Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 32(4): 1061-1079, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38382529

RESUMEN

Complement-mediated diseases can be treated using systemic inhibitors. However, complement components are abundant in circulation, affecting systemic inhibitors' exposure and efficacy. Furthermore, because of complement's essential role in immunity, systemic treatments raise infection risk in patients. To address these challenges, we developed antibody fusion proteins combining the alternative-pathway complement inhibitor factor H (fH1-5) with an anti-C3d monoclonal antibody (C3d-mAb-2fH). Because C3d is deposited at sites of complement activity, this molecule localizes to tissue complement while minimizing circulating complement engagement. These fusion proteins bind to deposited complement in diseased human skin sections and localize to activated complement in a primate skin injury model. We further explored the pharmacology of C3d-mAb-2fH proteins in rodent models with robust tissue complement activation. Doses of C3d-mAb-2fH >1 mg/kg achieved >75% tissue complement inhibition in mouse and rat injury models while avoiding circulating complement blockade. Glomerular-specific complement inhibition reduced proteinuria and preserved podocyte foot-process architecture in rat membranous nephropathy, indicating disease-modifying efficacy. These data indicate that targeting local tissue complement results in durable and efficacious complement blockade in skin and kidney while avoiding systemic inhibition, suggesting broad applicability of this approach in treating a range of complement-mediated diseases.


Asunto(s)
Factor H de Complemento , Enfermedades Renales , Humanos , Ratones , Ratas , Animales , Factor H de Complemento/genética , Complemento C3d/metabolismo , Enfermedades Renales/etiología , Anticuerpos , Activación de Complemento
2.
Front Immunol ; 14: 1290272, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38054006

RESUMEN

Historically platelets are mostly known for their crucial contribution to hemostasis, but there is growing understanding of their role in inflammation and immunity. The immunomodulatory role of platelets entails interaction with pathogens, but also with immune cells including macrophages and dendritic cells (DCs), to activate adaptive immune responses. In our previous work, we have demonstrated that splenic CD169+ macrophages scavenge liposomes and collaborate with conventional type 1 DCs (cDC1) to induce expansion of CD8+ T cells. Here, we show that platelets associate with liposomes and bind to DNGR-1/Clec9a and CD169/Siglec-1 receptors in vitro. In addition, platelets interacted with splenic CD169+ macrophages and cDC1 and further increased liposome internalization by cDC1. Most importantly, platelet depletion prior to liposomal immunization resulted in significantly diminished antigen-specific CD8+ T cell responses, but not germinal center B cell responses. Previously, complement C3 was shown to be essential for platelet-mediated CD8+ T cell activation during bacterial infection. However, after liposomal vaccination CD8+ T cell priming was not dependent on complement C3. While DCs from platelet-deficient mice exhibited unaltered maturation status, they did express lower levels of CCR7. In addition, in the absence of platelets, CCL5 plasma levels were significantly reduced. Overall, our findings demonstrate that platelets engage in a cross-talk with CD169+ macrophages and cDC1 and emphasize the importance of platelets in induction of CD8+ T cell responses in the context of liposomal vaccination.


Asunto(s)
Linfocitos T CD8-positivos , Liposomas , Animales , Ratones , Liposomas/metabolismo , Complemento C3/metabolismo , Macrófagos , Antígenos
3.
Biomed Pharmacother ; 167: 115467, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37696087

RESUMEN

1,8-cineole (Eucalyptol), a naturally occurring compound derived from botanical sources such as eucalyptus, rosemary, and camphor laurel, has a long history of use in traditional medicine and exhibits an array of biological properties, including anti-inflammatory, antioxidant, antimicrobial, bronchodilatory, analgesic, and pro-apoptotic effects. Recent evidence has also indicated its potential role in managing conditions such as Alzheimer's disease, neuropathic pain, and cancer. This review spotlights the health advantages of 1,8-cineole, as demonstrated in clinical trials involving patients with respiratory disorders, including chronic obstructive pulmonary disease, asthma, bronchitis, and rhinosinusitis. In addition, we shed light on potential therapeutic applications of 1,8-cineole in various conditions, such as depression, epilepsy, peptic ulcer disease, diarrhea, cardiac-related heart diseases, and diabetes mellitus. A comprehensive understanding of 1,8-cineole's pharmacodynamics and safety aspects as well as developing effective formulations, might help to leverage its therapeutic value. This thorough review sets the stage for future research on diverse health benefits and potential uses of 1,8-cineole in tackling complex medical conditions.

