Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cell Rep ; 42(2): 112046, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36708514

RESUMEN

The diversity of mononuclear phagocyte (MNP) subpopulations across tissues is one of the key physiological characteristics of the immune system. Here, we focus on understanding the metabolic variability of MNPs through metabolic network analysis applied to three large-scale transcriptional datasets: we introduce (1) an ImmGen MNP open-source dataset of 337 samples across 26 tissues; (2) a myeloid subset of ImmGen Phase I dataset (202 MNP samples); and (3) a myeloid mouse single-cell RNA sequencing (scRNA-seq) dataset (51,364 cells) assembled based on Tabula Muris Senis. To analyze such large-scale datasets, we develop a network-based computational approach, genes and metabolites (GAM) clustering, for unbiased identification of the key metabolic subnetworks based on transcriptional profiles. We define 9 metabolic subnetworks that encapsulate the metabolic differences within MNP from 38 different tissues. Obtained modules reveal that cholesterol synthesis appears particularly active within the migratory dendritic cells, while glutathione synthesis is essential for cysteinyl leukotriene production by peritoneal and lung macrophages.


Asunto(s)
Fagocitos , Análisis de la Célula Individual , Animales , Ratones
2.
Nat Immunol ; 22(6): 699-710, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34040226

RESUMEN

It is increasingly recognized that immune development within mucosal tissues is under the control of environmental factors during early life. However, the cellular mechanisms that underlie such temporally and regionally restrictive governance of these processes are unclear. Here, we uncover an extrathymic pathway of immune development within the colon that is controlled by embryonic but not bone marrow-derived macrophages, which determines the ability of these organs to receive invariant natural killer T (iNKT) cells and allow them to establish local residency. Consequently, early-life perturbations of fetal-derived macrophages result in persistent decreases of mucosal iNKT cells and is associated with later-life susceptibility or resistance to iNKT cell-associated mucosal disorders. These studies uncover a host developmental program orchestrated by ontogenically distinct macrophages that is regulated by microbiota, and they reveal an important postnatal function of macrophages that emerge in fetal life.


Asunto(s)
Colitis/inmunología , Mucosa Intestinal/inmunología , Listeriosis/inmunología , Macrófagos/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Animales , Proliferación Celular/genética , Colitis/microbiología , Colitis/patología , Colon/citología , Colon/embriología , Colon/inmunología , Colon/patología , Citocinas/metabolismo , Toxina Diftérica/administración & dosificación , Toxina Diftérica/inmunología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Microbioma Gastrointestinal/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Vida Libre de Gérmenes , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/embriología , Mucosa Intestinal/patología , Listeriosis/microbiología , Listeriosis/patología , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , RNA-Seq , Transducción de Señal/genética , Transducción de Señal/inmunología
3.
Cell ; 181(3): 557-573.e18, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32259484

RESUMEN

Central nervous system (CNS) macrophages comprise microglia and border-associated macrophages (BAMs) residing in the meninges, the choroid plexus, and the perivascular spaces. Most CNS macrophages emerge during development, with the exception of choroid plexus and dural macrophages, which are replaced by monocytes in adulthood. Whether microglia and BAMs share a developmental program or arise from separate lineages remains unknown. Here, we identified two phenotypically, transcriptionally, and locally distinct brain macrophages throughout development, giving rise to either microglia or BAMs. Two macrophage populations were already present in the yolk sac suggesting an early segregation. Fate-mapping models revealed that BAMs mostly derived from early erythro-myeloid progenitors in the yolk sac. The development of microglia was dependent on TGF-ß, whereas the genesis of BAMs occurred independently of this cytokine. Collectively, our data show that developing parenchymal and non-parenchymal brain macrophages are separate entities in terms of ontogeny, gene signature, and requirement for TGF-ß.


Asunto(s)
Encéfalo/citología , Macrófagos/citología , Microglía/citología , Animales , Encéfalo/metabolismo , Linaje de la Célula , Ratones , Monocitos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
4.
Life Sci Alliance ; 3(1)2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31818882

RESUMEN

Acquired immune responses are initiated by activation of CD4+ helper T (Th) cells via recognition of antigens presented by conventional dendritic cells (cDCs). DCs instruct Th-cell polarization program into specific effector Th subset, which will dictate the type of immune responses. Hence, it is important to unravel how differentiation and/or activation of DC are linked with Th-cell-intrinsic mechanism that directs differentiation toward a specific effector Th subset. Here, we show that loss of Runx/Cbfß transcription factors complexes during DC development leads to loss of CD103+CD11b+ cDC2s and alters characteristics of CD103-CD11b+ cDCs in the intestine, which was accompanied with impaired differentiation of Rorγt+ Th17 cells and type 3 Rorγt+ regulatory T cells. We also show that a Runx-binding enhancer in the Rorc gene is essential for T cells to integrate cDC-derived signals to induce Rorγt expression. These findings reveal that Runx/Cbfß complexes play crucial and complementary roles in cDCs and Th cells to shape converging type 3 immune responses.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Células Dendríticas/metabolismo , Mucosa Intestinal/citología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Linfocitos T Reguladores/metabolismo , Células Th17/metabolismo , Inmunidad Adaptativa , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Células Dendríticas/inmunología , Mucosa Intestinal/inmunología , Ratones , Ratones Transgénicos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
5.
Methods Mol Biol ; 2034: 217-230, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31392688

RESUMEN

Microglia are the main population of macrophage residing in the central nervous system (CNS). Depletion experiments gave important insights into the physiology and function of microglia in healthy and diseased CNS. Ablation of microglia can be achieved by application of pharmacological or genetic tools. Here, we describe two approaches to ablate microglia: an efficient genetic model that utilizes DTRMG mouse line that has diphtheria toxin receptor (DTR) expression regulated by the promoter activity of the fractalkine receptor (CX3CR1) gene, and a pharmacological model that utilizes the blocking of macrophage colony-stimulating factor 1 receptor (CSF-1R) with a blocking antibody. Both the administration of systemic diphtheria toxin or anti-CSF-1R blocking antibody result in highly efficient and reversible depletion of microglia population in the CNS, which can be easily assessed by flow cytometry.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Receptor 1 de Quimiocinas CX3C/genética , Toxina Diftérica/farmacología , Expresión Génica , Factor de Crecimiento Similar a EGF de Unión a Heparina , Microglía , Regiones Promotoras Genéticas , Receptor de Factor Estimulante de Colonias de Macrófagos , Animales , Factor de Crecimiento Similar a EGF de Unión a Heparina/biosíntesis , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Ratones , Ratones Transgénicos , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo
6.
J Exp Med ; 216(10): 2265-2281, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31350310

RESUMEN

Microglia, the brain resident macrophages, critically shape forebrain neuronal circuits. However, their precise function in the cerebellum is unknown. Here we show that human and mouse cerebellar microglia express a unique molecular program distinct from forebrain microglia. Cerebellar microglial identity was driven by the CSF-1R ligand CSF-1, independently of the alternate CSF-1R ligand, IL-34. Accordingly, CSF-1 depletion from Nestin+ cells led to severe depletion and transcriptional alterations of cerebellar microglia, while microglia in the forebrain remained intact. Strikingly, CSF-1 deficiency and alteration of cerebellar microglia were associated with reduced Purkinje cells, altered neuronal function, and defects in motor learning and social novelty interactions. These findings reveal a novel CSF-1-CSF-1R signaling-mediated mechanism that contributes to motor function and social behavior.


Asunto(s)
Conducta Animal/fisiología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Microglía/metabolismo , Actividad Motora/fisiología , Células de Purkinje/metabolismo , Transducción de Señal/fisiología , Conducta Social , Animales , Humanos , Factor Estimulante de Colonias de Macrófagos/genética , Ratones , Ratones Transgénicos , Células de Purkinje/citología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo
7.
Science ; 363(6432)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30872492

RESUMEN

Macrophages are a heterogeneous cell population involved in tissue homeostasis, inflammation, and various pathologies. Although the major tissue-resident macrophage populations have been extensively studied, interstitial macrophages (IMs) residing within the tissue parenchyma remain poorly defined. Here we studied IMs from murine lung, fat, heart, and dermis. We identified two independent IM subpopulations that are conserved across tissues: Lyve1loMHCIIhiCX3CR1hi (Lyve1loMHCIIhi) and Lyve1hiMHCIIloCX3CR1lo (Lyve1hiMHCIIlo) monocyte-derived IMs, with distinct gene expression profiles, phenotypes, functions, and localizations. Using a new mouse model of inducible macrophage depletion (Slco2b1 flox/DTR), we found that the absence of Lyve1hiMHCIIlo IMs exacerbated experimental lung fibrosis. Thus, we demonstrate that two independent populations of IMs coexist across tissues and exhibit conserved niche-dependent functional programming.


