van Ham, P, Drewniak, A, Boucher, Charles, Geijtenbeek, TBH, Nijhuis, M, Pingen, Marieke, Sarrami-Forooshani, R, Wensing, AMJ, van Ham, P, Drewniak, A, Boucher, Charles, Geijtenbeek, TBH, and Nijhuis, M
- Abstract
Background: Different patterns of drug resistance are observed in treated and therapy naive HIV-1 infected populations. Especially the NRTI-related M184I/V variants, which are among the most frequently encountered mutations in treated patients, are underrepresented in the antiretroviral naive population. M184I/V mutations are known to have a profound effect on viral replication and tend to revert over time in the new host. However it is debated whether a diminished transmission efficacy of HIV variants with a reduced replication capacity can also contribute to the observed discrepancy in genotypic patterns. As dendritic cells (DCs) play a pivotal role in HIV-1 transmission, we used a model containing primary human Langerhans cells (LCs) and DCs to compare the transmission efficacy M184 variants (HIV-M184V/I/T) to HIV wild type (HIV-WT). As control, we used HIV harboring the NNRTI mutation K103N (HIV-K103N) which has a minor effect on replication and is found at a similar prevalence in treated and untreated individuals. Results: In comparison to HIV-WT, the HIV-M184 variants were less efficiently transmitted to CCR5(+) Jurkat T cells by both LCs and DCs. The transmission rate of HIV-K103N was slightly reduced to HIV-WT in LCs and even higher than HIV-WT in DCs. Replication experiments in CCR5(+) Jurkat T cells revealed no apparent differences in replication capacity between the mutant viruses and HIV-WT. However, viral replication in LCs and DCs was in concordance with the transmission results; replication by the HIV-M184 variants was lower than replication by HIV-WT, and the level of replication of HIV-K103N was intermediate for LCs and higher than HIV-WT for DCs. Conclusions: Our data demonstrate that drug resistant M184-variants display a reduced replication capacity in LCs and DCs which directly impairs their transmission efficacy. As such, diminished transmission efficacy may contribute to the lower prevalence of drug resistant variants in therapy naive individu
- Published
- 2014
8. A Prominent Role for DC-SIGN(+) Dendritic Cells in Initiation and Dissemination of Measles Virus Infection in Non-Human Primates
- Author
-
Mesman, AW, de Vries, Rory, McQuaid, S, Duprex, WP, de Swart, Rik, Geijtenbeek, TBH, Mesman, AW, de Vries, Rory, McQuaid, S, Duprex, WP, de Swart, Rik, and Geijtenbeek, TBH
- Abstract
Measles virus (MV) is a highly contagious virus that is transmitted by aerosols. During systemic infection, CD150(+) T and B lymphocytes in blood and lymphoid tissues are the main cells infected by pathogenic MV. However, it is unclear which cell types are the primary targets for MV in the lungs and how the virus reaches the lymphoid tissues. In vitro studies have shown that dendritic cell (DC) C-type lectin DC-SIGN captures MV, leading to infection of DCs as well as transmission to lymphocytes. However, evidence of DC-SIGN-mediated transmission in vivo has not been established. Here we identified DC-SIGN hi DCs as first target cells in vivo and demonstrate that macaque DC-SIGN functions as an attachment receptor for MV. Notably, DC-SIGN hi cells from macaque broncho-alveolar lavage and lymph nodes transmit MV to B lymphocytes, providing in vivo support for an important role for DCs in both initiation and dissemination of MV infection. Citation: Mesman AW, de Vries RD, McQuaid S, Duprex WP, de Swart RL, et al. (2012) A Prominent Role for DC-SIGN(+) Dendritic Cells in Initiation and Dissemination of Measles Virus Infection in Non-Human Primates. PLoS ONE 7(12): e49573. doi:10.1371/journal.pone.0049573
- Published
- 2012
9. Early Target Cells of Measles Virus after Aerosol Infection of Non-Human Primates
- Author
-
Lemon, K, de Vries, Rory, Mesman, AW, McQuaid, S, Amerongen, Geert, Yüksel, Selma, Ludlow, Martin, Rennick, LJ, Kuiken, Thijs, Rima, BK, Geijtenbeek, TBH, Osterhaus, Ab, Duprex, WP, de Swart, Rik, Lemon, K, de Vries, Rory, Mesman, AW, McQuaid, S, Amerongen, Geert, Yüksel, Selma, Ludlow, Martin, Rennick, LJ, Kuiken, Thijs, Rima, BK, Geijtenbeek, TBH, Osterhaus, Ab, Duprex, WP, and de Swart, Rik
- Abstract
Measles virus (MV) is highly infectious, and has long been thought to enter the host by infecting epithelial cells of the respiratory tract. However, epithelial cells do not express signaling lymphocyte activation molecule (CD150), which is the high-affinity cellular receptor for wild-type MV strains. We have generated a new recombinant MV strain expressing enhanced green fluorescent protein (EGFP), based on a wild-type genotype B3 virus isolate from Khartoum, Sudan (KS). Cynomolgus macaques were infected with a high dose of rMV(KS) EGFP by aerosol inhalation to ensure that the virus could reach the full range of potential target cells throughout the entire respiratory tract. Animals were euthanized 2, 3, 4 or 5 days post-infection (d.p.i., n = 3 per time point) and infected (EGFP(+)) cells were identified at all four time points, albeit at low levels 2 and 3 d.p.i. At these earliest time points, MV-infected cells were exclusively detected in the lungs by fluorescence microscopy, histopathology and/or virus isolation from broncho-alveolar lavage cells. On 2 d.p.i., EGFP(+) cells were phenotypically typed as large mononuclear cells present in the alveolar lumen or lining the alveolar epithelium. One to two days later, larger clusters of MV-infected cells were detected in bronchus-associated lymphoid tissue (BALT) and in the tracheo-bronchial lymph nodes. From 4 d.p.i. onward, MV-infected cells were detected in peripheral blood and various lymphoid tissues. In spite of the possibility for the aerosolized virus to infect cells and lymphoid tissues of the upper respiratory tract, MV-infected cells were not detected in either the tonsils or the adenoids until after onset of viremia. These data strongly suggest that in our model MV entered the host at the alveolar level by infecting macrophages or dendritic cells, which traffic the virus to BALT or regional lymph nodes, resulting in local amplification and subsequent systemic dissemination by viremia.
- Published
- 2011
10. HSV Neutralization by the Microbicidal Candidate C5A
- Author
-
Witte, Lot, Bobardt, MD, Chatterji, U, Loenen, Freek, Verjans, Georges, Geijtenbeek, TBH, Gallay, PA, Witte, Lot, Bobardt, MD, Chatterji, U, Loenen, Freek, Verjans, Georges, Geijtenbeek, TBH, and Gallay, PA
- Abstract
Genital herpes is a major risk factor in acquiring human immunodeficiency virus type-1 (HIV-1) infection and is caused by both Herpes Simplex virus type 1 (HSV-1) and HSV-2. The amphipathic peptide C5A, derived from the non-structural hepatitis C virus (HCV) protein 5A, was shown to prevent HIV-1 infection but neither influenza nor vesicular stomatitis virus infections. Here we investigated the antiviral function of C5A on HSV infections. C5A efficiently inhibited both HSV-1 and HSV-2 infection in epithelial cells in vitro as well as in an ex vivo epidermal infection model. C5A destabilized the integrity of the viral HSV membrane. Furthermore, drug resistant HSV strains were inhibited by this peptide. Notably, C5A-mediated neutralization of HSV-1 prevented HIV-1 transmission. An in vitro HIV-1 transmigration assay was developed using primary genital epithelial cells and HSV infection increased HIV-1 transmigration. Treatment with C5A abolished HIV-1 transmigration by preventing HSV infection and by preserving the integrity of the genital epithelium that was severely compromised by HSV infection. In conclusion, this study demonstrates that C5A represents a multipurpose microbicide candidate, which neutralizes both HIV-1 and HSV, and which may interfere with HIV-1 transmission through the genital epithelium.
- Published
- 2011
11. The Synthetic Bacterial Lipopeptide Pam3CSK4 Modulates Respiratory Syncytial Virus Infection Independent of TLR Activation
- Author
-
Nguyen, Tien, Witte, Lot, Ludlow, Martin, Yüksel, Selma, Wiesmuller, KH, Geijtenbeek, TBH, Osterhaus, Ab, de Swart, Rik, Nguyen, Tien, Witte, Lot, Ludlow, Martin, Yüksel, Selma, Wiesmuller, KH, Geijtenbeek, TBH, Osterhaus, Ab, and de Swart, Rik
- Abstract
Respiratory syncytial virus (RSV) is an important cause of acute respiratory disease in infants, immunocompromised subjects and the elderly. However, it is unclear why most primary RSV infections are associated with relatively mild symptoms, whereas some result in severe lower respiratory tract infections and bronchiolitis. Since RSV hospitalization has been associated with respiratory bacterial co-infections, we have tested if bacterial Toll-like receptor (TLR) agonists influence RSV-A2-GFP infection in human primary cells or cell lines. The synthetic bacterial lipopeptide Pam3-Cys-Ser-Lys4 (Pam3CSK4), the prototype ligand for the heterodimeric TLR1/TLR2 complex, enhanced RSV infection in primary epithelial, myeloid and lymphoid cells. Surprisingly, enhancement was optimal when lipopeptides and virus were added simultaneously, whereas addition of Pam3CSK4 immediately after infection had no effect. We have identified two structurally related lipopeptides without TLR-signaling capacity that also modulate RSV infection, whereas Pam3CSK4-reminiscent TLR1/2 agonists did not, and conclude that modulation of infection is independent of TLR activation. A similar TLR-independent enhancement of infection could also be demonstrated for wild-type RSV strains, and for HIV-1, measles virus and human metapneumovirus. We show that the effect of Pam3CSK4 is primarily mediated by enhanced binding of RSV to its target cells. The N-palmitoylated cysteine and the cationic lysines were identified as pivotal for enhanced virus binding. Surprisingly, we observed inhibition of RSV infection in immortalized epithelial cell lines, which was shown to be related to interactions between Pam3CSK4 and negatively charged glycosaminoglycans on these cells, which are known targets for binding of laboratory-adapted but not wild-type RSV. These data suggest a potential role for bacterial lipopeptides in enhanced binding of RSV and other viruses to their target cells, thus affecting viral entry or spre
- Published
- 2010
12. DC-SIGN and CD150 have distinct roles in transmission of measles virus from dendritic cells to T-lymphocytes
- Author
-
de Witte, L, de Vries, Rory, van der Vlist, M, Yüksel, Selma, Litjens, M, de Swart, Rik, Geijtenbeek, TBH, de Witte, L, de Vries, Rory, van der Vlist, M, Yüksel, Selma, Litjens, M, de Swart, Rik, and Geijtenbeek, TBH
- Abstract
Measles virus (MV) is among the most infectious viruses that affect humans and is transmitted via the respiratory route. In macaques, MV primarily infects lymphocytes and dendritic cells (DCs). Little is known about the initial target cell for MV infection. Since DCs bridge the peripheral mucosal tissues with lymphoid tissues, we hypothesize that DCs are the initial target cells that capture MV in the respiratory tract and transport the virus to the lymphoid tissues where MV is transmitted to lymphocytes. Recently, we have demonstrated that the C-type lectin DC-SIGN interacts with MV and enhances infection of DCs in cis. Using immunofluorescence microscopy, we demonstrate that DC-SIGN(+) DCs are abundantly present just below the epithelia of the respiratory tract. DC-SIGN(+) DCs efficiently present MV-derived antigens to CD4(+) T-lymphocytes after antigen uptake via either CD150 or DC-SIGN in vitro. However, DC-SIGN(+) DCs also mediate transmission of MV to CD4(+) and CD8(+) T-lymphocytes. We distinguished two different transmission routes that were either dependent or independent on direct DC infection. DC-SIGN and CD150 are both involved in direct DC infection and subsequent transmission of de novo synthesized virus. However, DC-SIGN, but not CD150, mediates trans-infection of MV to T-lymphocytes independent of DC infection. Together these data suggest a prominent role for DCs during the initiation, dissemination, and clearance of MV infection.
- Published
- 2008
13. Transcriptomic HIV-1 reservoir profiling reveals a role for mitochondrial functionality in HIV-1 latency.
- Author
-
Man S, Jansen J, Kroeze S, Geijtenbeek TBH, and Kootstra NA
- Subjects
- Humans, Transcriptome, Gene Expression Profiling methods, In Situ Hybridization, Fluorescence, Adult, Male, Flow Cytometry, HIV-1 physiology, HIV-1 genetics, Virus Latency physiology, HIV Infections virology, Mitochondria metabolism, CD4-Positive T-Lymphocytes virology
- Abstract
Identifying cellular and molecular mechanisms maintaining HIV-1 latency in the viral reservoir is crucial for devising effective cure strategies. Here we developed an innovative flow cytometry-fluorescent in situ hybridization (flow-FISH) approach for direct ex vivo reservoir detection without the need for reactivation using a combination of probes detecting abortive and elongated HIV-1 transcripts. Our flow-FISH assay distinguished between HIV-1-infected CD4+ T cells expressing abortive or elongated HIV-1 transcripts in PBMC from untreated and ART-treated PWH from the Amsterdam Cohort Studies. This flow-FISH method was employed to isolate CD4+ T cells expressing abortive or elongated HIV-1 transcripts from five ART-naïve PWH for transcriptomic analysis by 3' RNA sequencing. Supervised cluster analysis identified several differentially expressed mitochondrial genes in infected CD4+ T cells with abortive HIV-1 transcripts compared to cells containing elongated HIV-1 transcripts. Notably, enhancing mitochondrial function induced HIV-1 transcription in PBMC from PWH. Our data strongly suggests that cellular metabolism is involved in maintaining HIV-1 latency and show that improving mitochondrial functions induces HIV-1 transcriptional activity in PWH. These findings underline the relevance of metabolic regulation in HIV-1 infection, and support the development of strategies modulating immunometabolism to target viral latency., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2025 Man et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2025
- Full Text
- View/download PDF
14. Energy demanding RNA and protein metabolism drive dysfunctionality of HIV-specific T cell changes during chronic HIV infection.
- Author
-
van Pul L, Stunnenberg M, Kroeze S, van Dort KA, Boeser-Nunnink BDM, Harskamp AM, Geijtenbeek TBH, and Kootstra NA
- Subjects
- Humans, Male, Energy Metabolism, Adult, HIV-1 immunology, RNA genetics, RNA metabolism, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing genetics, Female, Chronic Disease, HLA-B Antigens genetics, HLA-B Antigens metabolism, Middle Aged, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, HIV Infections immunology, HIV Infections genetics, HIV Infections virology
- Abstract
Antiretroviral treatment of HIV infected individuals cannot eliminate the HIV reservoir and immune control of HIV is rarely seen upon treatment interruption. In long-term non-progressors (LTNP), an effective CD8 T cell response is thought to contribute to be immune control of HIV. Here we studied the transcriptional profile of virus specific CD8 T cells during the asymptomatic phase of disease, to gain molecular insights in CD8 T cell functionality in HIV progressors and different groups of LTNP: HLA-B*57 LTNP, non-HLA-B*57 LTNP and individuals carrying the MAVS minor genotype (rs7262903/rs7269320). Principal component analysis revealed distinct overall transcriptional profiles between the groups. The transcription profile of HIV-specific CD8 T cells of LTNP groups was associated with increased cytokine/IL-12 signaling and protein/RNA metabolism pathways, indicating an increased CD8 T cell functionality. Although the transcription profile of CMV-specific CD8 T cells differed from that of HIV-specific CD8 T cells, with mainly an upregulation of gene expression in progressors, similar affected pathways were identified. Moreover, CMV-specific CD8 T cells from progressors showed increased expression of genes related to effector functions and suggests recent antigen exposure. Our data shows that changes in cytokine signaling and the energy demanding RNA and protein metabolism are related to CD8 T cell dysfunction, which may indicate that mitochondrial dysfunction is an important driver of T cell dysfunctionality during chronic HIV infection. Indeed, improvement of mitochondrial function by IL-12 and mitoTempo treatment, enhanced in vitro IFNγ release by PBMC from PWH upon HIV gag and CMV pp65 peptide stimulation. Our study provides new insights into the molecular pathways associated with CD8 T cell mediated immune control of chronic HIV infection which is important for the design of novel treatment strategies to restore or improve the HIV-specific immune response., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 van Pul et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
15. Inhibition of HIV-1 replication by nanobodies targeting tetraspanin CD9.
- Author
-
Umotoy JC, Kroon PZ, Man S, van Dort KA, Atabey T, Schriek AI, Dekkers G, Herrera-Carrillo E, Geijtenbeek TBH, Heukers R, Kootstra NA, van Gils MJ, and de Taeye SW
- Abstract
HIV-1 alters the dynamics and distribution of tetraspanins, a group of proteins integral to membrane organization, to facilitate both entry and egress. Notably, the tetraspanin CD9 is dysregulated during HIV-1 infection, correlating with multifaceted effects on viral replication. Here, we generated llama-derived nanobodies against CD9 to restrict HIV-1 replication. We immunized llamas with recombinant large extracellular loop of CD9 and identified eight clonally distinct nanobodies targeting CD9, each exhibiting a range of affinities and differential binding to cell surface-expressed CD9. Notably, nanobodies T2C001 and T2C002 demonstrated low nanomolar affinities and exhibited differential sensitivities against endogenous and overexpressed CD9 on the cell surface. Although CD9-directed nanobodies did not impede the early stages of HIV-1 life cycle, they effectively inhibited virus-induced syncytia formation and virus replication in T cells and monocyte-derived macrophages. This discovery opens new avenues for host-targeted therapeutic strategies, potentially augmenting existing antiretroviral treatments for HIV-1., Competing Interests: The authors declare no competing interests., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
16. Human CD127 negative ILC2s show immunological memory.
- Author
-
Mathä L, Krabbendam L, Martinez Høyer S, Heesters BA, Golebski K, Kradolfer C, Ghaedi M, Ma J, Stadhouders R, Bachert C, Cardell LO, Zhang N, Holtappels G, Reitsma S, Helgers LC, Geijtenbeek TBH, Coquet JM, Takei F, Spits H, and Martinez-Gonzalez I
- Subjects
- Humans, Animals, Mice, Leukocyte Common Antigens metabolism, Cytokines metabolism, Inflammation immunology, Female, Mice, Inbred C57BL, Immunologic Memory immunology, Interleukin-7 Receptor alpha Subunit metabolism, Lymphocytes immunology, Immunity, Innate immunology
- Abstract
ILC2s are key players in type 2 immunity and contribute to maintaining homeostasis. ILC2s are also implicated in the development of type 2 inflammation-mediated chronic disorders like asthma. While memory ILC2s have been identified in mouse, it is unknown whether human ILC2s can acquire immunological memory. Here, we demonstrate the persistence of CD45RO, a marker previously linked to inflammatory ILC2s, in resting ILC2s that have undergone prior activation. A high proportion of these cells concurrently reduce the expression of the canonical ILC marker CD127 in a tissue-specific manner. Upon isolation and in vitro stimulation of CD127-CD45RO+ ILC2s, we observed an augmented ability to proliferate and produce cytokines. CD127-CD45RO+ ILC2s are found in both healthy and inflamed tissues and display a gene signature of cell activation. Similarly, mouse memory ILC2s show reduced expression of CD127. Our findings suggest that human ILC2s can acquire innate immune memory and warrant a revision of the current strategies to identify human ILC2s., (© 2024 Mathä et al.)
- Published
- 2024
- Full Text
- View/download PDF
17. Prevotella timonensis Bacteria Associated With Vaginal Dysbiosis Enhance Human Immunodeficiency Virus Type 1 Susceptibility Of Vaginal CD4+ T Cells.
- Author
-
van Teijlingen NH, van Smoorenburg MY, Sarrami-Forooshani R, Zijlstra-Willems EM, van Hamme JL, Borgdorff H, van de Wijgert JHHM, van Leeuwen E, van der Post JAM, Strijbis K, Ribeiro CMS, and Geijtenbeek TBH
- Subjects
- Humans, Female, Disease Susceptibility, Microbiota, Virus Internalization, Prevotella isolation & purification, Dysbiosis microbiology, Vagina microbiology, Vagina virology, Vagina immunology, CD4-Positive T-Lymphocytes immunology, HIV-1, HIV Infections microbiology, HIV Infections immunology, HIV Infections virology
- Abstract
Dysbiosis of the vaginal microbiome poses a serious risk for sexual human immunodeficiency virus type 1 (HIV-1) transmission. Prevotella spp are abundant during vaginal dysbiosis and associated with enhanced HIV-1 susceptibility; however, underlying mechanisms remain unclear. Here, we investigated the direct effect of vaginal bacteria on HIV-1 susceptibility of vaginal CD4+ T cells. Notably, pre-exposure to Prevotella timonensis enhanced HIV-1 uptake by vaginal T cells, leading to increased viral fusion and enhanced virus production. Pre-exposure to antiretroviral inhibitors abolished P timonensis-enhanced infection. Our study shows that the vaginal microbiome directly affects mucosal CD4+ T-cell susceptibility, emphasizing importance of vaginal dysbiosis diagnosis and treatment., Competing Interests: Potential conflicts of interest. All authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2024. Published by Oxford University Press on behalf of Infectious Diseases Society of America.)
- Published
- 2024
- Full Text
- View/download PDF
18. Retinoic acid-loaded liposomes induce human mucosal CD103 + dendritic cells that inhibit Th17 cells and drive regulatory T-cell development in vitro.
- Author
-
Nagy NA, Hafkamp FMJ, Sparrius R, Bas R, Lozano Vigario F, van Capel TMM, van Ree R, Geijtenbeek TBH, Slütter B, Tas SW, and de Jong EC
- Subjects
- Humans, Immune Tolerance drug effects, Cells, Cultured, Interleukin-10 metabolism, Interleukin-10 immunology, Forkhead Transcription Factors metabolism, Inflammatory Bowel Diseases immunology, Lymphocyte Activation drug effects, Lymphocyte Activation immunology, Celiac Disease immunology, Tretinoin pharmacology, Integrin alpha Chains metabolism, Th17 Cells immunology, Dendritic Cells immunology, Dendritic Cells drug effects, Antigens, CD immunology, Antigens, CD metabolism, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory drug effects, Liposomes, Cell Differentiation drug effects, Cell Differentiation immunology, Retinal Dehydrogenase metabolism, Aldehyde Dehydrogenase 1 Family
- Abstract
The active vitamin A metabolite, all-trans-retinoic acid (RA), primes precursor dendritic cells (DCs) into a mucosal phenotype with tolerogenic properties characterized by the expression of integrin CD103. CD103
+ DCs can counteract pathogenic Th1 and Th17 in inflammatory bowel disease (IBD) or celiac disease (CD). Tolerogenic manipulation of DCs using nanoparticles carrying tolerogenic adjuvants and disease-specific antigens is a valuable treatment strategy to induce antigen-specific mucosal tolerance in vivo. Here, we investigated the effects of RA-loaded liposomes on human DC phenotype and function, including DC-driven T-cell development, both during the generation of monocyte-derived DCs (moDCs) as well as by priming immature moDCs. RA liposomes drove CD103+ DC differentiation as well as ALDH1A2 expression in DCs. Neutrophil-dependent Th17 cell development was reduced by RA-liposome-differentiated and RA-liposome-primed DCs. Moreover, RA liposome treatment shifted T-cell development toward a Th2 cell profile. Importantly, RA liposomes induced the development of IL-10-producing and FoxP3+ regulatory T cells (Tregs) of various Treg subsets, including ICOS+ Tregs, that were potent inhibitors of bystander memory T-cell proliferation. Taken together, RA-loaded liposomes could be a novel treatment avenue for IBD or CD patients., (© 2024 The Authors. European Journal of Immunology published by Wiley‐VCH GmbH.)- Published
- 2024
- Full Text
- View/download PDF
19. Dengue Virus Infects Human Skin Langerhans Cells through Langerin for Dissemination to Dendritic Cells.
- Author
-
Helgers LC, Keijzer NCH, van Hamme JL, Sprokholt JK, and Geijtenbeek TBH
- Subjects
- Humans, Cell Movement, Cells, Cultured, Skin virology, Skin pathology, Epidermis virology, Epidermis pathology, Epidermis metabolism, Langerhans Cells virology, Langerhans Cells immunology, Lectins, C-Type metabolism, Dengue Virus physiology, Mannose-Binding Lectins metabolism, Dendritic Cells virology, Dendritic Cells immunology, Antigens, CD metabolism, Dengue virology, Dengue immunology
- Abstract
Dengue virus (DENV) is the most disease-causative flavivirus worldwide. DENV as a mosquito-borne virus infects human hosts through the skin; however, the initial target cells in the skin remain unclear. In this study, we have investigated whether epidermal Langerhans cells (LCs) play a role in DENV acquisition and dissemination. We have used a human epidermal ex vivo infection model as well as isolated LCs to investigate infection by DENV. Notably, both immature and mature LCs were permissive to DENV infection in vitro and ex vivo, and infection was dependent on C-type lectin receptor langerin because blocking antibodies against langerin significantly reduced DENV infection in vitro and ex vivo. DENV-infected LCs efficiently transmitted DENV to target cells such as dendritic cells. Moreover, DENV exposure increased the migration of LCs from epidermal explants. These results strongly suggest that DENV targets epidermal LCs for infection and dissemination in the human host. These findings could provide potential drug targets to combat the early stage of DENV infection., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. Plant-produced Der p 2-bearing bioparticles activate Th1/Treg-related activation patterns in dendritic cells irrespective of the allergic background.
- Author
-
Busold S, Aglas L, Menage C, Desgagnés R, Faye L, Fitchette AC, de Jong EC, Martel C, Stigler M, Catala-Stordeur V, Tropper G, Auger L, Morel B, Versteeg SA, Vézina LP, Gomord V, Layhadi JA, Shamji M, Geijtenbeek TBH, and van Ree R
- Subjects
- Humans, Antigens, Dermatophagoides, Arthropod Proteins, Dendritic Cells, Allergens, Cytokines, T-Lymphocytes, Regulatory, Hypersensitivity
- Published
- 2024
- Full Text
- View/download PDF
21. Noncanonical-NF-κB activation and DDX3 inhibition reduces the HIV-1 reservoir by elimination of latently infected cells ex-vivo .
- Author
-
Jansen J, Kroeze S, Man S, Andreini M, Bakker J-W, Zamperini C, Tarditi A, Kootstra NA, and Geijtenbeek TBH
- Subjects
- Humans, NF-kappa B metabolism, CD4-Positive T-Lymphocytes, Gene Expression Regulation, Virus Latency, HIV-1, HIV Infections drug therapy, HIV Infections genetics
- Abstract
Importance: HIV-1 continues to be a major global health challenge. Current HIV-1 treatments are effective but need lifelong adherence. An HIV-1 cure should eliminate the latent viral reservoir that persists in people living with HIV-1. Different methods have been investigated that focus on reactivation and subsequent elimination of the HIV-1 reservoir, and it is becoming clear that a combination of compounds with different mechanisms of actions might be more effective. Here, we target two host factors, inhibitor of apoptosis proteins that control apoptosis and the DEAD-box helicase DDX3, facilitating HIV mRNA transport/translation. We show that targeting of these host factors with SMAC mimetics and DDX3 inhibitors induce reversal of viral latency and eliminate HIV-1-infected cells in vitro and ex vivo ., Competing Interests: M.A., J.B., C.Z., and A.T. are employees of First Health Pharmaceuticals B.V.
- Published
- 2024
- Full Text
- View/download PDF
22. SARS-CoV-2 suppresses TLR4-induced immunity by dendritic cells via C-type lectin receptor DC-SIGN.
- Author
-
van der Donk LEH, Bermejo-Jambrina M, van Hamme JL, Volkers MMW, van Nuenen AC, Kootstra NA, and Geijtenbeek TBH
- Subjects
- Humans, SARS-CoV-2 metabolism, Toll-Like Receptor 4 metabolism, Lipopolysaccharides pharmacology, Lipopolysaccharides metabolism, Lectins, C-Type metabolism, Cytokines metabolism, Dendritic Cells, Superinfection, COVID-19 metabolism
- Abstract
SARS-CoV-2 causes COVID-19, an infectious disease with symptoms ranging from a mild cold to severe pneumonia, inflammation, and even death. Although strong inflammatory responses are a major factor in causing morbidity and mortality, superinfections with bacteria during severe COVID-19 often cause pneumonia, bacteremia and sepsis. Aberrant immune responses might underlie increased sensitivity to bacteria during COVID-19 but the mechanisms remain unclear. Here we investigated whether SARS-CoV-2 directly suppresses immune responses to bacteria. We studied the functionality of human dendritic cells (DCs) towards a variety of bacterial triggers after exposure to SARS-CoV-2 Spike (S) protein and SARS-CoV-2 primary isolate (hCoV-19/Italy). Notably, pre-exposure of DCs to either SARS-CoV-2 S protein or a SARS-CoV-2 isolate led to reduced type I interferon (IFN) and cytokine responses in response to Toll-like receptor (TLR)4 agonist lipopolysaccharide (LPS), whereas other TLR agonists were not affected. SARS-CoV-2 S protein interacted with the C-type lectin receptor DC-SIGN and, notably, blocking DC-SIGN with antibodies restored type I IFN and cytokine responses to LPS. Moreover, blocking the kinase Raf-1 by a small molecule inhibitor restored immune responses to LPS. These results suggest that SARS-CoV-2 modulates DC function upon TLR4 triggering via DC-SIGN-induced Raf-1 pathway. These data imply that SARS-CoV-2 actively suppresses DC function via DC-SIGN, which might account for the higher mortality rates observed in patients with COVID-19 and bacterial superinfections., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 van der Donk et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
23. Toll-like receptor 4 and Syk kinase shape dendritic cell-induced immune activation to major house dust mite allergens.
- Author
-
Busold S, Akkerdaas JH, Zijlstra-Willems EM, van der Graaf K, Tas SW, de Jong EC, van Ree R, and Geijtenbeek TBH
- Abstract
Background: House dust mite (HDM) is a major cause of respiratory allergic diseases. Dendritic cells (DCs) play a central role in orchestrating adaptive allergic immune responses. However, it remains unclear how DCs become activated by HDM. Biochemical functions of the major HDM allergens Der p 1 (cysteine protease) and Der p 2 (MD2-mimick) have been implicated to contribute to DC activation., Methods: We investigated the immune activating potential of HDM extract and its major allergens Der p 1 and Der p 2 using monocyte-derived DCs (moDCs). Maturation and activation markers were monitored by flow cytometry and cytokine production by ELISA. Allergen depletion and proteinase K digestion were used to investigate the involvement of proteins, and in particular of the major allergens. Inhibitors of spleen tyrosine kinase (Syk), Toll-like receptor 4 (TLR4) and of C-type lectin receptors (CLRs) were used to identify the involved receptors. The contribution of endotoxins in moDC activation was assessed by their removal from HDM extract., Results: HDM extract induced DC maturation and cytokine responses in contrast to the natural purified major allergens Der p 1 and Der p 2. Proteinase K digestion and removal of Der p 1 or Der p 2 did not alter the immune stimulatory capacity of HDM extract. Antibodies against the CLRs Dectin-1, Dectin-2, and DC-SIGN did not affect cytokine responses. In contrast, Syk inhibition partially reduced IL-6, IL-12 and completely blocked IL-10. Blocking TLR4 signaling reduced the HDM-induced IL-10 and IL-12p70 induction, but not IL-6, while endotoxin removal potently abolished the induced cytokine response., Conclusion: Our data strongly suggest that HDM-induced DC activation is neither dependent on Der p 1 nor Der p 2, but depend on Syk and TLR4 activation, which might suggest a crosstalk between Syk and TLR4 pathways. Our data highlight that endotoxins play a potent role in immune responses targeting HDM., Competing Interests: RR received consulting fees and/or speaker’s fees from Angany Inc., HAL Allergy BV, and Citeq BV, ThermoFisher Scientific Reacta Healthcare Ltd., Mission MightyMe, AB Enzymes and ALK Abello. KG is the CEO/owner of Citeq Biologics. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Busold, Akkerdaas, Zijlstra-Willems, van der Graaf, Tas, de Jong, van Ree and Geijtenbeek.)
- Published
- 2023
- Full Text
- View/download PDF
24. Anti-cancer effect of COVID-19 vaccines in mice models.
- Author
-
Deldadeh N, Haghighat S, Omidi Z, Sarrami-Foroushani R, Ansari AM, Sanati H, Azizi A, Zayeri F, Forouzesh F, Geijtenbeek TBH, and Javidi MA
- Subjects
- Humans, Female, Animals, Mice, COVID-19 Vaccines, CD4-CD8 Ratio, Biomarkers, Tumor, Vaccination, COVID-19 prevention & control, Neoplasms
- Abstract
Aims: Without any doubt, vaccination was the best choice for Coronavirus disease 2019 (COVID-19) pandemic control. According to the American Society of Clinical Oncology (ASCO) and European Society for Medical Oncology (ESMO), people with cancer or a history of cancer have a higher risk of dying from Covid-19 than ordinary people; hence, they should be considered a high-priority group for vaccination. On the other hand, the effect of the Covid-19 vaccination on cancer is not transparent enough. This study is one of the first in vivo studies that try to show the impact of Sinopharm (S) and AstraZeneca (A) vaccines on breast cancer, the most common cancer among women worldwide., Materials and Methods: Vaccination was performed with one and two doses of Sinopharm (S1/S2) or AstraZeneca (A1/A2) on the 4T1 triple-negative breast cancer (TNBC) mice model. The tumor size and body weight of mice were monitored every two days. After one month, mice were euthanized, and the existence of Tumor-infiltrating lymphocytes (TILs) and expression of the important markers in the tumor site was assessed. Metastasis in the vital organs was also investigated., Key Findings: Strikingly, all of the vaccinated mice showed a decrease in tumor size and this decrease was highest after two vaccinations. Moreover, we observed more TILs in the tumor after vaccination. Vaccinated mice demonstrated a decrease in the expression of tumor markers (VEGF, Ki-67, MMP-2/9), CD4/CD8 ratio, and metastasis to the vital organs., Significance: Our results strongly suggest that COVID-19 vaccinations decrease tumor growth and metastasis., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
25. Author Correction: Low energy nebulization preserves integrity of SARS-CoV-2 mRNA vaccines for respiratory delivery.
- Author
-
van Rijn CJM, Vlaming KE, Bem RA, Dekker RJ, Poortinga A, Breit T, van Leeuwen S, Ensink WA, van Wijnbergen K, van Hamme JL, Bonn D, and Geijtenbeek TBH
- Published
- 2023
- Full Text
- View/download PDF
26. Liposomes loaded with vitamin D3 induce regulatory circuits in human dendritic cells.
- Author
-
Nagy NA, Lozano Vigario F, Sparrius R, van Capel TMM, van Ree R, Tas SW, de Vries IJM, Geijtenbeek TBH, Slütter B, and de Jong EC
- Subjects
- Humans, Dendritic Cells, Immune Tolerance, Skin, Cholecalciferol pharmacology, Liposomes
- Abstract
Introduction: Nanomedicine provides a promising platform for manipulating dendritic cells (DCs) and the ensuing adaptive immune response. For the induction of regulatory responses, DCs can be targeted in vivo with nanoparticles incorporating tolerogenic adjuvants and auto-antigens or allergens., Methods: Here, we investigated the tolerogenic effect of different liposome formulations loaded with vitamin D3 (VD3). We extensively phenotyped monocyte-derived DCs (moDCs) and skin DCs and assessed DC-induced regulatory CD4+ T cells in coculture., Results: Liposomal VD3 primed-moDCs induced the development of regulatory CD4+ T cells (Tregs) that inhibited bystander memory T cell proliferation. Induced Tregs were of the FoxP3+ CD127low phenotype, also expressing TIGIT. Additionally, liposome-VD3 primed moDCs inhibited the development of T helper 1 (Th1) and T helper 17 (Th17) cells. Skin injection of VD3 liposomes selectively stimulated the migration of CD14+ skin DCs., Discussion: These results suggest that nanoparticulate VD3 is a tolerogenic tool for DC-mediated induction of regulatory T cell responses., Competing Interests: Authors BS and EdJ: Payment to institute by Health Holland and Samenwerkende Gezondheidsorganisaties SGF Grantnr: LSHM18065-SGF. Author RvR: Payment to institute by Health Holland and Samenwerkende Gezondheidsorganisaties SGF Grantnr: LSHM18065-SGF, Payment to institute by Health Holland − TKI-LSH PPP Allowance − Grant nr. LSHM19073, European Commission, NWO-TKI, AB Enzymes, Angany Inc., payment to self consulting fees received from HAL Allergy, Citeq BV, Angany Inc, Reacta Healthcare, and Mission MightyMe, payment to self for lectures, speakers bureaus, or educational events by HAL Allergy BV, ALK and ThermoFisher Scientific. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Nagy, Lozano Vigario, Sparrius, van Capel, van Ree, Tas, de Vries, Geijtenbeek, Slütter, de Jong and for the DC4Balance consortium.)
- Published
- 2023
- Full Text
- View/download PDF
27. Low energy nebulization preserves integrity of SARS-CoV-2 mRNA vaccines for respiratory delivery.
- Author
-
van Rijn CJM, Vlaming KE, Bem RA, Dekker RJ, Poortinga A, Breit T, van Leeuwen S, Ensink WA, van Wijnbergen K, van Hamme JL, Bonn D, and Geijtenbeek TBH
- Subjects
- Humans, COVID-19 Vaccines, SARS-CoV-2 genetics, RNA, Messenger genetics, mRNA Vaccines, COVID-19 prevention & control, Nanoparticles
- Abstract
Nebulization of mRNA therapeutics can be used to directly target the respiratory tract. A promising prospect is that mucosal administration of lipid nanoparticle (LNP)-based mRNA vaccines may lead to a more efficient protection against respiratory viruses. However, the nebulization process can rupture the LNP vehicles and degrade the mRNA molecules inside. Here we present a novel nebulization method able to preserve substantially the integrity of vaccines, as tested with two SARS-CoV-2 mRNA vaccines. We compare the new method with well-known nebulization methods used for medical respiratory applications. We find that a lower energy level in generating LNP droplets using the new nebulization method helps safeguard the integrity of the LNP and vaccine. By comparing nebulization techniques with different energy dissipation levels we find that LNPs and mRNAs can be kept largely intact if the energy dissipation remains below a threshold value, for LNP integrity 5-10 J/g and for mRNA integrity 10-20 J/g for both vaccines., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
28. Ectopic expression of cGAS in Salmonella typhimurium enhances STING-mediated IFN-β response in human macrophages and dendritic cells.
- Author
-
Waanders L, van der Donk LEH, Ates LS, Maaskant J, van Hamme JL, Eldering E, van Bruggen JAC, Rietveld JM, Bitter W, Geijtenbeek TBH, and Kuijl CP
- Subjects
- Humans, Salmonella typhimurium metabolism, Ectopic Gene Expression, Nucleotidyltransferases genetics, Nucleotidyltransferases metabolism, Macrophages metabolism, Dendritic Cells metabolism, Tumor Microenvironment, Interferon Type I, Neoplasms metabolism
- Abstract
Background: Interferon (IFN)-β induction via activation of the stimulator of interferon genes (STING) pathway has shown promising results in tumor models. STING is activated by cyclic dinucleotides such as cyclic GMP-AMP dinucleotides with phosphodiester linkages 2'-5' and 3'-5' (cGAMPs), that are produced by cyclic GMP-AMP synthetase (cGAS). However, delivery of STING pathway agonists to the tumor site is a challenge. Bacterial vaccine strains have the ability to specifically colonize hypoxic tumor tissues and could therefore be modified to overcome this challenge. Combining high STING-mediated IFN-β levels with the immunostimulatory properties of Salmonella typhimurium could have potential to overcome the immune suppressive tumor microenvironment., Methods: We have engineered S. typhimurium to produce cGAMP by expression of cGAS. The ability of cGAMP to induce IFN-β and its IFN-stimulating genes was addressed in infection assays of THP-I macrophages and human primary dendritic cells (DCs). Expression of catalytically inactive cGAS is used as a control. DC maturation and cytotoxic T-cell cytokine and cytotoxicity assays were conducted to assess the potential antitumor response in vitro. Finally, by making use of different S. typhimurium type III secretion (T3S) mutants, the mode of cGAMP transport was elucidated., Results: Expression of cGAS in S. typhimurium results in a 87-fold stronger IFN-β response in THP-I macrophages. This effect was mediated by cGAMP production and is STING dependent. Interestingly, the needle-like structure of the T3S system was necessary for IFN-β induction in epithelial cells. DC activation included upregulation of maturation markers and induction of type I IFN response. Coculture of challenged DCs with cytotoxic T cells revealed an improved cGAMP-mediated IFN-γ response. In addition, coculture of cytotoxic T cells with challenged DCs led to improved immune-mediated tumor B-cell killing., Conclusion: S. typhimurium can be engineered to produce cGAMPs that activate the STING pathway in vitro. Furthermore, they enhanced the cytotoxic T-cell response by improving IFN-γ release and tumor cell killing. Thus, the immune response triggered by S. typhimurium can be enhanced by ectopic cGAS expression. These data show the potential of S. typhimurium -cGAS in vitro and provides rationale for further research in vivo., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
29. Immune activation of vaginal human Langerhans cells increases susceptibility to HIV-1 infection.
- Author
-
van Teijlingen NH, Eder J, Sarrami-Forooshani R, Zijlstra-Willems EM, Roovers JWR, van Leeuwen E, Ribeiro CMS, and Geijtenbeek TBH
- Subjects
- Humans, Female, Langerhans Cells, Ligands, HIV-1 physiology, HIV Infections, Sexually Transmitted Diseases, HIV Seropositivity
- Abstract
Vaginal inflammation increases the risk for sexual HIV-1 transmission but underlying mechanisms remain unclear. In this study we assessed the impact of immune activation on HIV-1 susceptibility of primary human vaginal Langerhans cells (LCs). Vaginal LCs isolated from human vaginal tissue expressed a broad range of TLRs and became activated after exposure to both viral and bacterial TLR ligands. HIV-1 replication was restricted in immature vaginal LCs as only low levels of infection could be detected. Notably, activation of immature vaginal LCs by bacterial TLR ligands increased HIV-1 infection, whereas viral TLR ligands were unable to induce HIV-1 replication in vaginal LCs. Furthermore, mature vaginal LCs transmitted HIV-1 to CD4 T cells. This study emphasizes the role for vaginal LCs in protection against mucosal HIV-1 infection, which is abrogated upon activation. Moreover, our data suggest that bacterial STIs can increase the risk of HIV-1 acquisition in women., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
30. CXCR5 + PD-1 ++ CD4 + T cells colonize infant intestines early in life and promote B cell maturation.
- Author
-
Jordan-Paiz A, Martrus G, Steinert FL, Kaufmann M, Sagebiel AF, Schreurs RRCE, Rechtien A, Baumdick ME, Jung JM, Möller KJ, Wegner L, Grüttner C, Richert L, Thünauer R, Schroeder-Schwarz J, van Goudoever JB, Geijtenbeek TBH, Altfeld M, Pals ST, Perez D, Klarenbeek PL, Tomuschat C, Sauter G, Königs I, Schumacher U, Friese MA, Melling N, Reinshagen K, and Bunders MJ
- Subjects
- Adult, Child, Humans, Infant, B-Lymphocytes, Receptors, CXCR5, Programmed Cell Death 1 Receptor, T-Lymphocytes, Helper-Inducer, CD4-Positive T-Lymphocytes immunology
- Abstract
Gastrointestinal infections are a major cause for serious clinical complications in infants. The induction of antibody responses by B cells is critical for protective immunity against infections and requires CXCR5
+ PD-1++ CD4+ T cells (TFH cells). We investigated the ontogeny of CXCR5+ PD-1++ CD4+ T cells in human intestines. While CXCR5+ PD-1++ CD4+ T cells were absent in fetal intestines, CXCR5+ PD-1++ CD4+ T cells increased after birth and were abundant in infant intestines, resulting in significant higher numbers compared to adults. These findings were supported by scRNAseq analyses, showing increased frequencies of CD4+ T cells with a TFH gene signature in infant intestines compared to blood. Co-cultures of autologous infant intestinal CXCR5+ PD-1+/- CD4+ T cells with B cells further demonstrated that infant intestinal TFH cells were able to effectively promote class switching and antibody production by B cells. Taken together, we demonstrate that functional TFH cells are numerous in infant intestines, making them a promising target for oral pediatric vaccine strategies., (© 2022. The Author(s), under exclusive licence to CSI and USTC.)- Published
- 2023
- Full Text
- View/download PDF
31. Human Dendritic Cells Transmit Enterovirus A71 via Heparan Sulfates to Target Cells Independent of Viral Replication.
- Author
-
Helgers LC, Bhoekhan MS, Pajkrt D, Wolthers KC, Geijtenbeek TBH, and Sridhar A
- Subjects
- Humans, Antigens, Viral metabolism, Dendritic Cells, Heparin metabolism, Heparitin Sulfate metabolism, Sulfates metabolism, Virus Replication physiology, Enterovirus, Enterovirus A, Human genetics, Enterovirus Infections
- Abstract
Enterovirus A71 (EV-A71) is a causative agent of life-threatening neurological diseases in young children. EV-A71 is highly infectious but it remains unclear how the virus disseminates from primary entry sites-the mucosa of the respiratory tract or the intestine-to secondary replication sites-skin or brain. Here, we investigated the role of dendritic cells (DCs) in EV-A71 dissemination. DCs reside in the mucosa of the airway and gut, and migrate to lymphoid tissues upon activation and, therefore, could facilitate EV-A71 dissemination to secondary replication sites. Monocyte-derived DCs were not permissive to different genotypes of EV-A71 but, notably, coculture with EV-A71-susceptiblle RD99 cells led to very efficient infection of RD99 cells. Notably, EV-A71 transmission of DCs to RD99 was independent of viral replication as a replication inhibitor did not affect transmission. Soluble heparin blocked EV-A71 transmission by DCs to RD99 cells, in contrast to antibodies against known attachment receptor DC-SIGN. These results strongly suggest that DCs might be a first target for EV-A71 and involved in viral dissemination via heparan sulfates and heparin derivatives might be an effective treatment to attenuate dissemination. IMPORTANCE EV-A71 is an emerging neurotropic virus that is of emerging concern and can result in polio-like illness. The exact mechanism of how EV-A71 results in neurological symptoms are unknown. In particular, the early dissemination of the virus from primary replication sites (airway and intestine) to secondary sites (central nervous system and skin) needs to be elucidated. There is evidence pointing toward a role for dendritic cells (DC) in EV-A71 transmission. Moreover, heparan sulfate (HS) binding mutations are observed in patients with severe diseases. Therefore, we evaluated the potential role of HS on DC in transmission. We find that HS are critical for transmitting EV-A71 by DC to target cells. Our data are consistent with other clinical and in vitro observations highlighting the importance of HS in EV-A71-induced disease.
- Published
- 2022
- Full Text
- View/download PDF
32. Inhalation of Low Molecular Weight Heparins as Prophylaxis against SARS-CoV-2.
- Author
-
Eder J, Bermejo-Jambrina M, Vlaming KE, Kaptein TM, Zaderer V, Kemper EM, Wilflingseder D, Reitsma S, de Bree GJ, Cohn DM, and Geijtenbeek TBH
- Subjects
- Humans, SARS-CoV-2, Enoxaparin therapeutic use, Heparin, Low-Molecular-Weight adverse effects, COVID-19
- 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.
- Published
- 2022
- Full Text
- View/download PDF
33. Fungal sensing by dectin-1 directs the non-pathogenic polarization of T H 17 cells through balanced type I IFN responses in human DCs.
- Author
-
Gringhuis SI, Kaptein TM, Remmerswaal EBM, Drewniak A, Wevers BA, Theelen B, D'Haens GRAM, Boekhout T, and Geijtenbeek TBH
- Subjects
- Humans, Cytokines metabolism, Lectins, C-Type genetics, Lectins, C-Type metabolism, Th17 Cells metabolism, Transforming Growth Factor beta metabolism, Dendritic Cells metabolism, Interferon Type I metabolism, Mycoses immunology
- Abstract
The non-pathogenic T
H 17 subset of helper T cells clears fungal infections, whereas pathogenic TH 17 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 TH 17 cells. Dectin-1 signaling triggered transient and intermediate expression of interferon (IFN)-β in DCs, which was mediated by the opposed activities of transcription factors IRF1 and IRF5. IFN-β-induced signaling led to integrin αvβ8 expression directly and to the release of the active form of the cytokine transforming growth factor (TGF)-β indirectly. Uncontrolled IFN-β responses as a result of IRF1 deficiency induced high expression of the IFN-stimulated gene BST2 in DCs and restrained TGF-β activation. Active TGF-β was required for polarization of non-pathogenic TH 17 cells, whereas pathogenic TH 17 cells developed in the absence of active TGF-β. Thus, dectin-1-mediated modulation of type I IFN responses allowed TGF-β activation and non-pathogenic TH 17 cell development during fungal infections in humans., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
34. Corrigendum: Anti-HIV-1 nanobody-IgG1 constructs with improved neutralization potency and the ability to mediate Fc effector functions.
- Author
-
Schriek AI, van Haaren MM, Poniman M, Dekkers G, Bentlage AEH, Grobben M, Vidarsson G, Sanders RW, Verrips T, Geijtenbeek TBH, Heukers R, Kootstra NA, de Taeye SW, and van Gils MJ
- Abstract
[This corrects the article DOI: 10.3389/fimmu.2022.893648.]., (Copyright © 2022 Schriek, van Haaren, Poniman, Dekkers, Bentlage, Grobben, Vidarsson, Sanders, Verrips, Geijtenbeek, Heukers, Kootstra, de Taeye and van Gils.)
- Published
- 2022
- Full Text
- View/download PDF
35. Fel d 1 surface expression on plant-made eBioparticles combines potent immune activation and hypoallergenicity.
- Author
-
Busold S, Aglas L, Menage C, Auger L, Desgagnés R, Faye L, Fitchette AC, de Jong EC, Martel C, Stigler M, Catala-Stordeur V, Tropper G, Vézina LP, Gomord V, Geijtenbeek TBH, and van Ree R
- Subjects
- Humans, Allergens, Immunoglobulin E metabolism
- Published
- 2022
- Full Text
- View/download PDF
36. Crosstalk between R848 and abortive HIV-1 RNA-induced signaling enhances antiviral immunity.
- Author
-
Stunnenberg M, van Hamme JL, Zijlstra-Willems EM, Gringhuis SI, and Geijtenbeek TBH
- Subjects
- Adjuvants, Immunologic, CD8-Positive T-Lymphocytes, Dendritic Cells, Immunity, Innate, RNA, Receptors, Pattern Recognition, HIV-1 physiology
- Abstract
Pathogens trigger multiple pattern recognition receptors (PRRs) that together dictate innate and adaptive immune responses. Understanding the crosstalk between PRRs is important to enhance vaccine efficacy. Abortive HIV-1 RNA transcripts are produced during acute and chronic HIV-1 infection and are known ligands for different PRRs, leading to antiviral and proinflammatory responses. Here, we have investigated the crosstalk between responses induced by these 58 nucleotide-long HIV-1 RNA transcripts and different TLR ligands. Costimulation of dendritic cells (DCs) with abortive HIV-1 RNA and TLR7/8 agonist R848, but not other TLR agonists, resulted in enhanced antiviral type I IFN responses as well as adaptive immune responses via the induction of DC-mediated T helper 1 (T
H 1) responses and IFNγ+ CD8+ T cells. Our data underscore the importance of crosstalk between abortive HIV-1 RNA and R848-induced signaling for the induction of effective antiviral immunity., (© 2022 The Authors. Journal of Leukocyte Biology published by Wiley Periodicals LLC on behalf of Society for Leukocyte Biology.)- Published
- 2022
- Full Text
- View/download PDF
37. The Long Non-Coding Antisense RNA JHDM1D-AS1 Regulates Inflammatory Responses in Human Monocytes.
- Author
-
Malmström E, Khan HN, Veer CV', Stunnenberg M, Meijer MT, Matsumoto H, Otto NA, Geijtenbeek TBH, de Vos AF, van der Poll T, and Scicluna BP
- Subjects
- Humans, Lipopolysaccharides metabolism, Macrophages metabolism, Monocytes, RNA, Antisense metabolism, RNA, Long Noncoding metabolism
- Abstract
Monocytes are key players in innate immunity, with their ability to regulate inflammatory responses and combat invading pathogens. There is a growing body of evidence indicating that long non-coding RNA (lncRNA) participate in various cellular biological processes, including the innate immune response. The immunoregulatory properties of numerous lncRNAs discovered in monocytes remain largely unexplored. Here, by RNA sequencing, we identified a lncRNA JHDM1D-AS1, which was upregulated in blood monocytes obtained from patients with sepsis relative to healthy controls. JHDM1D-AS1 expression was induced in primary human monocytes exposed to Toll-like receptor ligands, such as lipopolysaccharide (LPS), or bacteria. The inducibility of JHDM1D-AS1 expression in monocytes depended, at least in part, on nuclear factor-κB activation. JHDM1D-AS1 knockdown experiments in human monocyte-derived macrophages revealed significantly enhanced expression of inflammatory mediators, before and after exposure to LPS, relative to control cells. Specifically, genes involved in inflammatory responses were upregulated (e.g., CXCL2 , CXCL8 , IL1RN , TREM1 , TNF , and IL6 ), whereas genes involved in anti-inflammatory pathways were downregulated (e.g., SOCS1 and IL10RA ). JHDM1D-AS1 overexpression in a pro-monocytic cell line revealed diminished pro-inflammatory responses subsequent to LPS challenge. Collectively, these findings identify JHDM1D-AS1 as a potential anti-inflammatory mediator induced in response to inflammatory stimuli., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Malmström, Khan, Veer, Stunnenberg, Meijer, Matsumoto, Otto, Geijtenbeek, de Vos, Poll and Scicluna.)
- Published
- 2022
- Full Text
- View/download PDF
38. DDX3X structural analysis: Implications in the pharmacology and innate immunity.
- Author
-
De Colibus L, Stunnenberg M, and Geijtenbeek TBH
- Abstract
The human DEAD-Box Helicase 3 X-Linked (DDX3X) is an ATP-dependent RNA helicase involved in virtually every step of RNA metabolism, ranging from transcription regulation in the nucleus to translation initiation and stress granule (SG) formation, and plays crucial roles in innate immunity, as well as tumorigenesis and viral infections. This review discusses latest advances in DDX3X biology and structure-function relationship, including the implications of the recent DDX3X crystal structure in complex with double stranded RNA for RNA metabolism, DDX3X involvement in the cross-talk between innate immune responses and cell stress adaptation, and the roles of DDX3X in controlling cell fate., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
39. Anti-HIV-1 Nanobody-IgG1 Constructs With Improved Neutralization Potency and the Ability to Mediate Fc Effector Functions.
- Author
-
Schriek AI, van Haaren MM, Poniman M, Dekkers G, Bentlage AEH, Grobben M, Vidarsson G, Sanders RW, Verrips T, Geijtenbeek TBH, Heukers R, Kootstra NA, de Taeye SW, and van Gils MJ
- Subjects
- Antibodies, Neutralizing pharmacology, Broadly Neutralizing Antibodies, HIV Antibodies, Humans, Immunoglobulin G, HIV Seropositivity, HIV-1, Single-Domain Antibodies pharmacology
- Abstract
The most effective treatment for HIV-1, antiretroviral therapy, suppresses viral replication and averts the disease from progression. Nonetheless, there is a need for alternative treatments as it requires daily administration with the possibility of side effects and occurrence of drug resistance. Broadly neutralizing antibodies or nanobodies targeting the HIV-1 envelope glycoprotein are explored as alternative treatment, since they mediate viral suppression and contribute to the elimination of virus-infected cells. Besides neutralization potency and breadth, Fc-mediated effector functions of bNAbs also contribute to the in vivo efficacy. In this study multivalent J3, 2E7 and 1F10 anti-HIV-1 broadly neutralizing nanobodies were generated to improve neutralization potency and IgG1 Fc fusion was utilized to gain Fc-mediated effector functions. Bivalent and trivalent nanobodies, coupled using long glycine-serine linkers, showed increased binding to the HIV-1 Env and enhanced neutralization potency compared to the monovalent variant. Fusion of an IgG1 Fc domain to J3 improved neutralization potency compared to the J3-bihead and restored Fc-mediated effector functions such as antibody-dependent cellular phagocytosis and trogocytosis, and natural killer cell activation. Due to their neutralization breadth and potency and their ability to induce effector functions these nanobody-IgG1 constructs may prove to be valuable towards alternative HIV-1 therapies., Competing Interests: Authors GD and RH were employed by QVQ Holding BV. Author TV was employed by VerLin BV. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Schriek, van Haaren, Poniman, Dekkers, Bentlage, Grobben, Vidarsson, Sanders, Verrips, Geijtenbeek, Heukers, Kootstra, de Taeye and van Gils.)
- Published
- 2022
- Full Text
- View/download PDF
40. Uptake Kinetics Of Liposomal Formulations of Differing Charge Influences Development of in Vivo Dendritic Cell Immunotherapy.
- Author
-
Nagy NA, Castenmiller C, Vigario FL, Sparrius R, van Capel TMM, de Haas AM, van Kooyk Y, van Ree R, Tas SW, Geijtenbeek TBH, Jiskoot W, Slütter B, and de Jong EC
- Subjects
- Immunologic Factors, Immunotherapy methods, Kinetics, Dendritic Cells, Liposomes
- Abstract
Dendritic cells (DCs) control adaptive immunity and are therefore attractive for in vivo targeting to either induce immune activation or tolerance, depending on disease. Liposomes, nanoparticles comprised of a lipid bi-layer, provide a nanoplatform for loading disease-relevant antigen, adjuvant and DC-targeting molecules simultaneously. However, it is yet not fully understood how liposomal formulations affect uptake by DCs and DC function. Here, we examined monocyte-derived DC (moDC) and skin DC uptake of six different liposomal formulations, together with their DC-modulating effect. Contrary to literature, we show using imaging flow cytometry that anionic or neutral liposomes are taken up more efficiently than cationic liposomes by moDCs, or by skin DCs after intradermal injection. None of the formulations yielded significant modulation of DC function as determined by the upregulation of maturation markers and cytokine production. These results suggest that anionic liposomes would be more suitable as vaccine carriers for a dermal application., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Payment to institute by Health Holland and Samenwerkende Gezondheidsorganisaties (SGF) Grantnr: LSHM18065-SGF. Payment to institute by Health Holland – TKI-LSH PPP Allowance – Grant nr. LSHM19073, European Commission, NWO-TKI, AB Enzymes, Angany Inc., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
41. SARS-CoV-2 infection activates dendritic cells via cytosolic receptors rather than extracellular TLRs.
- Author
-
van der Donk LEH, Eder J, van Hamme JL, Brouwer PJM, Brinkkemper M, van Nuenen AC, van Gils MJ, Sanders RW, Kootstra NA, Bermejo-Jambrina M, and Geijtenbeek TBH
- Subjects
- Cell Line, Humans, SARS-CoV-2, COVID-19 immunology, Dendritic Cells immunology, Spike Glycoprotein, Coronavirus immunology, Toll-Like Receptor 4 immunology
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), an infectious disease characterized by strong induction of inflammatory cytokines, progressive lung inflammation, and potentially multiorgan dysfunction. It remains unclear how SARS-CoV-2 infection leads to immune activation. The Spike (S) protein of SARS-CoV-2 has been suggested to trigger TLR4 and thereby activate immunity. Here, we have investigated the role of TLR4 in SARS-CoV-2 infection and immunity. Neither exposure of isolated S protein, SARS-CoV-2 pseudovirus nor primary SARS-CoV-2 isolate induced TLR4 activation in a TLR4-expressing cell line. Human monocyte-derived DCs express TLR4 but not angiotensin converting enzyme 2 (ACE2), and DCs were not infected by SARS-CoV-2. Notably, neither S protein nor SARS-CoV-2 induced DC maturation or cytokines, indicating that both S protein and SARS-CoV-2 virus particles do not trigger extracellular TLRs including TLR4. Ectopic expression of ACE2 in DCs led to efficient infection by SARS-CoV-2 and, strikingly, efficient type I IFN and cytokine responses. These data strongly suggest that not extracellular TLRs but intracellular viral sensors are key players in sensing SARS-CoV-2. These data imply that SARS-CoV-2 escapes direct sensing by TLRs, which might underlie the lack of efficient immunity to SARS-CoV-2 early during infection., (© 2022 The Authors. European Journal of Immunology published by Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
42. An optimized retroviral toolbox for overexpression and genetic perturbation of primary lymphocytes.
- Author
-
van der Donk LEH, van der Spek J, van Duivenvoorde T, Ten Brink MS, Geijtenbeek TBH, Kuijl CP, van Heijst JWJ, and Ates LS
- Subjects
- Animals, Humans, Lymphocytes, Mice, Promoter Regions, Genetic, Genetic Vectors genetics, Retroviridae genetics
- Abstract
Genetic manipulation of primary lymphocytes is crucial for both clinical purposes and fundamental research. Despite their broad use, we encountered a paucity of data on systematic comparison and optimization of retroviral vectors, the workhorses of genetic modification of primary lymphocytes. Here, we report the construction and validation of a versatile range of retroviral expression vectors. These vectors can be used for the knockdown or overexpression of genes of interest in primary human and murine lymphocytes, in combination with a wide choice of selection and reporter strategies. By streamlining the vector backbone and insert design, these publicly available vectors allow easy interchangeability of the independent building blocks, such as different promoters, fluorescent proteins, surface markers and antibiotic resistance cassettes. We validated these vectors and tested the optimal promoters for in vitro and in vivo overexpression and knockdown of the murine T cell antigen receptor. By publicly sharing these vectors and the data on their optimization, we aim to facilitate genetic modification of primary lymphocytes for researchers entering this field., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
43. Insertion of atypical glycans into the tumor antigen-binding site identifies DLBCLs with distinct origin and behavior.
- Author
-
Chiodin G, Allen JD, Bryant DJ, Rock P, Martino EA, Valle-Argos B, Duriez PJ, Watanabe Y, Henderson I, Blachly JS, McCann KJ, Strefford JC, Packham G, Geijtenbeek TBH, Figdor CG, Wright GW, Staudt LM, Burack R, Bowden TA, Crispin M, Stevenson FK, and Forconi F
- Subjects
- Binding Sites, Cell Adhesion Molecules chemistry, Glycosylation, Humans, Lectins, C-Type chemistry, Lymphoma, Large B-Cell, Diffuse chemistry, Protein Interaction Domains and Motifs, Receptors, Cell Surface chemistry, Tumor Cells, Cultured, Complementarity Determining Regions chemistry, Lymphoma, Large B-Cell, Diffuse pathology, Polysaccharides analysis
- Abstract
Glycosylation of the surface immunoglobulin (Ig) variable region is a remarkable follicular lymphoma-associated feature rarely seen in normal B cells. Here, we define a subset of diffuse large B-cell lymphomas (DLBCLs) that acquire N-glycosylation sites selectively in the Ig complementarity-determining regions (CDRs) of the antigen-binding sites. Mass spectrometry and X-ray crystallography demonstrate how the inserted glycans are stalled at oligomannose-type structures because they are buried in the CDR loops. Acquisition of sites occurs in ∼50% of germinal-center B-cell-like DLBCL (GCB-DLBCL), mainly of the genetic EZB subtype, irrespective of IGHV-D-J use. This markedly contrasts with the activated B-cell-like DLBCL Ig, which rarely has sites in the CDR and does not seem to acquire oligomannose-type structures. Acquisition of CDR-located acceptor sites associates with mutations of epigenetic regulators and BCL2 translocations, indicating an origin shared with follicular lymphoma. Within the EZB subtype, these sites are associated with more rapid disease progression and with significant gene set enrichment of the B-cell receptor, PI3K/AKT/MTORC1 pathway, glucose metabolism, and MYC signaling pathways, particularly in the fraction devoid of MYC translocations. The oligomannose-type glycans on the lymphoma cells interact with the candidate lectin dendritic cell-specific intercellular adhesion molecule 3 grabbing non-integrin (DC-SIGN), mediating low-level signals, and lectin-expressing cells form clusters with lymphoma cells. Both clustering and signaling are inhibited by antibodies specifically targeting the DC-SIGN carbohydrate recognition domain. Oligomannosylation of the tumor Ig is a posttranslational modification that readily identifies a distinct GCB-DLBCL category with more aggressive clinical behavior, and it could be a potential precise therapeutic target via antibody-mediated inhibition of the tumor Ig interaction with DC-SIGN-expressing M2-polarized macrophages., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
44. Complement Potentiates Immune Sensing of HIV-1 and Early Type I Interferon Responses.
- Author
-
Posch W, Bermejo-Jambrina M, Steger M, Witting C, Diem G, Hörtnagl P, Hackl H, Lass-Flörl C, Huber LA, Geijtenbeek TBH, and Wilflingseder D
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing immunology, Dendritic Cells immunology, Dendritic Cells virology, HIV Infections genetics, HIV Infections virology, HIV-1 genetics, Humans, Integrin alphaXbeta2 genetics, Integrin alphaXbeta2 immunology, Interferon Type I genetics, Interferon-Induced Helicase, IFIH1 genetics, Interferon-Induced Helicase, IFIH1 immunology, Receptors, CCR5 genetics, Receptors, CCR5 immunology, Complement System Proteins immunology, HIV Infections immunology, HIV-1 immunology, Interferon Type I immunology
- Abstract
Complement-opsonized HIV-1 triggers efficient antiviral type I interferon (IFN) responses in dendritic cells (DCs), which play an important role in protective responses at the earliest stages in retroviral infection. In contrast, HIV-1 suppresses or escapes sensing by STING- and MAVS-associated sensors. Here, we identified a complement receptor-mediated sensing pathway, where DCs are activated in CCR5/RLR (RIG-I/MDA5)/MAVS/TBK1-dependent fashion. Increased fusion of complement-opsonized HIV-1 via complement receptor 4 and CCR5 leads to increased incoming HIV-1 RNA in the cytoplasm, sensed by a nonredundant cooperative effect of RIG-I and MDA5. Moreover, complement-opsonized HIV-1 down-modulated the MAVS-suppressive Raf-1/PLK1 pathway, thereby opening the antiviral recognition pathway via MAVS. This in turn was followed by MAVS aggregation and subsequent TBK1/IRF3/NF-κB activation in DCs exposed to complement- but not non-opsonized HIV-1. Our data strongly suggest that complement is important in the induction of efficient antiviral immune responses by preventing HIV-1 suppressive mechanisms as well as inducing specific cytosolic sensors. IMPORTANCE Importantly, our study highlights an unusual target on DCs-the α chain of complement receptor 4 (CR4) (CD11c)-for therapeutic interventions in HIV-1 treatment. Targeting CD11c on DCs mediated a potent antiviral immune response via clustering of CR4 and CCR5 and subsequent opening of an antiviral recognition pathway in DCs via MAVS. This novel finding might provide novel tools for specifically boosting endogenous antiviral immunity via CR4, abundantly expressed on multiple DC subsets.
- Published
- 2021
- Full Text
- View/download PDF
45. Infection and transmission of SARS-CoV-2 depend on heparan sulfate proteoglycans.
- Author
-
Bermejo-Jambrina M, Eder J, Kaptein TM, van Hamme JL, Helgers LC, Vlaming KE, Brouwer PJM, van Nuenen AC, Spaargaren M, de Bree GJ, Nijmeijer BM, Kootstra NA, van Gils MJ, Sanders RW, and Geijtenbeek TBH
- Subjects
- 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, COVID-19 transmission, Heparan Sulfate Proteoglycans metabolism, Heparin, Low-Molecular-Weight pharmacology, SARS-CoV-2 pathogenicity
- 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., (© 2021 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
46. Abortive HIV-1 RNA induces pro-IL-1β maturation via protein kinase PKR and inflammasome activation in humans.
- Author
-
Stunnenberg M, van Hamme JL, Trimp M, Gringhuis SI, and Geijtenbeek TBH
- Subjects
- Host-Pathogen Interactions, Humans, MAP Kinase Signaling System, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, RNA, Viral genetics, Reactive Oxygen Species metabolism, Receptors, Pattern Recognition metabolism, Signal Transduction, HIV Infections metabolism, HIV Infections virology, HIV-1 physiology, Inflammasomes metabolism, Interleukin-1beta metabolism, RNA, Viral metabolism, eIF-2 Kinase metabolism
- Abstract
The proinflammatory cytokine IL-1β mediates high levels of immune activation observed during acute and chronic human immunodeficiency virus 1 (HIV-1) infection. Little is known about the mechanisms that drive IL-1β activation during HIV-1 infection. Here, we have identified a crucial role for abortive HIV-1 RNAs in inducing IL-1β in humans. Abortive HIV-1 RNAs were sensed by protein kinase RNA-activated (PKR), which triggered activation of the canonical NLRP3 inflammasome and caspase-1, leading to pro-IL-1β processing and secretion. PKR activated the inflammasome via ROS generation and MAP kinases ERK1/2, JNK, and p38. Inhibition of PKR during HIV-1 infection blocked IL-1β production. As abortive HIV-1 RNAs are produced during productive infection and latency, our data strongly suggest that targeting PKR signaling might attenuate immune activation during acute and chronic HIV-1 infection., (© 2021 The Authors. European Journal of Immunology published by Wiley-VCH GmbH.)
- Published
- 2021
- Full Text
- View/download PDF
47. Variations in the Abortive HIV-1 RNA Hairpin Do Not Impede Viral Sensing and Innate Immune Responses.
- Author
-
Stunnenberg M, van Hamme JL, Das AT, Berkhout B, and Geijtenbeek TBH
- Abstract
The highly conserved trans-acting response element (TAR) present in the RNA genome of human immunodeficiency virus 1 (HIV-1) is a stably folded hairpin structure involved in viral replication. However, TAR is also sensed by viral sensors, leading to antiviral immunity. While high variation in the TAR RNA structure renders the virus replication-incompetent, effects on viral sensing remain unclear. Here, we investigated the role of TAR RNA structure and stability on viral sensing. TAR mutants with deletions in the TAR hairpin that enhanced thermodynamic stability increased antiviral responses. Strikingly, TAR mutants with lower stability due to destabilization of the TAR hairpin also increased antiviral responses without affecting pro-inflammatory responses. Moreover, mutations that affected the TAR RNA sequence also enhanced specific antiviral responses. Our data suggest that mutations in TAR of replication-incompetent viruses can still induce immune responses via viral sensors, hereby underscoring the robustness of HIV-1 RNA sensing mechanisms.
- Published
- 2021
- Full Text
- View/download PDF
48. Therapeutic Liposomal Vaccines for Dendritic Cell Activation or Tolerance.
- Author
-
Nagy NA, de Haas AM, Geijtenbeek TBH, van Ree R, Tas SW, van Kooyk Y, and de Jong EC
- Subjects
- Drug Delivery Systems methods, Humans, Dendritic Cells immunology, Immune Tolerance immunology, Liposomes chemistry, Liposomes immunology, Nanoparticles chemistry, Vaccines chemistry, Vaccines immunology
- Abstract
Dendritic cells (DCs) are paramount in initiating and guiding immunity towards a state of activation or tolerance. This bidirectional capacity of DCs sets them at the center stage for treatment of cancer and autoimmune or allergic conditions. Accordingly, many clinical studies use ex vivo DC vaccination as a strategy to boost anti-tumor immunity or to suppress immunity by including vitamin D3, NF-κB inhibitors or retinoic acid to create tolerogenic DCs. As harvesting DCs from patients and differentiating these cells in vitro is a costly and cumbersome process, in vivo targeting of DCs has huge potential as nanoparticulate platforms equipped with activating or tolerogenic adjuvants can modulate DCs in their natural environment. There is a rapid expansion of the choices of nanoparticles and activation- or tolerance-promoting adjuvants for a therapeutic vaccine platform. In this review we highlight the most recent nanomedical approaches aimed at inducing immune activation or tolerance via targeting DCs, together with novel fundamental insights into the mechanisms inherent to fostering anti-tumor or tolerogenic immunity., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Nagy, de Haas, Geijtenbeek, van Ree, Tas, van Kooyk and de Jong.)
- Published
- 2021
- Full Text
- View/download PDF
49. HIV-1 subverts the complement system in semen to enhance viral transmission.
- Author
-
Nijmeijer BM, Bermejo-Jambrina M, Kaptein TM, Ribeiro CMS, Wilflingseder D, and Geijtenbeek TBH
- Subjects
- 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, HIV Infections immunology, HIV-1 physiology, Langerhans Cells immunology, Semen immunology
- 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.
- Published
- 2021
- Full Text
- View/download PDF
50. Intestinal CD8 + T cell responses are abundantly induced early in human development but show impaired cytotoxic effector capacities.
- Author
-
Schreurs RRCE, Sagebiel AF, Steinert FL, Highton AJ, Klarenbeek PL, Drewniak A, Bakx R, The SML, Ribeiro CMS, Perez D, Reinshagen K, Geijtenbeek TBH, van Goudoever JB, and Bunders MJ
- Subjects
- Cytotoxicity, Immunologic, Disease Susceptibility, Female, Fetus, Gene Expression Regulation, Developmental, Humans, Immune Tolerance, Infant, Male, Middle Aged, Programmed Cell Death 1 Receptor genetics, Programmed Cell Death 1 Receptor metabolism, CD8-Positive T-Lymphocytes immunology, Intestines immunology, Memory T Cells immunology, Virus Diseases immunology
- Abstract
Gastrointestinal viral infections are a major global cause of disease and mortality in infants. Cytotoxic CD8
+ T cells are critical to achieve viral control. However, studies investigating the development of CD8+ T cell immunity in human tissues early in life are lacking. Here, we investigated the maturation of the CD8+ T cell compartment in human fetal, infant and adult intestinal tissues. CD8+ T cells exhibiting a memory phenotype were already detected in fetal intestines and increased after birth. Infant intestines preferentially harbored effector CCR7- CD45RA- CD127- KLRG1+/- CD8+ T cells compared to tissue-resident memory CD69+ CD103+ CD8+ T cells detected in adults. Functional cytotoxic capacity, including cytokine and granzyme B production of infant intestinal effector CD8+ T cells was, however, markedly reduced compared to adult intestinal CD8+ T cells. This was in line with the high expression of the inhibitory molecule PD-1 by infant intestinal effector CD8+ T cells. Taken together, we demonstrate that intestinal CD8+ T cell responses are induced early in human development, however exhibit a reduced functionality. The impaired CD8+ T cell functionality early in life contributes to tolerance during foreign antigen exposure after birth, however functions as an immune correlate for the increased susceptibility to gastrointestinal viral infections in infancy.- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.