36 results on '"van Hamme JL"'
Search Results
2. 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
3. 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
4. Control of complement-induced inflammatory responses to SARS-CoV-2 infection by anti-SARS-CoV-2 antibodies.
- Author
-
Bermejo-Jambrina M, van der Donk LE, van Hamme JL, Wilflingseder D, de Bree G, Prins M, de Jong M, Nieuwkerk P, van Gils MJ, Kootstra NA, and Geijtenbeek TB
- Subjects
- Humans, SARS-CoV-2, Antibodies, Viral, Complement System Proteins, Inflammation, Immunity, Innate, COVID-19
- Abstract
Dysregulated immune responses contribute to the excessive and uncontrolled inflammation observed in severe COVID-19. However, how immunity to SARS-CoV-2 is induced and regulated remains unclear. Here, we uncover the role of the complement system in the induction of innate and adaptive immunity to SARS-CoV-2. Complement rapidly opsonizes SARS-CoV-2 particles via the lectin pathway. Complement-opsonized SARS-CoV-2 efficiently induces type-I interferon and pro-inflammatory cytokine responses via activation of dendritic cells, which are inhibited by antibodies against the complement receptors (CR) 3 and 4. Serum from COVID-19 patients, or monoclonal antibodies against SARS-CoV-2, attenuate innate and adaptive immunity induced by complement-opsonized SARS-CoV-2. Blocking of CD32, the FcγRII antibody receptor of dendritic cells, restores complement-induced immunity. These results suggest that opsonization of SARS-CoV-2 by complement is involved in the induction of innate and adaptive immunity to SARS-CoV-2 in the acute phase of infection. Subsequent antibody responses limit inflammation and restore immune homeostasis. These findings suggest that dysregulation of the complement system and FcγRII signaling may contribute to severe COVID-19., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. 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
6. 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
7. 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
8. 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
9. Vaginal bacterium Prevotella timonensis turns protective Langerhans cells into HIV-1 reservoirs for virus dissemination.
- Author
-
van Teijlingen NH, Helgers LC, Sarrami-Forooshani R, Zijlstra-Willems EM, van Hamme JL, Segui-Perez C, van Smoorenburg MY, Borgdorff H, van de Wijgert JH, van Leeuwen E, van der Post JA, Strijbis K, Ribeiro CM, and Geijtenbeek TB
- Subjects
- Antiviral Agents, Dysbiosis, Female, Humans, Langerhans Cells, Prevotella, HIV Infections, HIV-1
- Abstract
Dysbiosis of vaginal microbiota is associated with increased HIV-1 acquisition, but the underlying cellular mechanisms remain unclear. Vaginal Langerhans cells (LCs) protect against mucosal HIV-1 infection via autophagy-mediated degradation of HIV-1. As LCs are in continuous contact with bacterial members of the vaginal microbiome, we investigated the impact of commensal and dysbiosis-associated vaginal (an)aerobic bacterial species on the antiviral function of LCs. Most of the tested bacteria did not affect the HIV-1 restrictive function of LCs. However, Prevotella timonensis induced a vast uptake of HIV-1 by vaginal LCs. Internalized virus remained infectious for days and uptake was unaffected by antiretroviral drugs. P. timonensis-exposed LCs efficiently transmitted HIV-1 to target cells both in vitro and ex vivo. Additionally, P. timonensis exposure enhanced uptake and transmission of the HIV-1 variants that establish infection after sexual transmission, the so-called Transmitted Founder variants. Our findings, therefore, suggest that P. timonensis might set the stage for enhanced HIV-1 susceptibility during vaginal dysbiosis and advocate targeted treatment of P. timonensis during bacterial vaginosis to limit HIV-1 infection., (© 2022 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
10. 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, Antiviral Agents, 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
11. 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
12. 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
13. 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
14. 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
15. Autophagy-enhancing drugs limit mucosal HIV-1 acquisition and suppress viral replication ex vivo.
- Author
-
Cloherty APM, van Teijlingen NH, Eisden TTHD, van Hamme JL, Rader AG, Geijtenbeek TBH, Schreurs RRCE, and Ribeiro CMS
- Subjects
- Cell Line, Cells, Cultured, HIV-1 physiology, Humans, Mucous Membrane drug effects, Mucous Membrane virology, Anti-HIV Agents pharmacology, Autophagy drug effects, HIV Infections drug therapy, HIV-1 drug effects, Virus Replication drug effects
- Abstract
Current direct-acting antiviral therapies are highly effective in suppressing HIV-1 replication. However, mucosal inflammation undermines prophylactic treatment efficacy, and HIV-1 persists in long-lived tissue-derived dendritic cells (DCs) and CD4
+ T cells of treated patients. Host-directed strategies are an emerging therapeutic approach to improve therapy outcomes in infectious diseases. Autophagy functions as an innate antiviral mechanism by degrading viruses in specialized vesicles. Here, we investigated the impact of pharmaceutically enhancing autophagy on HIV-1 acquisition and viral replication. To this end, we developed a human tissue infection model permitting concurrent analysis of HIV-1 cellular targets ex vivo. Prophylactic treatment with autophagy-enhancing drugs carbamazepine and everolimus promoted HIV-1 restriction in skin-derived CD11c+ DCs and CD4+ T cells. Everolimus also decreased HIV-1 susceptibility to lab-adapted and transmitted/founder HIV-1 strains, and in vaginal Langerhans cells. Notably, we observed cell-specific effects of therapeutic treatment. Therapeutic rapamycin treatment suppressed HIV-1 replication in tissue-derived CD11c+ DCs, while all selected drugs limited viral replication in CD4+ T cells. Strikingly, both prophylactic and therapeutic treatment with everolimus or rapamycin reduced intestinal HIV-1 productive infection. Our findings highlight host autophagy pathways as an emerging target for HIV-1 therapies, and underscore the relevancy of repurposing clinically-approved autophagy drugs to suppress mucosal HIV-1 replication.- Published
- 2021
- Full Text
- View/download PDF
16. Neutrophil specific granule and NETosis defects in gray platelet syndrome.
- Author
-
Aarts CEM, Downes K, Hoogendijk AJ, Sprenkeler EGG, Gazendam RP, Favier R, Favier M, Tool ATJ, van Hamme JL, Kostadima MA, Waller K, Zieger B, van Bergen MGJM, Langemeijer SMC, van der Reijden BA, Janssen H, van den Berg TK, van Bruggen R, Meijer AB, Ouwehand WH, and Kuijpers TW
- Subjects
- Animals, Blood Platelets, Blood Proteins, Cytoplasmic Granules, Humans, Mice, Neutrophils, Gray Platelet Syndrome genetics
- Abstract
Gray platelet syndrome (GPS) is an autosomal recessive bleeding disorder characterized by a lack of α-granules in platelets and progressive myelofibrosis. Rare loss-of-function variants in neurobeachin-like 2 (NBEAL2), a member of the family of beige and Chédiak-Higashi (BEACH) genes, are causal of GPS. It is suggested that BEACH domain containing proteins are involved in fusion, fission, and trafficking of vesicles and granules. Studies in knockout mice suggest that NBEAL2 may control the formation and retention of granules in neutrophils. We found that neutrophils obtained from the peripheral blood from 13 patients with GPS have a normal distribution of azurophilic granules but show a deficiency of specific granules (SGs), as confirmed by immunoelectron microscopy and mass spectrometry proteomics analyses. CD34+ hematopoietic stem cells (HSCs) from patients with GPS differentiated into mature neutrophils also lacked NBEAL2 expression but showed similar SG protein expression as control cells. This is indicative of normal granulopoiesis in GPS and identifies NBEAL2 as a potentially important regulator of granule release. Patient neutrophil functions, including production of reactive oxygen species, chemotaxis, and killing of bacteria and fungi, were intact. NETosis was absent in circulating GPS neutrophils. Lack of NETosis is suggested to be independent of NBEAL2 expression but associated with SG defects instead, as indicated by comparison with HSC-derived neutrophils. Since patients with GPS do not excessively suffer from infections, the consequence of the reduced SG content and lack of NETosis for innate immunity remains to be explored., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
17. Vaginal dysbiosis associated-bacteria Megasphaera elsdenii and Prevotella timonensis induce immune activation via dendritic cells.
- Author
-
van Teijlingen NH, Helgers LC, Zijlstra-Willems EM, van Hamme JL, Ribeiro CMS, Strijbis K, and Geijtenbeek TBH
- Subjects
- Cells, Cultured, Cytokines metabolism, Dendritic Cells metabolism, Dysbiosis immunology, Dysbiosis microbiology, Female, Humans, Inflammation Mediators metabolism, Leukocytes, Mononuclear, Prevotella isolation & purification, Primary Cell Culture, Vagina cytology, Vagina immunology, Vagina microbiology, Vaginosis, Bacterial microbiology, Dendritic Cells immunology, Dysbiosis complications, Megasphaera elsdenii immunology, Prevotella immunology, Vaginosis, Bacterial immunology
- Abstract
Dysbiosis of the vaginal microbiome as a result of overgrowth of anaerobic bacteria leads to bacterial vaginosis (BV) which is associated with increased inflammation in the genital mucosa. Moreover, BV increases susceptibility to sexual transmitted infections (STIs) and is associated with adverse pregnancy outcomes. It remains unclear how specific vaginal aerobic and anaerobic bacteria affect health and disease. We selected different vaginal bacteria ranging from true commensals to species associated with dysbiosis and investigated their effects on activation of dendritic cells (DCs). Commensal Lactobacilli crispatus did not induce DC maturation nor led to production of pro-inflammatory cytokines. In contrast, BV-associated bacteria Megasphaera elsdenii and Prevotella timonensis induced DC maturation and increased levels of pro-inflammatory cytokines. Notably, DCs stimulated with Prevotella timonensis suppressed Th2 responses and induced Th1 skewing, typically associated with preterm birth. In contrast, Lactobacillus crispatus and Megasphaera elsdenii did not affect Th cell polarization. These results strongly indicate that the interaction of vaginal bacteria with mucosal DCs determines mucosal inflammation and we have identified the anaerobic bacterium Prevotella timonensis as a strong inducer of inflammatory responses. Specifically targeting these inflammation-inducing bacteria might be a therapeutic strategy to prevent BV and associated risks in STI susceptibility and preterm birth., Competing Interests: Declaration of Competing Interest We wish to confirm that there are no known conflicts of interest associated with this publication and there has been no significant financial support for this work that could have influenced its outcome., (Copyright © 2020 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
18. Synthetic Abortive HIV-1 RNAs Induce Potent Antiviral Immunity.
- Author
-
Stunnenberg M, Sprokholt JK, van Hamme JL, Kaptein TM, Zijlstra-Willems EM, Gringhuis SI, and Geijtenbeek TBH
- Subjects
- 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, Adaptive Immunity, HIV Infections immunology, HIV-1 genetics, Host Microbial Interactions immunology, Immunity, Innate, RNA, Viral pharmacology
- 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-RNA
58 ) 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 TH 2 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., (Copyright © 2020 Stunnenberg, Sprokholt, van Hamme, Kaptein, Zijlstra-Willems, Gringhuis and Geijtenbeek.)- Published
- 2020
- Full Text
- View/download PDF
19. Sexually transmitted founder HIV-1 viruses are relatively resistant to Langerhans cell-mediated restriction.
- Author
-
Hertoghs N, Nijmeijer BM, van Teijlingen NH, Fenton-May AE, Kaptein TM, van Hamme JL, Kappes JC, Kootstra NA, Hahn BH, Borrow P, Ribeiro CMS, and Geijtenbeek TBH
- Subjects
- Antigens, CD metabolism, Cells, Cultured, HEK293 Cells, Humans, Langerhans Cells immunology, Lectins, C-Type metabolism, Mannose-Binding Lectins metabolism, HIV-1 pathogenicity, Host-Pathogen Interactions, Langerhans Cells virology
- 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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
20. Inherited p40phox deficiency differs from classic chronic granulomatous disease.
- Author
-
van de Geer A, Nieto-Patlán A, Kuhns DB, Tool AT, Arias AA, Bouaziz M, de Boer M, Franco JL, Gazendam RP, van Hamme JL, van Houdt M, van Leeuwen K, Verkuijlen PJ, van den Berg TK, Alzate JF, Arango-Franco CA, Batura V, Bernasconi AR, Boardman B, Booth C, Burns SO, Cabarcas F, Bensussan NC, Charbit-Henrion F, Corveleyn A, Deswarte C, Azcoiti ME, Foell D, Gallin JI, Garcés C, Guedes M, Hinze CH, Holland SM, Hughes SM, Ibañez P, Malech HL, Meyts I, Moncada-Velez M, Moriya K, Neves E, Oleastro M, Perez L, Rattina V, Oleaga-Quintas C, Warner N, Muise AM, López JS, Trindade E, Vasconcelos J, Vermeire S, Wittkowski H, Worth A, Abel L, Dinauer MC, Arkwright PD, Roos D, Casanova JL, Kuijpers TW, and Bustamante J
- Subjects
- Adolescent, Adult, Alleles, Child, Child, Preschool, Female, Gene Knockout Techniques, Granulomatous Disease, Chronic diagnosis, Granulomatous Disease, Chronic metabolism, HEK293 Cells, Humans, Male, Middle Aged, Mutant Proteins genetics, Mutant Proteins metabolism, NADPH Oxidases genetics, NADPH Oxidases metabolism, Pedigree, Phagocytes immunology, Phagocytes metabolism, Phagocytes microbiology, Phenotype, Phosphoproteins metabolism, Prognosis, RNA, Messenger genetics, RNA, Messenger metabolism, Transduction, Genetic, Young Adult, Granulomatous Disease, Chronic genetics, Loss of Function Mutation, Phosphoproteins deficiency, Phosphoproteins genetics
- Abstract
Biallelic loss-of-function (LOF) mutations of the NCF4 gene, encoding the p40phox subunit of the phagocyte NADPH oxidase, have been described in only 1 patient. We report on 24 p40phox-deficient patients from 12 additional families in 8 countries. These patients display 8 different in-frame or out-of-frame mutations of NCF4 that are homozygous in 11 of the families and compound heterozygous in another. When overexpressed in NB4 neutrophil-like cells and EBV-transformed B cells in vitro, the mutant alleles were found to be LOF, with the exception of the p.R58C and c.120_134del alleles, which were hypomorphic. Particle-induced NADPH oxidase activity was severely impaired in the patients' neutrophils, whereas PMA-induced dihydrorhodamine-1,2,3 (DHR) oxidation, which is widely used as a diagnostic test for chronic granulomatous disease (CGD), was normal or mildly impaired in the patients. Moreover, the NADPH oxidase activity of EBV-transformed B cells was also severely impaired, whereas that of mononuclear phagocytes was normal. Finally, the killing of Candida albicans and Aspergillus fumigatus hyphae by neutrophils was conserved in these patients, unlike in patients with CGD. The patients suffer from hyperinflammation and peripheral infections, but they do not have any of the invasive bacterial or fungal infections seen in CGD. Inherited p40phox deficiency underlies a distinctive condition, resembling a mild, atypical form of CGD.
- Published
- 2018
- Full Text
- View/download PDF
21. RIG-I-like receptor activation by dengue virus drives follicular T helper cell formation and antibody production.
- Author
-
Sprokholt JK, Kaptein TM, van Hamme JL, Overmars RJ, Gringhuis SI, and Geijtenbeek TBH
- Subjects
- 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, Antibodies, Viral immunology, Dengue immunology, Dengue Virus physiology, T-Lymphocytes, Helper-Inducer immunology
- 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/β 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.
- Published
- 2017
- Full Text
- View/download PDF
22. DCs facilitate B cell responses against microbial DNA via DC-SIGN.
- Author
-
Sprokholt JK, Heineke MH, Kaptein TM, van Hamme JL, and Geijtenbeek TBH
- Subjects
- Cytokines biosynthesis, Humans, Interferon Type I biosynthesis, B-Lymphocytes immunology, Cell Adhesion Molecules physiology, DNA, Bacterial immunology, Dendritic Cells immunology, Escherichia coli genetics, Lectins, C-Type physiology, Receptors, Cell Surface physiology
- 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.
- Published
- 2017
- Full Text
- View/download PDF
23. RIG-I-like Receptor Triggering by Dengue Virus Drives Dendritic Cell Immune Activation and T H 1 Differentiation.
- Author
-
Sprokholt JK, Kaptein TM, van Hamme JL, Overmars RJ, Gringhuis SI, and Geijtenbeek TBH
- Subjects
- 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, DEAD Box Protein 58 immunology, Dendritic Cells immunology, Dengue Virus immunology, Th1 Cells immunology
- 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-1β, 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/β 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 T
H cell differentiation, and we show that RIG-I and MDA5 triggering by DENV leads to TH 1 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., (Copyright © 2017 by The American Association of Immunologists, Inc.)- Published
- 2017
- Full Text
- View/download PDF
24. Dexamethasone promotes granulocyte mobilization by prolonging the half-life of granulocyte-colony-stimulating factor in healthy donors for granulocyte transfusions.
- Author
-
Hiemstra IH, van Hamme JL, Janssen MH, van den Berg TK, and Kuijpers TW
- Subjects
- Adult, Annexin A5 metabolism, Cell Survival drug effects, Filgrastim administration & dosage, Filgrastim pharmacokinetics, GPI-Linked Proteins metabolism, Half-Life, Humans, Male, Middle Aged, Mifepristone pharmacology, Neutrophils cytology, Plasma metabolism, Receptors, CXCR4 metabolism, Receptors, IgG metabolism, Dexamethasone administration & dosage, Dexamethasone pharmacokinetics, Leukocyte Transfusion, Neutrophils metabolism
- Abstract
Background: Granulocyte transfusion (GTX) is a potential approach to correcting neutropenia and relieving the increased risk of infection in patients who are refractory to antibiotics. To mobilize enough granulocytes for transfusion, healthy donors are premedicated with granulocyte-colony-stimulating factor (G-CSF) and dexamethasone. Granulocytes have a short circulatory half-life. Consequently, patients need to receive GTX every other day to keep circulating granulocyte counts at an acceptable level. We investigated whether plasma from premedicated donors was capable of prolonging neutrophil survival and, if so, which factor could be held responsible., Study Design and Methods: The effects of plasma from G-CSF/dexamethasone-treated donors on neutrophil survival were assessed by annexin-V, CD16. and CXCR4 staining and nuclear morphology. We isolated an albumin-bound protein using α-chymotrypsin and albumin-depletion and further characterized it using protein analysis. The effects of dexamethasone and G-CSF were assessed using mifepristone and G-CSF-neutralizing antibody. G-CSF plasma concentrations were determined by Western blot and Luminex analyses., Results: G-CSF/dexamethasone plasma contained a survival-promoting factor for at least 2 days. This factor was recognized as an albumin-associated protein and was identified as G-CSF itself, which was surprising considering its reported half-life of only 4.5 hours. Compared with coadministration of dexamethasone, administration of G-CSF alone to the same GTX donors led to a faster decline in circulating G-CSF levels, whereas dexamethasone itself did not induce any G-CSF, demonstrating a role for dexamethasone in increasing G-CSF half-life., Conclusion: Dexamethasone increases granulocyte yield upon coadministration with G-CSF by extending G-CSF half-life. This observation might also be exploited in the coadministration of dexamethasone with other recombinant proteins to modulate their half-life., (© 2016 AABB.)
- Published
- 2017
- Full Text
- View/download PDF
25. Characterization of buffy coat-derived granulocytes for clinical use: a comparison with granulocyte colony-stimulating factor/dexamethasone-pretreated donor-derived products.
- Author
-
van de Geer A, Gazendam RP, Tool AT, van Hamme JL, de Korte D, van den Berg TK, Zeerleder SS, and Kuijpers TW
- Subjects
- Adult, Antigens, Surface metabolism, Blood Component Removal, Blood Donors, Blood Platelets cytology, Cell Adhesion drug effects, Cell Survival drug effects, Chemotaxis drug effects, Granulocytes cytology, Granulocytes immunology, Granulocytes metabolism, Humans, Immunophenotyping, Leukocyte Count, Male, NADPH Oxidases metabolism, Reactive Oxygen Species metabolism, Blood Buffy Coat cytology, Dexamethasone pharmacology, Granulocyte Colony-Stimulating Factor pharmacology, Granulocytes drug effects
- Abstract
Background and Objectives: Buffy coat-derived granulocytes have been described as an alternative to the apheresis product from donors pretreated with dexamethasone and granulocyte colony-stimulating factor (G-CSF). The latter is - dependent on the local and national settings - obtained following a demanding and time-consuming procedure, which is undesirable in critically ill septic patients. In contrast, buffy coat-derived products have a large volume and are often heavily contaminated with red cells and platelets. We developed a new pooled buffy coat-derived product with high purity and small volume, and performed a comprehensive functional characterization of these granulocytes., Materials and Methods: We pooled ten buffy coats following the production of platelet concentrates. Saline 0·9% was added to decrease the viscosity and the product was split into plasma, red cells and a 'super' buffy coat. Functional data of the granulocytes were compared to those obtained with granulocytes from healthy controls and G-CSF/dexamethasone-pretreated donors., Results: Buffy coat-derived granulocytes showed adhesion, chemotaxis, reactive oxygen species production, degranulation, NETosis and in vitro killing of Staphylococcus aureus, Escherichia coli and Aspergillus species comparable to control and G-CSF/dexamethasone-derived granulocytes. Candida killing was superior compared to G-CSF/dexamethasone-derived granulocytes. Immunophenotyping was normal; especially no signs of activation in the buffy coat-derived granulocytes were seen. Viability was reduced. Buffy coats are readily available in the regular blood production process and would take away the concerns around the apheresis product., Conclusion: The product described appears a promising alternative for transfusion purposes., (© 2017 International Society of Blood Transfusion.)
- Published
- 2017
- Full Text
- View/download PDF
26. Receptor usage dictates HIV-1 restriction by human TRIM5α in dendritic cell subsets.
- Author
-
Ribeiro CM, Sarrami-Forooshani R, Setiawan LC, Zijlstra-Willems EM, van Hamme JL, Tigchelaar W, van der Wel NN, Kootstra NA, Gringhuis SI, and Geijtenbeek TB
- Subjects
- Antiviral Restriction Factors, Cell Adhesion Molecules metabolism, Cell Line, HIV Infections immunology, HIV Infections prevention & control, HIV Infections transmission, HIV-1 immunology, Host-Pathogen Interactions, Humans, Immunity, Mucosal, Langerhans Cells cytology, Langerhans Cells immunology, Receptors, Cell Surface metabolism, Tripartite Motif Proteins, Ubiquitin-Protein Ligases, Antigens, CD metabolism, Autophagy, Carrier Proteins metabolism, HIV-1 physiology, Langerhans Cells metabolism, Langerhans Cells virology, Lectins, C-Type metabolism, Mannose-Binding Lectins metabolism, Receptors, HIV metabolism
- Abstract
The most prevalent route of HIV-1 infection is across mucosal tissues after sexual contact. Langerhans cells (LCs) belong to the subset of dendritic cells (DCs) that line the mucosal epithelia of vagina and foreskin and have the ability to sense and induce immunity to invading pathogens. Anatomical and functional characteristics make LCs one of the primary targets of HIV-1 infection. Notably, LCs form a protective barrier against HIV-1 infection and transmission. LCs restrict HIV-1 infection through the capture of HIV-1 by the C-type lectin receptor Langerin and subsequent internalization into Birbeck granules. However, the underlying molecular mechanism of HIV-1 restriction in LCs remains unknown. Here we show that human E3-ubiquitin ligase tri-partite-containing motif 5α (TRIM5α) potently restricts HIV-1 infection of LCs but not of subepithelial DC-SIGN
+ DCs. HIV-1 restriction by TRIM5α was thus far considered to be reserved to non-human primate TRIM5α orthologues, but our data strongly suggest that human TRIM5α is a cell-specific restriction factor dependent on C-type lectin receptor function. Our findings highlight the importance of HIV-1 binding to Langerin for the routeing of HIV-1 into the human TRIM5α-mediated restriction pathway. TRIM5α mediates the assembly of an autophagy-activating scaffold to Langerin, which targets HIV-1 for autophagic degradation and prevents infection of LCs. By contrast, HIV-1 binding to DC-SIGN+ DCs leads to disassociation of TRIM5α from DC-SIGN, which abrogates TRIM5α restriction. Thus, our data strongly suggest that restriction by human TRIM5α is controlled by C-type-lectin-receptor-dependent uptake of HIV-1, dictating protection or infection of human DC subsets. Therapeutic interventions that incorporate C-type lectin receptors and autophagy-targeting strategies could thus provide cell-mediated resistance to HIV-1 in humans.- Published
- 2016
- Full Text
- View/download PDF
27. Proinflammatory cytokine response toward fungi but not bacteria in chronic granulomatous disease.
- Author
-
Gazendam RP, van de Geer A, van Hamme JL, Helgers L, Rohr J, Chrabieh M, Picard C, Roos D, van den Berg JM, van den Berg TK, and Kuijpers TW
- Subjects
- Cell Line, Granulomatous Disease, Chronic genetics, Humans, Inflammation genetics, Monocytes immunology, Mutation genetics, NADPH Oxidases genetics, Neutrophils immunology, Bacteria immunology, Cytokines immunology, Fungi immunology, Granulomatous Disease, Chronic immunology, Inflammation immunology
- Published
- 2016
- Full Text
- View/download PDF
28. Impaired killing of Candida albicans by granulocytes mobilized for transfusion purposes: a role for granule components.
- Author
-
Gazendam RP, van de Geer A, van Hamme JL, Tool AT, van Rees DJ, Aarts CE, van den Biggelaar M, van Alphen F, Verkuijlen P, Meijer AB, Janssen H, Roos D, van den Berg TK, and Kuijpers TW
- Subjects
- Biomarkers, Cell Degranulation drug effects, Cell Degranulation immunology, Cytoplasmic Granules immunology, Cytoplasmic Granules metabolism, Dexamethasone pharmacology, Granulocyte Colony-Stimulating Factor pharmacology, Granulocytes drug effects, Granulocytes metabolism, Granulocytes microbiology, Humans, Immunophenotyping, NADPH Oxidases metabolism, Neutrophils drug effects, Neutrophils immunology, Neutrophils metabolism, Neutrophils microbiology, Phagocytosis drug effects, Phagocytosis immunology, Phagosomes immunology, Phagosomes microbiology, Candida albicans immunology, Cytotoxicity, Immunologic, Granulocytes immunology, Leukocyte Transfusion, Microbial Viability immunology
- Abstract
Granulocyte transfusions are used to treat neutropenic patients with life-threatening bacterial or fungal infections that do not respond to anti-microbial drugs. Donor neutrophils that have been mobilized with granulocyte-colony stimulating factor (G-CSF) and dexamethasone are functional in terms of antibacterial activity, but less is known about their fungal killing capacity. We investigated the neutrophil-mediated cytotoxic response against C. albicans and A. fumigatus in detail. Whereas G-CSF/dexamethasone-mobilized neutrophils appeared less mature as compared to neutrophils from untreated controls, these cells exhibited normal ROS production by the NADPH oxidase system and an unaltered granule mobilization capacity upon stimulation. G-CSF/dexamethasone-mobilized neutrophils efficiently inhibited A. fumigatus germination and killed Aspergillus and Candida hyphae, but the killing of C. albicans yeasts was distinctly impaired. Following normal Candida phagocytosis, analysis by mass spectrometry of purified phagosomes after fusion with granules demonstrated that major constituents of the antimicrobial granule components, including major basic protein (MBP), were reduced. Purified MBP showed candidacidal activity, and neutrophil-like Crisp-Cas9 NB4-KO-MBP differentiated into phagocytes were impaired in Candida killing. Together, these findings indicate that G-CSF/dexamethasone-mobilized neutrophils for transfusion purposes have a selectively impaired capacity to kill Candida yeasts, as a consequence of an altered neutrophil granular content., (Copyright© Ferrata Storti Foundation.)
- Published
- 2016
- Full Text
- View/download PDF
29. Human Neutrophils Use Different Mechanisms To Kill Aspergillus fumigatus Conidia and Hyphae: Evidence from Phagocyte Defects.
- Author
-
Gazendam RP, van Hamme JL, Tool AT, Hoogenboezem M, van den Berg JM, Prins JM, Vitkov L, van de Veerdonk FL, van den Berg TK, Roos D, and Kuijpers TW
- Subjects
- Cytotoxicity, Immunologic immunology, Extracellular Traps immunology, Humans, Immunity, Innate, Immunologic Deficiency Syndromes immunology, Microscopy, Confocal, Phagocytes immunology, Aspergillosis immunology, Aspergillus fumigatus immunology, Hyphae immunology, Neutrophils immunology, Spores, Fungal immunology
- Abstract
Neutrophils are known to play a pivotal role in the host defense against Aspergillus infections. This is illustrated by the prevalence of Aspergillus infections in patients with neutropenia or phagocyte functional defects, such as chronic granulomatous disease. However, the mechanisms by which human neutrophils recognize and kill Aspergillus are poorly understood. In this work, we have studied in detail which neutrophil functions, including neutrophil extracellular trap (NET) formation, are involved in the killing of Aspergillus fumigatus conidia and hyphae, using neutrophils from patients with well-defined genetic immunodeficiencies. Recognition of conidia involves integrin CD11b/CD18 (and not dectin-1), which triggers a PI3K-dependent nonoxidative intracellular mechanism of killing. When the conidia escape from early killing and germinate, the extracellular destruction of the Aspergillus hyphae needs opsonization by Abs and involves predominantly recognition via Fcγ receptors, signaling via Syk, PI3K, and protein kinase C to trigger the production of toxic reactive oxygen metabolites by the NADPH oxidase and myeloperoxidase. A. fumigatus induces NET formation; however, NETs did not contribute to A. fumigatus killing. Thus, our findings reveal distinct killing mechanisms of Aspergillus conidia and hyphae by human neutrophils, leading to a comprehensive insight in the innate antifungal response., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
30. ADAR1 Facilitates HIV-1 Replication in Primary CD4+ T Cells.
- Author
-
Cuadrado E, Booiman T, van Hamme JL, Jansen MH, van Dort KA, Vanderver A, Rice GI, Crow YJ, Kootstra NA, and Kuijpers TW
- Subjects
- Adenosine Deaminase genetics, Adolescent, Blotting, Western, Cells, Cultured, Child, Child, Preschool, Female, Flow Cytometry, HIV Infections metabolism, Humans, Jurkat Cells, Male, RNA, Messenger genetics, RNA, Viral genetics, RNA-Binding Proteins genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Adenosine Deaminase metabolism, CD4-Positive T-Lymphocytes virology, HIV Infections virology, HIV-1 pathogenicity, RNA-Binding Proteins metabolism, Virus Replication
- Abstract
Unlike resting CD4+ T cells, activated CD4+T cells are highly susceptible to infection of human immunodeficiency virus 1 (HIV-1). HIV-1 infects T cells and macrophages without activating the nucleic acid sensors and the anti-viral type I interferon response. Adenosine deaminase acting on RNA 1 (ADAR1) is an RNA editing enzyme that displays antiviral activity against several RNA viruses. Mutations in ADAR1 cause the autoimmune disorder Aicardi-Goutieères syndrome (AGS). This disease is characterized by an inappropriate activation of the interferon-stimulated gene response. Here we show that HIV-1 replication, in ADAR1-deficient CD4+T lymphocytes from AGS patients, is blocked at the level of protein translation. Furthermore, viral protein synthesis block is accompanied by an activation of interferon-stimulated genes. RNA silencing of ADAR1 in Jurkat cells also inhibited HIV-1 protein synthesis. Our data support that HIV-1 requires ADAR1 for efficient replication in human CD4+T cells.
- Published
- 2015
- Full Text
- View/download PDF
31. Impaired microbial killing by neutrophils from patients with protein kinase C delta deficiency.
- Author
-
Szilagyi K, Gazendam RP, van Hamme JL, Tool AT, van Houdt M, Vos WA, Verkuijlen P, Janssen H, Belot A, Juillard L, Förster-Waldl E, Boztug K, Kraal G, de Winther MP, Kuijpers TW, and van den Berg TK
- Subjects
- Adolescent, Adult, Anti-Infective Agents immunology, Autoimmune Diseases genetics, Cytoplasmic Granules immunology, Female, Humans, Infections genetics, Male, Mutation genetics, NADP metabolism, Protein Kinase C-delta genetics, Reactive Oxygen Species metabolism, Recurrence, Young Adult, Anti-Infective Agents metabolism, Autoimmune Diseases immunology, Cytoplasmic Granules metabolism, Cytotoxicity, Immunologic, Infections immunology, Neutrophils immunology, Protein Kinase C-delta deficiency
- Published
- 2015
- Full Text
- View/download PDF
32. Fetal exposure to HIV-1 alters chemokine receptor expression by CD4+T cells and increases susceptibility to HIV-1.
- Author
-
Bunders MJ, van Hamme JL, Jansen MH, Boer K, Kootstra NA, and Kuijpers TW
- Subjects
- CD4 Lymphocyte Count, CD4-Positive T-Lymphocytes immunology, Case-Control Studies, Cluster Analysis, Female, Fetal Blood cytology, Gene Expression Profiling, HIV Infections immunology, Humans, Immunophenotyping, Infant, Infant, Newborn, Phenotype, Pregnancy, CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes virology, Gene Expression, Genetic Predisposition to Disease, HIV Infections genetics, HIV Infections virology, HIV-1, Receptors, Chemokine genetics
- Abstract
Absolute numbers of lymphocytes are decreased in uninfected infants born to HIV-1-infected women (HIV-1-exposed). Although the exact mechanism is unknown, fetal exposure to maternal HIV-1-infection could prime the immune system and affect T cell trafficking. We compared the expression of chemokine receptors on cord blood CD4(+) T cells from HIV-1-exposed children and healthy controls. At baseline CD4(+) T cells had a largely naïve phenotype. However, stimulation with cytokines resulted in an upregulation of inflammatory response-related chemokine receptors on CD4(+) T cells, with HIV-1-exposed infants having a significantly higher frequency of CD4(+) T cells expressing, in particularly Th2 associated chemokine receptors (CCR3 p < 0.01, CCR8 p = 0.03). Numbers of naive CCR7(+) CD4(+) T cells were reduced (p = 0.01) in HIV-1-exposed infants. We further assessed whether the inflammatory phenotype was associated with susceptibility to HIV-1 and detected higher levels of p24 upon in in vitro infection of stimulated CD4(+) T cells of HIV-1-exposed infants. In summary, fetal exposure to HIV-1 primes the immune system in the infant leading to an enhanced immune activation and altered T cell homing, with potential ramifications regarding T cell responses and the acquisition of HIV-1 as an infant.
- Published
- 2014
- Full Text
- View/download PDF
33. Two independent killing mechanisms of Candida albicans by human neutrophils: evidence from innate immunity defects.
- Author
-
Gazendam RP, van Hamme JL, Tool AT, van Houdt M, Verkuijlen PJ, Herbst M, Liese JG, van de Veerdonk FL, Roos D, van den Berg TK, and Kuijpers TW
- Subjects
- Candida albicans growth & development, Candidiasis immunology, Candidiasis microbiology, Cells, Cultured, Humans, Intracellular Signaling Peptides and Proteins metabolism, Lectins, C-Type antagonists & inhibitors, Lectins, C-Type genetics, Lectins, C-Type metabolism, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases genetics, NADPH Oxidases metabolism, Phagocytosis, Protein Kinase C antagonists & inhibitors, Protein Kinase C genetics, Protein Kinase C metabolism, Protein-Tyrosine Kinases metabolism, Reactive Oxygen Species metabolism, Receptors, IgG antagonists & inhibitors, Receptors, IgG genetics, Receptors, IgG metabolism, Syk Kinase, Candida albicans immunology, Candidiasis prevention & control, Immunity, Innate immunology, Neutrophils immunology
- Abstract
Invasive fungal infections, accompanied by high rates of mortality, represent an increasing problem in medicine. Neutrophils are the major effector immune cells in fungal killing. Based on studies with neutrophils from patients with defined genetic defects, we provide evidence that human neutrophils use 2 distinct and independent phagolysosomal mechanisms to kill Candida albicans. The first mechanism for the killing of unopsonized C albicans was found to be dependent on complement receptor 3 (CR3) and the signaling proteins phosphatidylinositol-3-kinase and caspase recruitment domain-containing protein 9 (CARD9), but was independent of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity. The second mechanism for the killing of opsonized C albicans was strictly dependent on Fcγ receptors, protein kinase C (PKC), and reactive oxygen species production by the NADPH oxidase system. Each of the 2 pathways of Candida killing required Syk tyrosine kinase activity, but dectin-1 was dispensable for both of them. These data provide an explanation for the variable clinical presentation of fungal infection in patients suffering from different immune defects, including dectin-1 deficiency, CARD9 deficiency, or chronic granulomatous disease., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
34. Defects in neutrophil granule mobilization and bactericidal activity in familial hemophagocytic lymphohistiocytosis type 5 (FHL-5) syndrome caused by STXBP2/Munc18-2 mutations.
- Author
-
Zhao XW, Gazendam RP, Drewniak A, van Houdt M, Tool AT, van Hamme JL, Kustiawan I, Meijer AB, Janssen H, Russell DG, van de Corput L, Tesselaar K, Boelens JJ, Kuhnle I, Van Der Werff Ten Bosch J, Kuijpers TW, and van den Berg TK
- Subjects
- Cell Degranulation genetics, Cell Degranulation immunology, Cytoplasmic Granules metabolism, Cytoplasmic Granules microbiology, Escherichia coli immunology, Escherichia coli Infections genetics, Escherichia coli Infections immunology, Female, Gastroenteritis genetics, Genetic Predisposition to Disease, Humans, Killer Cells, Natural immunology, Killer Cells, Natural microbiology, Lymphohistiocytosis, Hemophagocytic genetics, Lymphohistiocytosis, Hemophagocytic microbiology, Male, Neutrophils microbiology, Staphylococcal Infections genetics, Staphylococcal Infections immunology, Staphylococcus aureus immunology, Gastroenteritis immunology, Lymphohistiocytosis, Hemophagocytic immunology, Munc18 Proteins genetics, Munc18 Proteins immunology, Neutrophils immunology
- Abstract
Familial hemophagocytic lymphohistiocytosis (FHL) is caused by genetic defects in cytotoxic granule components or their fusion machinery, leading to impaired natural killer cell and/or T lymphocyte degranulation and/or cytotoxicity. This may accumulate into a life-threatening condition known as macrophage activation syndrome. STXBP2, also known as MUNC18-2, has recently been identified as the disease-causing gene in FHL type 5 (FHL-5). A role for STXBP2 in neutrophils, and for neutrophils in FHL in general, has not been documented thus far. Here, we report that FHL-5 neutrophils have a profound defect in granule mobilization, resulting in inadequate bacterial killing, in particular, of gram-negative Escherichia coli, but not of Staphylococcus aureus, which rather depends on intact reduced NAD phosphate oxidase activity. This impairment of bacterial killing may contribute to the apparent susceptibility to gastrointestinal tract inflammation in patients with FHL-5.
- Published
- 2013
- Full Text
- View/download PDF
35. Invasive fungal infection and impaired neutrophil killing in human CARD9 deficiency.
- Author
-
Drewniak A, Gazendam RP, Tool AT, van Houdt M, Jansen MH, van Hamme JL, van Leeuwen EM, Roos D, Scalais E, de Beaufort C, Janssen H, van den Berg TK, and Kuijpers TW
- Subjects
- Adolescent, Brain Diseases diagnosis, Brain Diseases etiology, Brain Diseases immunology, CARD Signaling Adaptor Proteins immunology, Candida albicans immunology, Candida albicans isolation & purification, Candidiasis, Invasive complications, Candidiasis, Invasive genetics, Cells, Cultured, Cytophagocytosis genetics, Cytophagocytosis immunology, Female, Humans, Immunity, Innate genetics, Immunity, Innate physiology, CARD Signaling Adaptor Proteins deficiency, CARD Signaling Adaptor Proteins genetics, Candidiasis, Invasive immunology, Neutrophils immunology
- Abstract
Caspase recruitment domain-containing protein 9 (CARD9) is an adaptor molecule in the cytosol of myeloid cells, required for induction of T-helper cells producing interleukin-17 (Th17 cells) and important in antifungal immunity. In a patient suffering from Candida dubliniensis meningoencephalitis, mutations in the CARD9 gene were found to result in the loss of protein expression. Apart from the reduced numbers of CD4(+) Th17 lymphocytes, we identified a lack of monocyte-derived cytokines in response to Candida strains. Importantly, CARD9-deficient neutrophils showed a selective Candida albicans killing defect with abnormal ultrastructural phagolysosomes and outgrowth of hyphae. The neutrophil killing defect was independent of the generation of reactive oxygen species by the reduced NAD phosphate oxidase system. Taken together, this demonstrates that human CARD9 deficiency results in selective defect in the host defense against invasive fungal infection, caused by an impaired phagocyte killing.
- Published
- 2013
- Full Text
- View/download PDF
36. Memory CD4(+)CCR5(+) T cells are abundantly present in the gut of newborn infants to facilitate mother-to-child transmission of HIV-1.
- Author
-
Bunders MJ, van der Loos CM, Klarenbeek PL, van Hamme JL, Boer K, Wilde JC, de Vries N, van Lier RA, Kootstra N, Pals ST, and Kuijpers TW
- Subjects
- Adult, CD4 Lymphocyte Count, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Cells, Cultured, Female, HIV Infections immunology, HIV-1 immunology, Humans, Infant, Newborn, Intestinal Mucosa cytology, Intestinal Mucosa immunology, Intestinal Mucosa metabolism, Intestines cytology, Intestines virology, Male, Mother-Child Relations, Pregnancy, Pregnancy Complications, Infectious immunology, CD4-Positive T-Lymphocytes cytology, HIV Infections transmission, Immunologic Memory immunology, Immunologic Memory physiology, Infectious Disease Transmission, Vertical, Intestines immunology, Receptors, CCR5 metabolism
- Abstract
Despite potential clinical importance, target cells for mother-to-child transmission of HIV-1 have not yet been identified. Cord blood-derived CD4(+) T cells are largely naive and do not express CCR5, the mandatory coreceptor for transmitted HIV-1 R5 strains in infants. In the present study, we demonstrate that in the human fetal and infant gut mucosa, there is already a large subset of mucosal memory CD4(+)CCR5(+) T cells with predominantly a Th1 and Th17 phenotype. Using next-generation sequencing of the TCRβ chain, clonally expanded T cells as a hallmark for memory development predominated in the gut mucosa (30%), whereas few were found in the lymph nodes (1%) and none in cord blood (0%). The gut mucosal fetal and infant CD4(+) T cells were highly susceptible to HIV-1 without any prestimulation; pol proviral DNA levels were similar to infected phytohemagglutinin-stimulated adult PBMCs. In conclusion, in the present study, we show that extensive adaptive immunity is present before birth and the gut mucosa is the preferential site for memory CD4(+) T cells. These CD4(+)CCR5(+) T cells in the infant mucosa provide a large pool of susceptible cells for ingested HIV-1 at birth and during breastfeeding, indicating a mucosal route of mother-to-child transmission that can be targeted in prevention strategies.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.