Asunto(s)
Pulmón/inmunología , Pulmón/patología , Macrófagos/inmunología , Animales , Antígenos Ly , Receptor 1 de Quimiocinas CX3C/genética , Linaje de la Célula , Dermis/inmunología , Modelos Animales de Enfermedad , Fibrosis , Glicoproteínas/análisis , Antígenos de Histocompatibilidad Clase II/genética , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Miocardio/inmunología , Transportadores de Anión Orgánico/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcriptoma
8.
Immunity ; 49(2): 326-341.e7, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30054204

RESUMEN

The maintenance of appropriate arterial tone is critically important for normal physiological arterial function. However, the cellular and molecular mechanisms remain poorly defined. Here, we have shown that in the mouse aorta, resident macrophages prevented arterial stiffness and collagen deposition in the steady state. Using phenotyping, transcriptional profiling, and targeted deletion of Csf1r, we have demonstrated that these macrophages-which are a feature of blood vessels invested with smooth muscle cells (SMCs) in both mouse and human tissues-expressed the hyaluronan (HA) receptor LYVE-l. Furthermore, we have shown they possessed the unique ability to modulate collagen expression in SMCs by matrix metalloproteinase MMP-9-dependent proteolysis through engagement of LYVE-1 with the HA pericellular matrix of SMCs. Our study has unveiled a hitherto unknown homeostatic contribution of arterial LYVE-1+ macrophages through the control of collagen production by SMCs and has identified a function of LYVE-1 in leukocytes.


Asunto(s)
Colágeno/metabolismo , Glicoproteínas/metabolismo , Receptores de Hialuranos/metabolismo , Macrófagos/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Rigidez Vascular/fisiología , Animales , Aorta/fisiología , Femenino , Glicoproteínas/genética , Humanos , Ácido Hialurónico/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética
9.
Immunity ; 47(1): 183-198.e6, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723550

RESUMEN

Tissue macrophages arise during embryogenesis from yolk-sac (YS) progenitors that give rise to primitive YS macrophages. Until recently, it has been impossible to isolate or derive sufficient numbers of YS-derived macrophages for further study, but data now suggest that induced pluripotent stem cells (iPSCs) can be driven to undergo a process reminiscent of YS-hematopoiesis in vitro. We asked whether iPSC-derived primitive macrophages (iMacs) can terminally differentiate into specialized macrophages with the help of growth factors and organ-specific cues. Co-culturing human or murine iMacs with iPSC-derived neurons promoted differentiation into microglia-like cells in vitro. Furthermore, murine iMacs differentiated in vivo into microglia after injection into the brain and into functional alveolar macrophages after engraftment in the lung. Finally, iPSCs from a patient with familial Mediterranean fever differentiated into iMacs with pro-inflammatory characteristics, mimicking the disease phenotype. Altogether, iMacs constitute a source of tissue-resident macrophage precursors that can be used for biological, pathophysiological, and therapeutic studies.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Hematopoyesis , Macrófagos/fisiología , Neuronas/fisiología , Células Madre Pluripotentes/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Embrión de Mamíferos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis
10.
Nature ; 546(7660): 662-666, 2017 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-28614294

RESUMEN

During gestation the developing human fetus is exposed to a diverse range of potentially immune-stimulatory molecules including semi-allogeneic antigens from maternal cells, substances from ingested amniotic fluid, food antigens, and microbes. Yet the capacity of the fetal immune system, including antigen-presenting cells, to detect and respond to such stimuli remains unclear. In particular, dendritic cells, which are crucial for effective immunity and tolerance, remain poorly characterized in the developing fetus. Here we show that subsets of antigen-presenting cells can be identified in fetal tissues and are related to adult populations of antigen-presenting cells. Similar to adult dendritic cells, fetal dendritic cells migrate to lymph nodes and respond to toll-like receptor ligation; however, they differ markedly in their response to allogeneic antigens, strongly promoting regulatory T-cell induction and inhibiting T-cell tumour-necrosis factor-α production through arginase-2 activity. Our results reveal a previously unappreciated role of dendritic cells within the developing fetus and indicate that they mediate homeostatic immune-suppressive responses during gestation.


