Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunity ; 56(8): 1761-1777.e6, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37506694

RESUMEN

Conventional dendritic cells (cDCs) are professional antigen-presenting cells that control the adaptive immune response. Their subsets and developmental origins have been intensively investigated but are still not fully understood as their phenotypes, especially in the DC2 lineage and the recently described human DC3s, overlap with monocytes. Here, using LEGENDScreen to profile DC vs. monocyte lineages, we found sustained expression of FLT3 and CD45RB through the whole DC lineage, allowing DCs and their precursors to be distinguished from monocytes. Using fate mapping models, single-cell RNA sequencing and adoptive transfer, we identified a lineage of murine CD16/32+CD172a+ DC3, distinct from DC2, arising from Ly6C+ monocyte-DC progenitors (MDPs) through Lyz2+Ly6C+CD11c- pro-DC3s, whereas DC2s develop from common DC progenitors (CDPs) through CD7+Ly6C+CD11c+ pre-DC2s. Corresponding DC subsets, developmental stages, and lineages exist in humans. These findings reveal DC3 as a DC lineage phenotypically related to but developmentally different from monocytes and DC2s.


Asunto(s)
Monocitos , Células Madre , Ratones , Humanos , Animales , Fenotipo , Células Cultivadas , Células Dendríticas , Diferenciación Celular
2.
Methods Mol Biol ; 2618: 375-385, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36905527

RESUMEN

Single-cell technologies have become valuable tools to trace dendritic cell differentiation trajectories. Here, we illustrate the workflow used for processing of mouse bone marrow for single-cell RNA sequencing and trajectory analyses, as done in Dress et al. (Nat Immunol 20:852-864, 2019). This short methodology is presented as a starting point for researchers just beginning to dive into the complex field of dendritic cell ontogeny and cellular development trajectory analyses.


Asunto(s)
Células Dendríticas , Hematopoyesis , Animales , Ratones , Diferenciación Celular , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos
3.
Proc Natl Acad Sci U S A ; 120(13): e2219978120, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36940336

RESUMEN

We have previously shown that proteasome inhibitor bortezomib stabilizes p53 in stem and progenitor cells within gastrointestinal tissues. Here, we characterize the effect of bortezomib treatment on primary and secondary lymphoid tissues in mice. We find that bortezomib stabilizes p53 in significant fractions of hematopoietic stem and progenitor cells in the bone marrow, including common lymphoid and myeloid progenitors, granulocyte-monocyte progenitors, and dendritic cell progenitors. The stabilization of p53 is also observed in multipotent progenitors and hematopoietic stem cells, albeit at lower frequencies. In the thymus, bortezomib stabilizes p53 in CD4-CD8- T cells. Although there is less p53 stabilization in secondary lymphoid organs, cells in the germinal center of the spleen and Peyer's patch accumulate p53 in response to bortezomib. Bortezomib induces the upregulation of p53 target genes and p53 dependent/independent apoptosis in the bone marrow and thymus, suggesting that cells in these organs are robustly affected by proteasome inhibition. Comparative analysis of cell percentages in the bone marrow indicates expanded stem and multipotent progenitor pools in p53R172H mutant mice compared with p53 wild-type mice, suggesting a critical role for p53 in regulating the development and maturation of hematopoietic cells in the bone marrow. We propose that progenitors along the hematopoietic differentiation pathway express relatively high levels of p53 protein, which under steady-state conditions is constantly degraded by Mdm2 E3 ligase; however, these cells rapidly respond to stress to regulate stem cell renewal and consequently maintain the genomic integrity of hematopoietic stem/progenitor cell populations.


Asunto(s)
Inhibidores de Proteasoma , Proteína p53 Supresora de Tumor , Ratones , Animales , Bortezomib/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Progenitoras Mieloides/metabolismo , Ratones Endogámicos C57BL
4.
Eur J Immunol ; 2022 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-36563126

RESUMEN

This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. Recent studies have provided evidence for an increasing number of phenotypically distinct conventional DC (cDC) subsets that on one hand exhibit a certain functional plasticity, but on the other hand are characterized by their tissue- and context-dependent functional specialization. Here, we describe a selection of assays for the functional characterization of mouse and human cDC. The first two protocols illustrate analysis of cDC endocytosis and metabolism, followed by guidelines for transcriptomic and proteomic characterization of cDC populations. Then, a larger group of assays describes the characterization of cDC migration in vitro, ex vivo, and in vivo. The final guidelines measure cDC inflammasome and antigen (cross)-presentation activity. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.

5.
Eur J Immunol ; 51(12): 2708-3145, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34910301

RESUMEN

The third edition of Flow Cytometry Guidelines provides the key aspects to consider when performing flow cytometry experiments and includes comprehensive sections describing phenotypes and functional assays of all major human and murine immune cell subsets. Notably, the Guidelines contain helpful tables highlighting phenotypes and key differences between human and murine cells. Another useful feature of this edition is the flow cytometry analysis of clinical samples with examples of flow cytometry applications in the context of autoimmune diseases, cancers as well as acute and chronic infectious diseases. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid. All sections are written and peer-reviewed by leading flow cytometry experts and immunologists, making this edition an essential and state-of-the-art handbook for basic and clinical researchers.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Citometría de Flujo , Infecciones/inmunología , Neoplasias/inmunología , Animales , Enfermedad Crónica , Humanos , Ratones , Guías de Práctica Clínica como Asunto
6.
ACS Nano ; 15(10): 15754-15770, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34618423

RESUMEN

Multiple successful vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are urgently needed to address the ongoing coronavirus disease 2019 (Covid-19) pandemic. In the present work, we describe a subunit vaccine based on the SARS-CoV-2 spike protein coadministered with CpG adjuvant. To enhance the immunogenicity of our formulation, both antigen and adjuvant were encapsulated with our proprietary artificial cell membrane (ACM) polymersome technology. Structurally, ACM polymersomes are self-assembling nanoscale vesicles made up of an amphiphilic block copolymer comprising poly(butadiene)-b-poly(ethylene glycol) and a cationic lipid, 1,2-dioleoyl-3-trimethylammonium-propane. Functionally, ACM polymersomes serve as delivery vehicles that are efficiently taken up by dendritic cells (DC1 and DC2), which are key initiators of the adaptive immune response. Two doses of our formulation elicit robust neutralizing antibody titers in C57BL/6 mice that persist at least 40 days. Furthermore, we confirm the presence of functional memory CD4+ and CD8+ T cells that produce T helper type 1 cytokines. This study is an important step toward the development of an efficacious vaccine in humans.


Asunto(s)
Vacunas contra la COVID-19/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Linfocitos T CD8-positivos , COVID-19/prevención & control , Humanos , Ratones , Ratones Endogámicos C57BL , Nanopartículas , Subunidades de Proteína , SARS-CoV-2 , Vacunas de Subunidad
8.
Mol Immunol ; 122: 186-192, 2020 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-32388073

RESUMEN

Myeloid cells and their progenitors have historically been characterized based on their expression of a well-defined set of surface proteins, transcription factors and cytokines they depend on. These traditional analyses on "bulk" myeloid cell populations led to valuable early insights into the ontogeny of dendritic cells, granulocytes, monocytes and macrophages - a process called myelopoiesis. However, bulk approaches have limitations: they are unable to discern the individual stages and functions of progenitors and may be compromised by contaminating cells of non-myeloid lineages with similar or overlapping phenotypes. In recent years the emergence of high dimensional technologies to interrogate single cells at the molecular level, including single-cell mRNA sequencing and mass cytometry, has revolutionised our understanding of immune cell development and differentiation. Here, we highlight how the use of single-cell technologies has advanced our understanding of myelopoiesis and the emerging opportunities for it to continue to do so.

9.
Cell ; 178(6): 1509-1525.e19, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491389

RESUMEN

Most tissue-resident macrophage (RTM) populations are seeded by waves of embryonic hematopoiesis and are self-maintained independently of a bone marrow contribution during adulthood. A proportion of RTMs, however, is constantly replaced by blood monocytes, and their functions compared to embryonic RTMs remain unclear. The kinetics and extent of the contribution of circulating monocytes to RTM replacement during homeostasis, inflammation, and disease are highly debated. Here, we identified Ms4a3 as a specific gene expressed by granulocyte-monocyte progenitors (GMPs) and subsequently generated Ms4a3TdT reporter, Ms4a3Cre, and Ms4a3CreERT2 fate-mapping models. These models traced efficiently monocytes and granulocytes, but no lymphocytes or tissue dendritic cells. Using these models, we precisely quantified the contribution of monocytes to the RTM pool during homeostasis and inflammation. The unambiguous identification of monocyte-derived cells will permit future studies of their function under any condition.


Asunto(s)
Proteínas de Ciclo Celular/genética , Expresión Génica , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Granulocitos/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/genética , Monocitos/metabolismo , Animales , Células Progenitoras de Granulocitos y Macrófagos/citología , Granulocitos/citología , Hematopoyesis/fisiología , Homeostasis/fisiología , Inflamación/metabolismo , Macrófagos/citología , Ratones , Monocitos/citología
10.
Nat Immunol ; 20(7): 852-864, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31213723

RESUMEN

Dendritic cells (DC) are currently classified as conventional DCs (cDCs) and plasmacytoid DCs (pDCs). Through a combination of single-cell transcriptomic analysis, mass cytometry, in vivo fate mapping and in vitro clonal assays, here we show that, at the single-cell level, the priming of mouse hematopoietic progenitor cells toward the pDC lineage occurs at the common lymphoid progenitor stage, indicative of early divergence of the pDC and cDC lineages. We found the transcriptional signature of a pDC precursor stage, defined here, in the IL-7Rα+ common lymphoid progenitor population and identified Ly6D, IL-7Rα, CD81 and CD2 as key markers of pDC differentiation, which distinguish pDC precursors from cDC precursors. In conclusion, pDCs developed in the bone marrow from a Ly6DhiCD2hi lymphoid progenitor cell and differentiated independently of the myeloid cDC lineage.


Asunto(s)
Antígenos Ly/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Citometría de Flujo , Proteínas Ligadas a GPI/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Ratones , Transcriptoma
11.
Immunity ; 49(5): 819-828.e6, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30413362

RESUMEN

Inducing graft acceptance without chronic immunosuppression remains an elusive goal in organ transplantation. Using an experimental transplantation mouse model, we demonstrate that local macrophage activation through dectin-1 and toll-like receptor 4 (TLR4) drives trained immunity-associated cytokine production during allograft rejection. We conducted nanoimmunotherapeutic studies and found that a short-term mTOR-specific high-density lipoprotein (HDL) nanobiologic treatment (mTORi-HDL) averted macrophage aerobic glycolysis and the epigenetic modifications underlying inflammatory cytokine production. The resulting regulatory macrophages prevented alloreactive CD8+ T cell-mediated immunity and promoted tolerogenic CD4+ regulatory T (Treg) cell expansion. To enhance therapeutic efficacy, we complemented the mTORi-HDL treatment with a CD40-TRAF6-specific nanobiologic (TRAF6i-HDL) that inhibits co-stimulation. This synergistic nanoimmunotherapy resulted in indefinite allograft survival. Together, we show that HDL-based nanoimmunotherapy can be employed to control macrophage function in vivo. Our strategy, focused on preventing inflammatory innate immune responses, provides a framework for developing targeted therapies that promote immunological tolerance.


Asunto(s)
Supervivencia de Injerto/inmunología , Terapia de Inmunosupresión , Inflamación/inmunología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Trasplante de Órganos , Aloinjertos , Animales , Biomarcadores , Proteína HMGB1/genética , Tolerancia Inmunológica , Inmunidad Innata , Memoria Inmunológica , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Serina-Treonina Quinasas TOR/metabolismo , Vimentina/genética
12.
BMC Genomics ; 19(1): 194, 2018 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-29703139

RESUMEN

BACKGROUND: The murine discs large homolog 2 (DLG2; post synaptic density 93 (PSD-93); Chapsyn-110) is a member of the membrane-associated guanylate kinase (MAGUK) protein family involved in receptor assembly and associated with signaling enzymes on cell membranes. In neurons, DLG2 protein isoforms derived from alternatively spliced transcripts have been described to bind to NMDA (N-methyl-aspartate) receptors and K channels and to mediate clustering of these channels in the postsynaptic membrane. In myeloid cells of the immune system, such as dendritic cells (DCs), a lack of data exists on the expression or function of DLG2. In cDNA microarray transcriptome analyses, we found Dlg2 highly expressed in a subpopulation of plasmacytoid DCs (pDCs) stimulated to produce type I interferons (IFNs) such as IFNß. RESULTS: Using RACE- and RT-PCR as well as immunoprecipitation followed by Western blotting we characterised the differential expression of the Dlg2 splice variants in IFNß-producing pDCs. Besides Dlg2É£ this cell population expressed a novel short Dlg2η transcript we termed Dlg2η3. Our expression data were integrated into information from genome databases to obtain a novel and comprehensive overview of the mouse Dlg2 gene architecture. To elucidate the intracellular localisation pattern of protein isoforms, ectopical expression analysis of fluorescently tagged DLG2 splice variants was performed. Here we found an enrichment of the larger isoform DLG2α1 at the plasma membrane while the newly identified shorter (DLG2η) isoform as well as DLG2É£ were equally distributed throughout the cytoplasm. Additionally, DLG2η was also found in the nucleus. Analysis of Dlg2-knockout mice previously generated by deleting exon 9 surprisingly revealed that the protein for the novel DLG2η isoform was still expressed in the brain and in bone marrow-derived pDCs from mice carrying the homozygous deletion (Dlg2 ΔE9/ΔE9 ). CONCLUSION: We describe a novel splice variant of the mouse Dlg2 gene termed Dlg2η and define the differential expression pattern of DLG2 isoforms in IFNß-producing pDCs. The presence of DLG2η protein in the CNS of Dlg2 ΔE9/ΔE9 mice might influence the phenotype of these mice and has to be taken into account in the interpretation of results regarding the functional role of DLG2 in neuronal postsynaptic membranes.


Asunto(s)
Empalme Alternativo , Células Dendríticas/metabolismo , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Interferón beta/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Animales , Médula Ósea/metabolismo , Encéfalo/metabolismo , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Células Dendríticas/citología , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Análisis de Secuencia por Matrices de Oligonucleótidos , Isoformas de Proteínas/metabolismo , Regulación hacia Arriba
13.
Immunol Cell Biol ; 96(5): 463-476, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29473216

RESUMEN

Dendritic cells (DCs) are a diverse family of cells that play a crucial role in linking our innate and adaptive immune system to initiate adequate T-cell responses. They respond to pathogens by triggering the appropriate adaptive immune response that is required to clear invaders while balancing the need to limit tissue damage as a result of inflammation. Perturbation in DC regulation results in abnormal T-cell homeostasis, leading to development of autoimmune diseases and aberrant responses to pathogens. In recent years, it has become clear that the DC family is more complex than initially thought in terms of their identification, homeostasis and functions. In this review, we discuss recent findings that improve our understanding of development, and functional specializations of conventional and plasmacytoid DCs.


Asunto(s)
Células Dendríticas/inmunología , Inflamación/inmunología , Linfocitos T/inmunología , Inmunidad Adaptativa , Animales , Diferenciación Celular , Linaje de la Célula , Homeostasis , Humanos , Activación de Linfocitos
14.
J Immunol ; 196(11): 4447-51, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183572

RESUMEN

Type I IFNs are critical in initiating protective antiviral immune responses, and plasmacytoid dendritic cells (pDCs) represent a major source of these cytokines. We show that only few pDCs are capable of producing IFN-ß after virus infection or CpG stimulation. Using IFNß/YFP reporter mice, we identify these IFN-ß-producing cells in the spleen as a CCR9(+)CD9(-) pDC subset that is localized exclusively within the T/B cell zones. IFN-ß-producing pDCs exhibit a distinct transcriptome profile, with higher expression of genes encoding cytokines and chemokines, facilitating T cell recruitment and activation. These distinctive characteristics of IFN-ß-producing pDCs are independent of the type I IFNR-mediated feedback loop. Furthermore, IFN-ß-producing pDCs exhibit enhanced CCR7-dependent migratory properties in vitro. Additionally, they effectively recruit T cells in vivo in a peritoneal inflammation model. We define "professional type I IFN-producing cells" as a distinct subset of pDCs specialized in coordinating cellular immune responses.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interferón beta/genética , Bazo/citología , Bazo/inmunología , Transcriptoma , Animales , Interferón beta/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal
15.
Am J Respir Cell Mol Biol ; 52(1): 14-24, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24918472

RESUMEN

Treatment of primary or immortalized human airway epithelial cells (16HBE14o-, S9) or alveolar cancer cells (A549) with recombinant hemolysin A (rHla), a major virulence-associated factor of Staphylococcus aureus, induces alterations in cell shape and formation of paracellular gaps in the cell layer. Semiquantitative Western blotting using extracts of freshly isolated airway tissue (nasal epithelium) or 16HBE14o- model cells revealed that phosphorylation levels of focal adhesion kinase (Fak) and paxillin were altered upon treatment of tissue or cells with rHla. Immune fluorescence analyses showed that rHla treatment of 16HBE14o- cells results in losses of vinculin and paxillin from focal contacts and a net reduction in the number of focal contacts. The actin cytoskeleton was strongly remodeled. We concluded that treatment of cells with rHla activates Fak signaling, which accelerates focal contact turnover and prevents newly formed focal contacts (focal complexes) from maturation to focal adhesions. The inability of rHla-treated cells to form stable focal adhesions may be one factor that contributes to gap formation in the cell layer. In vivo, such changes may disturb the defensive barrier function of the airway epithelium and may facilitate lung infections by S. aureus.


Asunto(s)
Células Epiteliales/metabolismo , Matriz Extracelular/metabolismo , Neumonía Estafilocócica/metabolismo , Mucosa Respiratoria/metabolismo , Staphylococcus aureus/metabolismo , Toxinas Bacterianas/genética , Adhesión Celular/genética , Línea Celular , Células Epiteliales/patología , Matriz Extracelular/genética , Femenino , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Adhesiones Focales/patología , Proteínas Hemolisinas/genética , Humanos , Masculino , Paxillin/genética , Paxillin/metabolismo , Fosforilación/genética , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/patología , Mucosa Respiratoria/patología , Transducción de Señal/genética , Staphylococcus aureus/genética , Vinculina/genética , Vinculina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...