4.
Sci Rep ; 13(1): 274, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36609665

RESUMEN

The complement system provides vital immune protection against infectious agents by labeling them with complement fragments that enhance phagocytosis by immune cells. Many details of complement-mediated phagocytosis remain elusive, partly because it is difficult to study the role of individual complement proteins on target surfaces. Here, we employ serum-free methods to couple purified complement C3b onto E. coli bacteria and beads and then expose human neutrophils to these C3b-coated targets. We examine the neutrophil response using a combination of flow cytometry, confocal microscopy, luminometry, single-live-cell/single-target manipulation, and dynamic analysis of neutrophil spreading on opsonin-coated surfaces. We show that purified C3b can potently trigger phagocytosis and killing of bacterial cells via Complement receptor 1. Comparison of neutrophil phagocytosis of C3b- versus antibody-coated beads with single-bead/single-target analysis exposes a similar cell morphology during engulfment. However, bulk phagocytosis assays of C3b-beads combined with DNA-based quenching reveal that these are poorly internalized compared to their IgG1 counterparts. Similarly, neutrophils spread slower on C3b-coated compared to IgG-coated surfaces. These observations support the requirement of multiple stimulations for efficient C3b-mediated uptake. Together, our results establish the existence of a direct pathway of phagocytic uptake of C3b-coated targets and present methodologies to study this process.


Asunto(s)
Complemento C3b , Neutrófilos , Humanos , Neutrófilos/metabolismo , Complemento C3b/metabolismo , Escherichia coli/metabolismo , Fagocitosis , Receptores de Complemento 3b/metabolismo , Proteínas del Sistema Complemento/metabolismo , Inmunoglobulina G , Receptores de Complemento/metabolismo
5.
Front Immunol ; 13: 973702, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36059476

RESUMEN

Pemphigoid diseases are autoimmune chronic inflammatory skin diseases, which are characterized by blistering of the skin and/or mucous membranes, and circulating and tissue-bound autoantibodies. The well-established pathomechanisms comprise autoantibodies targeting various structural proteins located at the dermal-epidermal junction, leading to complement factor binding and activation. Several effector cells are thus attracted and activated, which in turn inflict characteristic tissue damage and subepidermal blistering. Moreover, the detection of linear complement deposits in the skin is a diagnostic hallmark of all pemphigoid diseases. However, recent studies showed that blistering might also occur independently of complement. This review reassesses the importance of complement in pemphigoid diseases based on current research by contrasting and contextualizing data from in vitro, murine and human studies.


Asunto(s)
Penfigoide Ampolloso , Animales , Autoanticuerpos , Vesícula , Proteínas del Sistema Complemento , Humanos , Ratones , Piel
7.
Platelets ; 33(8): 1192-1198, 2022 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-35701857

RESUMEN

We aimed to investigate the effects of integrin αIIbß3 inhibitor tirofiban on hallmarks of platelet activation, degranulation, and aggregation during its use to analyze activated but non-complexed platelets via flow cytometry. To do so, we used washed platelets from healthy human donors. We combined aggregometry, an assay of platelet functionality, with flow cytometry and ELISA to detect and correlate, respectively, platelet aggregation, activation, and granule release. While tirofiban effectively inhibited agonist-induced platelet aggregation (thrombin receptor-activating peptide 6 (TRAP), convulxin (CVX), U46619 and IV.3), the surface expression of P-selectin and CD63 and granule release of RANTES were significantly increased, indicating that tirofiban enhances degranulation, uncoupled from aggregation. The results show that tirofiban alters the activation phenotype of platelets, something that should be considered when using tirofiban to enable flow cytometric analysis of activated but unaggregated platelet suspensions.


Asunto(s)
Selectina-P , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Plaquetas/metabolismo , Quimiocina CCL5/metabolismo , Quimiocina CCL5/farmacología , Humanos , Selectina-P/metabolismo , Activación Plaquetaria , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/metabolismo , Inhibidores de Agregación Plaquetaria/farmacología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Receptores de Trombina/metabolismo , Tirofibán/farmacología , Tirosina/metabolismo , Tirosina/farmacología
8.
Nat Commun ; 12(1): 3352, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099640

RESUMEN

Platelets contribute to the regulation of tissue neovascularization, although the specific factors underlying this function are unknown. Here, we identified the complement anaphylatoxin C5a-mediated activation of C5a receptor 1 (C5aR1) on platelets as a negative regulatory mechanism of vessel formation. We showed that platelets expressing C5aR1 exert an inhibitory effect on endothelial cell functions such as migration and 2D and 3D tube formation. Growth factor- and hypoxia-driven vascularization was markedly increased in C5ar1-/- mice. Platelet-specific deletion of C5aR1 resulted in a proangiogenic phenotype with increased collateralization, capillarization and improved pericyte coverage. Mechanistically, we found that C5a induced preferential release of CXC chemokine ligand 4 (CXCL4, PF4) from platelets as an important antiangiogenic paracrine effector molecule. Interfering with the C5aR1-CXCL4 axis reversed the antiangiogenic effect of platelets both in vitro and in vivo.In conclusion, we identified a mechanism for the control of tissue neovascularization through C5a/C5aR1 axis activation in platelets and subsequent induction of the antiangiogenic factor CXCL4.


Asunto(s)
Plaquetas/metabolismo , Factor Plaquetario 4/metabolismo , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo , Inductores de la Angiogénesis , Animales , Activación de Complemento , Complemento C5a , Inflamación , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia , Receptores CXCR3/genética , Transducción de Señal
9.
Front Immunol ; 12: 635622, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33968028

RESUMEN

Introduction: Schistosomiasis is a neglected tropical disease (NTD) caused by blood-dwelling flatworms which develop from skin-penetrating cercariae, the freely swimming water-borne infective stage of Schistosoma mansoni, into adult worms. This natural course of infection can be mimicked in experimental mouse models of schistosomiasis. However, only a maximum of 20-30% of penetrated cercariae mature into fecund adults. The reasons for this are unknown but could potentially involve soluble factors of the innate immune system, such as complement factors and preexisting, natural antibodies. Materials and Methods: Using our recently developed novel serum- and cell-free in vitro culture system for newly transformed schistosomula (NTS), which supports long-term larval survival, we investigated the effects of mouse serum and its major soluble complement factors C1q, C3, C4 as well as preexisting, natural IgM in vitro and assessed worm development in vivo by infecting complement and soluble (s)IgM-deficient animals. Results: In contrast to sera from humans and a broad variety of mammalian species, serum from mice, surprisingly, killed parasites already at skin stage in vitro. Interestingly, the most efficient killing component(s) were heat-labile but did not include important members of the perhaps best known family of heat-labile serum factors, the complement system, nor consisted of complement-activating natural immunoglobulins. Infection of complement C1q and sIgM-deficient mice with S. mansoni as well as in vitro tests with sera from mice deficient in C3 and C4 revealed no major role for these soluble factors in vivo in regard to parasite maturation, fecundity and associated immunopathology. Rather, the reduction of parasite maturation from cercariae to adult worms was comparable to wild-type mice. Conclusion: This study reveals that not yet identified heat-labile serum factors are major selective determinants of the host-specificity of schistosomiasis, by directly controlling schistosomal development and survival.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , Inmunoglobulina M/sangre , Schistosoma mansoni/crecimiento & desarrollo , Esquistosomiasis mansoni/sangre , Esquistosomiasis mansoni/parasitología , Animales , Complemento C1q/genética , Complemento C1q/metabolismo , Complemento C3/genética , Complemento C3/metabolismo , Complemento C4/genética , Complemento C4/metabolismo , Proteínas del Sistema Complemento/genética , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Parásitos , Humanos , Macaca mulatta , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Especificidad de la Especie
11.
Int J Mol Sci ; 21(20)2020 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-33050608

RESUMEN

Allergic asthma is a chronical pulmonary disease with high prevalence. It manifests as a maladaptive immune response to common airborne allergens and is characterized by airway hyperresponsiveness, eosinophilia, type 2 cytokine-associated inflammation, and mucus overproduction. Alveolar macrophages (AMs), although contributing to lung homeostasis and tolerance to allergens at steady state, have attracted less attention compared to professional antigen-presenting and adaptive immune cells in their contributions. Using an acute model of house dust mite-driven allergic asthma in mice, we showed that a fraction of resident tissue-associated AMs, while polarizing to the alternatively activated M2 phenotype, exhibited signs of polynucleation and polyploidy. Mechanistically, in vitro assays showed that only Granulocyte-Macrophage Colony Stimulating Factor and interleukins IL-13 and IL-33, but not IL-4 or IL-5, participate in the establishment of this phenotype, which resulted from division defects and not cell-cell fusion as shown by microscopy. Intriguingly, mRNA analysis of AMs isolated from allergic asthmatic lungs failed to show changes in the expression of genes involved in DNA damage control except for MafB. Altogether, our data support the idea that upon allergic inflammation, AMs undergo DNA damage-induced stresses, which may provide new unconventional therapeutical approaches to treat allergic asthma.


Asunto(s)
Asma/etiología , Asma/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Interleucina-33/farmacología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Poliploidía , Animales , Asma/patología , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Expresión Génica , Células Gigantes/efectos de los fármacos , Células Gigantes/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Activación de Macrófagos , Macrófagos Alveolares/citología , Ratones
12.
PLoS Negl Trop Dis ; 13(1): e0006590, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30689639

RESUMEN

BACKGROUND: The arsenal in anthelminthic treatment against schistosomiasis is limited and relies almost exclusively on a single drug, praziquantel (PZQ). Thus, resistance to PZQ could constitute a major threat. Even though PZQ is potent in killing adult worms, its activity against earlier stages is limited. Current in vitro drug screening strategies depend on newly transformed schistosomula (NTS) for initial hit identification, thereby limiting sensitivity to new compounds predominantly active in later developmental stages. Therefore, the aim of this study was to establish a highly standardized, straightforward and reliable culture method to generate and maintain advanced larval stages in vitro. We present here how this method can be a valuable tool to test drug efficacy at each intermediate larval stage, reducing the reliance on animal use (3Rs). METHODOLOGY/PRINCIPAL FINDINGS: Cercariae were mechanically transformed into skin-stage (SkS) schistosomula and successfully cultured for up to four weeks with no loss in viability in a commercially available medium. Under these serum- and cell-free conditions, development halted at the lung-stage (LuS). However, the addition of human serum (HSe) propelled further development into liver stage (LiS) worms within eight weeks. Skin and lung stages, as well as LiS, were submitted to 96-well drug screening assays using known anti-schistosomal compounds such as PZQ, oxamniquine (OXM), mefloquine (MFQ) and artemether (ART). Our findings showed stage-dependent differences in larval susceptibility to these compounds. CONCLUSION: With this robust and highly standardized in vitro assay, important developmental stages of S. mansoni up to LiS worms can be generated and maintained over prolonged periods of time. The phenotype of LiS worms, when exposed to reference drugs, was comparable to most previously published works for ex vivo harvested adult worms. Therefore, this in vitro assay can help reduce reliance on animal experiments in search for new anti-schistosomal drugs.


Asunto(s)
Schistosoma mansoni/efectos de los fármacos , Schistosoma mansoni/crecimiento & desarrollo , Esquistosomicidas/farmacología , Animales , Arteméter/farmacología , Cercarias/efectos de los fármacos , Cercarias/crecimiento & desarrollo , Medio de Cultivo Libre de Suero/química , Medio de Cultivo Libre de Suero/farmacología , Evaluación Preclínica de Medicamentos , Humanos , Mefloquina/farmacología , Oxamniquina/farmacología , Praziquantel/farmacología , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomicidas/aislamiento & purificación
13.
Semin Immunol ; 37: 4-11, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29573978

RESUMEN

Rapid elimination of microbes from the bloodstream, along with the ability to mount an adaptive immune response, are essential for optimal host-defense. Kupffer cells are strategically positioned in the liver sinusoids and efficiently capture circulating microbes from the hepatic artery and portal vein, thus preventing bacterial dissemination. In vivo and in vitro studies have probed how complement receptor of the immunoglobulin superfamily (CRIg), also referred to as Z39Ig and V-set and Ig domain-containing 4 (VSIG4), acts as a critical player in pathogen recognition and clearance. While recent data suggested that CRIg may bind bacterial cell wall components directly, the single transmembrane receptor is best known for its interaction with complement C3 opsonization products on the microbial surface. On Kupffer cells, CRIg must capture opsonized microbes against the shear forces of the blood flow. In vivo work reveals how immune adherence (IA), a process in which blood platelets or erythrocytes associate with circulating bacteria, plays a critical role in regulating pathogen capture by CRIg under flow conditions. In addition to its typical innate immune functions, CRIg was shown to directly and indirectly influence adaptive immune responses. Here, we review our current understanding of the diverse roles of CRIg in pathogen elimination, anti-microbial immunity and autoimmunity. In particular, we will explore how, through selective capturing by CRIg, an important balance is achieved between the immunological and clearance functions of liver and spleen.


Asunto(s)
Infecciones Bacterianas/inmunología , Macrófagos del Hígado/fisiología , Proteínas Opsoninas/metabolismo , Receptores de Complemento/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Aglutinación , Animales , Complemento C3/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inmunomodulación , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología
14.
J Allergy Clin Immunol ; 142(5): 1558-1570, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29382591

RESUMEN

BACKGROUND: Nanoparticle (NP)-based vaccines are attractive immunotherapy tools because of their capability to codeliver antigen and adjuvant to antigen-presenting cells. Their cellular distribution and serum protein interaction ("protein corona") after systemic administration and their effect on the functional properties of NPs is poorly understood. OBJECTIVES: We analyzed the relevance of the protein corona on cell type-selective uptake of dextran-coated NPs and determined the outcome of vaccination with NPs that codeliver antigen and adjuvant in disease models of allergy. METHODS: The role of protein corona constituents for cellular binding/uptake of dextran-coated ferrous nanoparticles (DEX-NPs) was analyzed both in vitro and in vivo. DEX-NPs conjugated with the model antigen ovalbumin (OVA) and immunostimulatory CpG-rich oligodeoxynucleotides were administered to monitor the induction of cellular and humoral immune responses. Therapeutic effects of this DEX-NP vaccine in mouse models of OVA-induced anaphylaxis and allergic asthma were assessed. RESULTS: DEX-NPs triggered lectin-induced complement activation, yielding deposition of activated complement factor 3 on the DEX-NP surface. In the spleen DEX-NPs targeted predominantly B cells through complement receptors 1 and 2. The DEX-NP vaccine elicited much stronger OVA-specific IgG2a production than coadministered soluble OVA plus CpG oligodeoxynucleotides. B-cell binding of the DEX-NP vaccine was critical for IgG2a production. Treatment of OVA-sensitized mice with the DEX-NP vaccine prevented induction of anaphylactic shock and allergic asthma accompanied by IgE inhibition. CONCLUSIONS: Opsonization of lectin-coated NPs by activated complement components results in selective B-cell targeting. The intrinsic B-cell targeting property of lectin-coated NPs can be exploited for treatment of allergic immune responses.


Asunto(s)
Anafilaxia/inmunología , Linfocitos B/inmunología , Hipersensibilidad/inmunología , Nanopartículas/administración & dosificación , Corona de Proteínas/inmunología , Animales , Antígenos/administración & dosificación , Dextranos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Femenino , Compuestos Ferrosos/administración & dosificación , Lectinas/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligodesoxirribonucleótidos/administración & dosificación , Ovalbúmina/administración & dosificación , Linfocitos T/inmunología , Vacunas/administración & dosificación
15.
Cell ; 171(6): 1368-1382.e23, 2017 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-29195076

RESUMEN

Blood platelets are critical for hemostasis and thrombosis and play diverse roles during immune responses. Despite these versatile tasks in mammalian biology, their skills on a cellular level are deemed limited, mainly consisting in rolling, adhesion, and aggregate formation. Here, we identify an unappreciated asset of platelets and show that adherent platelets use adhesion receptors to mechanically probe the adhesive substrate in their local microenvironment. When actomyosin-dependent traction forces overcome substrate resistance, platelets migrate and pile up the adhesive substrate together with any bound particulate material. They use this ability to act as cellular scavengers, scanning the vascular surface for potential invaders and collecting deposited bacteria. Microbe collection by migrating platelets boosts the activity of professional phagocytes, exacerbating inflammatory tissue injury in sepsis. This assigns platelets a central role in innate immune responses and identifies them as potential targets to dampen inflammatory tissue damage in clinical scenarios of severe systemic infection.


Asunto(s)
Infecciones Bacterianas/inmunología , Plaquetas/inmunología , Animales , Bacterias/clasificación , Plaquetas/citología , Vasos Sanguíneos/lesiones , Vasos Sanguíneos/patología , Calcio/metabolismo , Movimiento Celular , Polaridad Celular , Humanos , Inflamación/inmunología , Integrinas/metabolismo , Ratones , Miosinas/metabolismo , Neutrófilos/citología
17.
Eur J Immunol ; 47(12): 2153-2162, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28833039

RESUMEN

The transfer of regulatory T cells, either freshly isolated, or modified, represents a promising therapeutic approach to dampen misdirected immune responses, like autoimmune diseases, chronic inflammatory syndromes and graft versus host disease. Clinical isolation of highly pure regulatory T cell (Treg) populations is still challenging and labeling reagents can influence their viability and functionality, potentially altering the potency of isolated Treg cell products. Here we show that reversible Fab multimer-based Treg purification can prevent conventional antibody label-induced interferences in vitro and in vivo. Remaining isolation reagents negatively interfere with Treg engraftment efficacy in C57BL/6 wild-type mice due to Fcγ-receptor- as well as IL-2 receptor-mediated mechanisms. Using a preclinical model for acute GvHD, we further show that purified 'label-freed' Tregs are protective at substantially lower cell numbers as compared to conventional nonreversible antibody staining, translating into significantly improved survival of mice treated with minimally manipulated Tregs. These findings have important clinical relevance for future Treg-based cell therapies.


Asunto(s)
Traslado Adoptivo/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Animales , Separación Celular/métodos , Células Cultivadas , Femenino , Citometría de Flujo , Enfermedad Injerto contra Huésped/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especificidad de Órganos/inmunología , Reproducibilidad de los Resultados , Factores de Tiempo
18.
Mol Immunol ; 90: 227-238, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28843904

RESUMEN

The complement system not only plays a critical role in efficient detection and clearance of bacteria, but also in intestinal immune homeostasis as mice deficient for key complement components display enhanced intestinal inflammation upon experimental colitis. Because underlying molecular mechanisms for this observation are unclear, we investigated the crosstalk between intestinal epithelial cells (IEC), bacteria and the complement system in the course of chronic colitis. Surprisingly, mouse intestinal epithelial cell lines constitutively express high mRNA levels of complement component 3 (C3), Toll-like receptor 2 (Tlr2) and Tlr4. Stimulation of these cells with lipopolysaccharide (LPS), but not with flagellin, LD-muramyldipeptide or peptidoglycan, triggered increased C3 expression, secretion and activation. Stimulation of the C3aR on these cell lines with C3a resulted in an increase of LPS-triggered pro-inflammatory response. Tissue biopsies from C57BL/6J mice revealed higher expression of C3, Tlr1, Tlr2 and Tlr4 in colonic primary IECs (pIECs) compared to ileal pIECs, while in germ-free mice no differences in C3 protein expression was observed. In DSS-induced chronic colitis mouse models, C3 mRNA expression was upregulated in colonic biopsies and ileal pIECs with elevated C3 protein in the lamina propria, IECs and the mucus. Notably, increased C3b opsonization of mucosa-attached bacteria and decreased fecal full-length C3 protein was observed in DSS-treated compared to untreated mice. Of significant interest, non-inflamed and inflamed colonic biopsy samples from CD but not UC patients displayed exacerbated C3 expression compared to controls. These findings suggest that a novel TLR4-C3 axis could control the intestinal immune response during chronic colitis.


Asunto(s)
Colitis Ulcerosa/patología , Complemento C3a/biosíntesis , Complemento C3b/biosíntesis , Células Epiteliales/metabolismo , Mucosa Intestinal/patología , Animales , Bacterias/inmunología , Línea Celular , Colitis Ulcerosa/inducido químicamente , Complemento C3a/metabolismo , Complemento C3b/metabolismo , Sulfato de Dextran/toxicidad , Humanos , Inflamación/patología , Mucosa Intestinal/inmunología , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Receptor Toll-Like 1/biosíntesis , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 4/biosíntesis
19.
Sci Rep ; 6: 34440, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27694929

RESUMEN

The mechanisms protecting from immunopathology during acute bacterial infections are incompletely known. We found that in response to apoptotic immune cells and live or dead Listeria monocytogenes scavenger receptor BI (SR-BI), an anti-atherogenic lipid exchange mediator, activated internalization mechanisms with characteristics of macropinocytosis and, assisted by Golgi fragmentation, initiated autophagic responses. This was supported by scavenger receptor-induced local increases in membrane cholesterol concentrations which generated lipid domains particularly in cell extensions and the Golgi. SR-BI was a key driver of beclin-1-dependent autophagy during acute bacterial infection of the liver and spleen. Autophagy regulated tissue infiltration of neutrophils, suppressed accumulation of Ly6C+ (inflammatory) macrophages, and prevented hepatocyte necrosis in the core of infectious foci. Perifocal levels of Ly6C+ macrophages and Ly6C- macrophages were unaffected, indicating predominant regulation of the focus core. SR-BI-triggered autophagy promoted co-elimination of apoptotic immune cells and dead bacteria but barely influenced bacterial sequestration and survival or inflammasome activation, thus exclusively counteracting damage inflicted by immune responses. Hence, SR-BI- and autophagy promote a surveillance pathway that partially responds to products of antimicrobial defenses and selectively prevents immunity-induced damage during acute infection. Our findings suggest that control of infection-associated immunopathology can be based on a unified defense operation.


Asunto(s)
Autofagia/inmunología , Macrófagos/inmunología , Microdominios de Membrana/inmunología , Pinocitosis/inmunología , Receptores Depuradores de Clase B/inmunología , Animales , Autofagia/genética , Beclina-1/genética , Beclina-1/inmunología , Aparato de Golgi/genética , Aparato de Golgi/inmunología , Listeria monocytogenes/inmunología , Listeriosis/genética , Listeriosis/inmunología , Listeriosis/patología , Hepatopatías/genética , Hepatopatías/inmunología , Hepatopatías/patología , Macrófagos/patología , Microdominios de Membrana/genética , Ratones , Ratones Noqueados , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/patología , Pinocitosis/genética , Receptores Depuradores de Clase B/genética , Enfermedades del Bazo/genética , Enfermedades del Bazo/inmunología , Enfermedades del Bazo/patología
20.
Immunol Rev ; 274(1): 112-126, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27782330

RESUMEN

The activation of the complement system by canonical and non-canonical mechanisms results in the generation of multiple C3 and C5 cleavage fragments including anaphylatoxins C3a and C5a as well as opsonizing C3b/iC3b. It is now well appreciated that anaphylatoxins not only act as pro-inflammatory mediators but as immunoregulatory molecules that control the activation status of cells and tissue at several levels. Likewise, C3b/iC3b is more than the opsonizing fragment that facilitates engulfment and destruction of targets by phagocytes. In the circulation, it also facilitates the transport and delivery of bacteria and immune complexes to phagocytes, through a process known as immune adherence, with consequences for adaptive immunity. Here, we will discuss non-classical immunoregulatory properties of C3 and C5 cleavage fragments. We highlight the influence of anaphylatoxins on Th2 and Th17 cell development during allergic asthma with a particular emphasis on their role in the modulation of CD11b+ conventional dendritic cells and monocyte-derived dendritic cells. Furthermore, we discuss the control of anaphylatoxin-mediated activation of dendritic cells and allergic effector cells by adaptive immune mechanisms that involve allergen-specific IgG1 antibodies and plasma or regulatory T cell-derived IL-10 production. Finally, we take a fresh look at immune adherence with a particular focus on the development of antibacterial cytotoxic T-cell responses.


Asunto(s)
Complemento C3/metabolismo , Complemento C5/metabolismo , Células Dendríticas/inmunología , Hipersensibilidad/inmunología , Células TH1/inmunología , Células Th17/inmunología , Inmunidad Adaptativa , Animales , Diferenciación Celular , Activación de Complemento , Complemento C3/inmunología , Complemento C5/inmunología , Humanos , Inmunidad Innata , Inmunomodulación , Proteolisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...