Asunto(s)
Arginasa/metabolismo , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Feto/inmunología , Tolerancia Inmunológica , Linfocitos T/inmunología , Adulto , Movimiento Celular , Proliferación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Feto/citología , Feto/enzimología , Humanos , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Linfocitos T/citología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Receptores Toll-Like/inmunología
11.
J Exp Med ; 213(11): 2293-2314, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27811056

RESUMEN

It is well established that Ly6Chi monocytes develop from common monocyte progenitors (cMoPs) and reside in the bone marrow (BM) until they are mobilized into the circulation. In our study, we found that BM Ly6Chi monocytes are not a homogenous population, as current data would suggest. Using computational analysis approaches to interpret multidimensional datasets, we demonstrate that BM Ly6Chi monocytes consist of two distinct subpopulations (CXCR4hi and CXCR4lo subpopulations) in both mice and humans. Transcriptome studies and in vivo assays revealed functional differences between the two subpopulations. Notably, the CXCR4hi subset proliferates and is immobilized in the BM for the replenishment of functionally mature CXCR4lo monocytes. We propose that the CXCR4hi subset represents a transitional premonocyte population, and that this sequential step of maturation from cMoPs serves to maintain a stable pool of BM monocytes. Additionally, reduced CXCR4 expression on monocytes, upon their exit into the circulation, does not reflect its diminished role in monocyte biology. Specifically, CXCR4 regulates monocyte peripheral cellular activities by governing their circadian oscillations and pulmonary margination, which contributes toward lung injury and sepsis mortality. Together, our study demonstrates the multifaceted role of CXCR4 in defining BM monocyte heterogeneity and in regulating their function in peripheral tissues.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Monocitos/citología , Receptores CXCR4/metabolismo , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Ritmo Circadiano/genética , Endotoxinas/toxicidad , Femenino , Perfilación de la Expresión Génica , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo
12.
Immunity ; 42(4): 665-78, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25902481

RESUMEN

Although classified as hematopoietic cells, tissue-resident macrophages (MFs) arise from embryonic precursors that seed the tissues prior to birth to generate a self-renewing population, which is maintained independently of adult hematopoiesis. Here we reveal the identity of these embryonic precursors using an in utero MF-depletion strategy and fate-mapping of yolk sac (YS) and fetal liver (FL) hematopoiesis. We show that YS MFs are the main precursors of microglia, while most other MFs derive from fetal monocytes (MOs). Both YS MFs and fetal MOs arise from erythro-myeloid progenitors (EMPs) generated in the YS. In the YS, EMPs gave rise to MFs without monocytic intermediates, while EMP seeding the FL upon the establishment of blood circulation acquired c-Myb expression and gave rise to fetal MOs that then seeded embryonic tissues and differentiated into MFs. Thus, adult tissue-resident MFs established from hematopoietic stem cell-independent embryonic precursors arise from two distinct developmental programs.


Asunto(s)
Envejecimiento/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Células Progenitoras Mieloides/inmunología , Proteínas Proto-Oncogénicas c-myb/inmunología , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula/inmunología , Rastreo Celular , Embrión de Mamíferos , Femenino , Feto , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Riñón/citología , Riñón/inmunología , Hígado/citología , Hígado/inmunología , Pulmón/citología , Pulmón/inmunología , Macrófagos/citología , Ratones , Microglía/citología , Microglía/inmunología , Monocitos/citología , Células Progenitoras Mieloides/citología , Embarazo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-myb/metabolismo , Piel/citología , Piel/inmunología , Saco Vitelino/citología , Saco Vitelino/inmunología
13.
Nat Immunol ; 16(5): 505-16, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25751747

RESUMEN

A cytosolic role for the histone methyltransferase Ezh2 in regulating lymphocyte activation has been suggested, but the molecular mechanisms underpinning this extranuclear function have remained unclear. Here we found that Ezh2 regulated the integrin signaling and adhesion dynamics of neutrophils and dendritic cells (DCs). Ezh2 deficiency impaired the integrin-dependent transendothelial migration of innate leukocytes and restricted disease progression in an animal model of multiple sclerosis. Direct methylation of talin, a key regulatory molecule in cell migration, by Ezh2 disrupted the binding of talin to F-actin and thereby promoted the turnover of adhesion structures. This regulatory effect was abolished by targeted disruption of the interactions of Ezh2 with the cytoskeletal-reorganization effector Vav1. Our studies reveal an unforeseen extranuclear function for Ezh2 in regulating adhesion dynamics, with implications for leukocyte migration, immune responses and potentially pathogenic processes.


Asunto(s)
Núcleo Celular/metabolismo , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/inmunología , Neutrófilos/inmunología , Complejo Represivo Polycomb 2/metabolismo , Talina/metabolismo , Actinas/metabolismo , Animales , Adhesión Celular/genética , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Activación de Linfocitos/genética , Metilación , Ratones , Ratones Noqueados , Complejo Represivo Polycomb 2/genética , Unión Proteica/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Talina/genética , Migración Transendotelial y Transepitelial/genética
14.
Immunity ; 38(5): 970-83, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23706669

RESUMEN

Mouse and human dendritic cells (DCs) are composed of functionally specialized subsets, but precise interspecies correlation is currently incomplete. Here, we showed that murine lung and gut lamina propria CD11b+ DC populations were comprised of two subsets: FLT3- and IRF4-dependent CD24(+)CD64(-) DCs and contaminating CSF-1R-dependent CD24(-)CD64(+) macrophages. Functionally, loss of CD24(+)CD11b(+) DCs abrogated CD4+ T cell-mediated interleukin-17 (IL-17) production in steady state and after Aspergillus fumigatus challenge. Human CD1c+ DCs, the equivalent of murine CD24(+)CD11b(+) DCs, also expressed IRF4, secreted IL-23, and promoted T helper 17 cell responses. Our data revealed heterogeneity in the mouse CD11b+ DC compartment and identifed mucosal tissues IRF4-expressing DCs specialized in instructing IL-17 responses in both mouse and human. The demonstration of mouse and human DC subsets specialized in driving IL-17 responses highlights the conservation of key immune functions across species and will facilitate the translation of mouse in vivo findings to advance DC-based clinical therapies.


Asunto(s)
Aspergillus fumigatus/inmunología , Células Dendríticas/metabolismo , Factores Reguladores del Interferón/metabolismo , Interleucina-17/metabolismo , Células Th17/metabolismo , Animales , Antígeno CD11b/metabolismo , Antígeno CD24/metabolismo , Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Humanos , Interleucina-17/biosíntesis , Interleucina-23/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Macrófagos/metabolismo , Ratones , Receptores de IgG/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Tirosina Quinasa 3 Similar a fms/metabolismo
15.
Immunity ; 38(4): 792-804, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23601688

RESUMEN

Despite accumulating evidence suggesting local self-maintenance of tissue macrophages in the steady state, the dogma remains that tissue macrophages derive from monocytes. Using parabiosis and fate-mapping approaches, we confirmed that monocytes do not show significant contribution to tissue macrophages in the steady state. Similarly, we found that after depletion of lung macrophages, the majority of repopulation occurred by stochastic cellular proliferation in situ in a macrophage colony-stimulating factor (M-Csf)- and granulocyte macrophage (GM)-CSF-dependent manner but independently of interleukin-4. We also found that after bone marrow transplantation, host macrophages retained the capacity to expand when the development of donor macrophages was compromised. Expansion of host macrophages was functional and prevented the development of alveolar proteinosis in mice transplanted with GM-Csf-receptor-deficient progenitors. Collectively, these results indicate that tissue-resident macrophages and circulating monocytes should be classified as mononuclear phagocyte lineages that are independently maintained in the steady state.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Pulmón/inmunología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/inmunología , Adulto , Animales , Trasplante de Médula Ósea , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Homeostasis , Humanos , Interleucina-4/metabolismo , Macrófagos/trasplante , Ratones , Ratones Noqueados , Ratones Mutantes , Parabiosis , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética
16.
Immunity ; 37(1): 60-73, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22795876

RESUMEN

Dendritic cell (DC)-mediated cross-presentation of exogenous antigens acquired in the periphery is critical for the initiation of CD8(+) T cell responses. Several DC subsets are described in human tissues but migratory cross-presenting DCs have not been isolated, despite their potential importance in immunity to pathogens, vaccines, and tumors and tolerance to self. Here, we identified a CD141(hi) DC present in human interstitial dermis, liver, and lung that was distinct from the majority of CD1c(+) and CD14(+) tissue DCs and superior at cross-presenting soluble antigens. Cutaneous CD141(hi) DCs were closely related to blood CD141(+) DCs, and migratory counterparts were found among skin-draining lymph node DCs. Comparative transcriptomic analysis with mouse showed tissue DC subsets to be conserved between species and permitted close alignment of human and mouse DC subsets. These studies inform the rational design of targeted immunotherapies and facilitate translation of mouse functional DC biology to the human setting.


Asunto(s)
Antígenos CD/metabolismo , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Cadenas alfa de Integrinas/metabolismo , Animales , Antígenos/inmunología , Movimiento Celular/inmunología , Quimiocina CXCL10/biosíntesis , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Piel/inmunología , Transcriptoma , Factor de Necrosis Tumoral alfa/biosíntesis
17.
J Exp Med ; 209(6): 1167-81, 2012 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-22565823

RESUMEN

Langerhans cells (LCs) are the dendritic cells (DCs) of the epidermis, forming one of the first hematopoietic lines of defense against skin pathogens. In contrast to other DCs, LCs arise from hematopoietic precursors that seed the skin before birth. However, the origin of these embryonic precursors remains unclear. Using in vivo lineage tracing, we identify a first wave of yolk sac (YS)-derived primitive myeloid progenitors that seed the skin before the onset of fetal liver hematopoiesis. YS progenitors migrate to the embryo proper, including the prospective skin, where they give rise to LC precursors, and the brain rudiment, where they give rise to microglial cells. However, in contrast to microglia, which remain of YS origin throughout life, YS-derived LC precursors are largely replaced by fetal liver monocytes during late embryogenesis. Consequently, adult LCs derive predominantly from fetal liver monocyte-derived cells with a minor contribution of YS-derived cells. Altogether, we establish that adult LCs have a dual origin, bridging early embryonic and late fetal myeloid development.


Asunto(s)
Células de Langerhans/citología , Hígado/citología , Hígado/embriología , Monocitos , Saco Vitelino/citología , Saco Vitelino/embriología , Factores de Edad , Animales , Linaje de la Célula , Femenino , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Células Progenitoras Mieloides , Embarazo , Piel/citología , Piel/embriología , Células Madre
18.
Immunol Res ; 53(1-3): 115-26, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22418726

RESUMEN

Malaria remains one of the main infectious diseases in intertropical regions. The malaria parasite has a complex life cycle in its mammalian host, switching between variable forms as it traverses through different tissues and anatomic locations, either intra- or intercellularly. During its journey, the parasite encounters and interacts with the host immune system, which functions to prevent infections and limit ensuing pathologies. One important component of the host immune system is the dendritic cells (DC) network. DC form a heterogeneous group of pathogen-sensing and antigen-presenting cells that play a crucial role in the initiation of adaptive immunity. Here, we review the known and unknown interactions between the malaria parasites and the DC system, starting from the inoculation of the parasite in the skin up to its exit from the liver, also known as the pre-erythrocytic stage of the infection, and discuss how deciphering these interactions may contribute to our understanding of the Plasmodium parasite biology as well as to the induction of immune protection via vaccination.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/parasitología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Plasmodium/inmunología , Inmunidad Adaptativa , Animales , Eritrocitos/inmunología , Humanos , Inmunoterapia , Malaria/prevención & control , Vacunas contra la Malaria/administración & dosificación , Terapia Molecular Dirigida
19.
Science ; 330(6005): 841-5, 2010 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-20966214

RESUMEN

Microglia are the resident macrophages of the central nervous system and are associated with the pathogenesis of many neurodegenerative and brain inflammatory diseases; however, the origin of adult microglia remains controversial. We show that postnatal hematopoietic progenitors do not significantly contribute to microglia homeostasis in the adult brain. In contrast to many macrophage populations, we show that microglia develop in mice that lack colony stimulating factor-1 (CSF-1) but are absent in CSF-1 receptor-deficient mice. In vivo lineage tracing studies established that adult microglia derive from primitive myeloid progenitors that arise before embryonic day 8. These results identify microglia as an ontogenically distinct population in the mononuclear phagocyte system and have implications for the use of embryonically derived microglial progenitors for the treatment of various brain disorders.


Asunto(s)
Encéfalo/citología , Macrófagos/citología , Microglía/citología , Células Progenitoras Mieloides/citología , Animales , Encéfalo/embriología , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/fisiología , Femenino , Técnicas de Sustitución del Gen , Hematopoyesis , Células Madre Hematopoyéticas/citología , Homeostasis , Factor Estimulante de Colonias de Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Saco Vitelino/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA