Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
EMBO J ; 43(7): 1135-1163, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38418557

RESUMEN

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.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Anticuerpos Antivirales , Proteínas del Sistema Complemento , Inflamación , Inmunidad Innata
2.
EMBO J ; 41(19): e110629, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-35968812

RESUMEN

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.


Asunto(s)
Infecciones por VIH , VIH-1 , Antivirales , Disbiosis , Femenino , Humanos , Células de Langerhans , Prevotella
3.
EMBO J ; 40(20): e106765, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34510494

RESUMEN

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.


Asunto(s)
COVID-19/transmisión , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina de Bajo-Peso-Molecular/farmacología , SARS-CoV-2/patogenicidad , Enzima Convertidora de Angiotensina 2/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Neutralizantes/farmacología , Chlorocebus aethiops , Células Dendríticas/metabolismo , Células Dendríticas/virología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Interacciones Huésped-Patógeno , Humanos , Membrana Mucosa/citología , Membrana Mucosa/virología , SARS-CoV-2/metabolismo , Sindecano-1/metabolismo , Sindecano-4/metabolismo , Células Vero , Tratamiento Farmacológico de COVID-19
4.
PLoS Pathog ; 19(10): e1011735, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37844099

RESUMEN

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.


Asunto(s)
COVID-19 , Sobreinfección , Humanos , SARS-CoV-2/metabolismo , Receptor Toll-Like 4/metabolismo , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , COVID-19/metabolismo , Lectinas Tipo C/metabolismo , Citocinas/metabolismo , Células Dendríticas
5.
J Infect Dis ; 230(1): e43-e47, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052703

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos , Disbiosis , Infecciones por VIH , VIH-1 , Prevotella , Vagina , Humanos , Femenino , Prevotella/aislamiento & purificación , Disbiosis/microbiología , Vagina/microbiología , Vagina/virología , Vagina/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/microbiología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Susceptibilidad a Enfermedades , Microbiota , Internalización del Virus
6.
Eur J Immunol ; 52(4): 646-655, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35099061

RESUMEN

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.


Asunto(s)
COVID-19 , Células Dendríticas , Glicoproteína de la Espiga del Coronavirus , Receptor Toll-Like 4 , COVID-19/inmunología , Línea Celular , Células Dendríticas/inmunología , Humanos , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/inmunología , Receptor Toll-Like 4/inmunología
7.
Eur J Immunol ; 51(10): 2464-2477, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34223639

RESUMEN

The proinflammatory cytokine IL-1ß 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-1ß activation during HIV-1 infection. Here, we have identified a crucial role for abortive HIV-1 RNAs in inducing IL-1ß 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-1ß 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-1ß 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.


Asunto(s)
Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , ARN Viral/metabolismo , eIF-2 Quinasa/metabolismo , Interacciones Huésped-Patógeno , Humanos , Sistema de Señalización de MAP Quinasas , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ARN Viral/genética , Especies Reactivas de Oxígeno/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal
8.
Nature ; 540(7633): 448-452, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27919079

RESUMEN

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.


Asunto(s)
Antígenos CD/metabolismo , Autofagia , Proteínas Portadoras/metabolismo , VIH-1/fisiología , Células de Langerhans/metabolismo , Células de Langerhans/virología , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Receptores del VIH/metabolismo , Factores de Restricción Antivirales , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/transmisión , VIH-1/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Mucosa , Células de Langerhans/citología , Células de Langerhans/inmunología , Receptores de Superficie Celular/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas
9.
PLoS Pathog ; 13(11): e1006738, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29186193

RESUMEN

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


Asunto(s)
Anticuerpos Antivirales/inmunología , Virus del Dengue/fisiología , Dengue/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Formación de Anticuerpos , Linfocitos B/inmunología , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/inmunología , Células Dendríticas/inmunología , Dengue/genética , Dengue/virología , Humanos , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/inmunología , Interleucina-27/genética , Interleucina-27/inmunología , Interleucinas/genética , Interleucinas/inmunología , Activación de Linfocitos , Receptores Inmunológicos
10.
J Immunol ; 198(12): 4764-4771, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28507028

RESUMEN

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


Asunto(s)
Proteína 58 DEAD Box/inmunología , Células Dendríticas/inmunología , Virus del Dengue/inmunología , Células TH1/inmunología , Diferenciación Celular , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiocina CCL3/genética , Quimiocina CCL3/inmunología , Quimiocina CCL4/genética , Quimiocina CCL4/inmunología , Proteína 58 DEAD Box/deficiencia , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Células Dendríticas/virología , Humanos , Helicasa Inducida por Interferón IFIH1/deficiencia , Helicasa Inducida por Interferón IFIH1/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Interleucina-6/inmunología , Interleucina-6/metabolismo , Receptores Inmunológicos , Células TH1/fisiología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
11.
J Immunol ; 196(3): 1272-83, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26718340

RESUMEN

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.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Hifa/inmunología , Neutrófilos/inmunología , Esporas Fúngicas/inmunología , Citotoxicidad Inmunológica/inmunología , Trampas Extracelulares/inmunología , Humanos , Inmunidad Innata , Síndromes de Inmunodeficiencia/inmunología , Microscopía Confocal , Fagocitos/inmunología
12.
Transfusion ; 57(3): 674-684, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28032635

RESUMEN

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.


Asunto(s)
Dexametasona , Transfusión de Leucocitos , Neutrófilos/metabolismo , Adulto , Anexina A5/metabolismo , Supervivencia Celular/efectos de los fármacos , Dexametasona/administración & dosificación , Dexametasona/farmacocinética , Filgrastim/administración & dosificación , Filgrastim/farmacocinética , Proteínas Ligadas a GPI/metabolismo , Semivida , Humanos , Masculino , Persona de Mediana Edad , Mifepristona/farmacología , Neutrófilos/citología , Plasma/metabolismo , Receptores CXCR4/metabolismo , Receptores de IgG/metabolismo
13.
Blood ; 124(4): 590-7, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-24948657

RESUMEN

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.


Asunto(s)
Candida albicans/inmunología , Candidiasis/prevención & control , Inmunidad Innata/inmunología , Neutrófilos/inmunología , Candida albicans/crecimiento & desarrollo , Candidiasis/inmunología , Candidiasis/microbiología , Células Cultivadas , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lectinas Tipo C/antagonistas & inhibidores , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Fagocitosis , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de IgG/antagonistas & inhibidores , Receptores de IgG/genética , Receptores de IgG/metabolismo , Quinasa Syk
14.
Haematologica ; 101(5): 587-96, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26802050

RESUMEN

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.


Asunto(s)
Candida albicans/inmunología , Citotoxicidad Inmunológica , Granulocitos/inmunología , Transfusión de Leucocitos , Viabilidad Microbiana/inmunología , Biomarcadores , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Gránulos Citoplasmáticos/inmunología , Gránulos Citoplasmáticos/metabolismo , Dexametasona/farmacología , Factor Estimulante de Colonias de Granulocitos/farmacología , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Granulocitos/microbiología , Humanos , Inmunofenotipificación , NADPH Oxidasas/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Fagosomas/inmunología , Fagosomas/microbiología
15.
Blood ; 121(13): 2385-92, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23335372

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/deficiencia , Proteínas Adaptadoras de Señalización CARD/genética , Candidiasis Invasiva/inmunología , Neutrófilos/inmunología , Adolescente , Encefalopatías/diagnóstico , Encefalopatías/etiología , Encefalopatías/inmunología , Proteínas Adaptadoras de Señalización CARD/inmunología , Candida albicans/inmunología , Candida albicans/aislamiento & purificación , Candidiasis Invasiva/complicaciones , Candidiasis Invasiva/genética , Células Cultivadas , Citofagocitosis/genética , Citofagocitosis/inmunología , Femenino , Humanos , Inmunidad Innata/genética , Inmunidad Innata/fisiología
16.
Blood ; 122(1): 109-11, 2013 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-23687090

RESUMEN

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.


Asunto(s)
Gastroenteritis/inmunología , Linfohistiocitosis Hemofagocítica/inmunología , Proteínas Munc18/genética , Proteínas Munc18/inmunología , Neutrófilos/inmunología , Degranulación de la Célula/genética , Degranulación de la Célula/inmunología , Gránulos Citoplasmáticos/metabolismo , Gránulos Citoplasmáticos/microbiología , Escherichia coli/inmunología , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/inmunología , Femenino , Gastroenteritis/genética , Predisposición Genética a la Enfermedad , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/microbiología , Linfohistiocitosis Hemofagocítica/genética , Linfohistiocitosis Hemofagocítica/microbiología , Masculino , Neutrófilos/microbiología , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología
17.
Blood ; 120(22): 4383-90, 2012 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-23033270

RESUMEN

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 TCRß 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.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Infecciones por VIH/transmisión , Memoria Inmunológica , Transmisión Vertical de Enfermedad Infecciosa , Intestinos/inmunología , Receptores CCR5/metabolismo , Adulto , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Femenino , Infecciones por VIH/inmunología , VIH-1/inmunología , Humanos , Memoria Inmunológica/inmunología , Memoria Inmunológica/fisiología , Recién Nacido , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Intestinos/citología , Intestinos/virología , Masculino , Relaciones Madre-Hijo , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología
18.
J Invest Dermatol ; 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37979773

RESUMEN

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.

19.
J Immunother Cancer ; 11(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37072345

RESUMEN

BACKGROUND: Interferon (IFN)-ß 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-ß 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-ß 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-ß 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-ß 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.


Asunto(s)
Interferón Tipo I , Neoplasias , Humanos , Salmonella typhimurium/metabolismo , Expresión Génica Ectópica , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Macrófagos/metabolismo , Neoplasias/metabolismo , Células Dendríticas/metabolismo , Microambiente Tumoral
20.
Sci Rep ; 13(1): 8851, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37258559

RESUMEN

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.


Asunto(s)
COVID-19 , Nanopartículas , Humanos , Vacunas contra la COVID-19 , SARS-CoV-2/genética , COVID-19/prevención & control , ARN Mensajero/genética , Vacunas de ARNm
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA