Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Cell ; 185(23): 4317-4332.e15, 2022 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-36302380

RESUMEN

Therapeutic cancer vaccines are designed to increase tumor-specific T cell immunity. However, suppressive mechanisms within the tumor microenvironment (TME) may limit T cell function. Here, we assessed how the route of vaccination alters intratumoral myeloid cells. Using a self-assembling nanoparticle vaccine that links tumor antigen peptides to a Toll-like receptor 7/8 agonist (SNP-7/8a), we treated tumor-bearing mice subcutaneously (SNP-SC) or intravenously (SNP-IV). Both routes generated antigen-specific CD8+ T cells that infiltrated tumors. However, only SNP-IV mediated tumor regression, dependent on systemic type I interferon at the time of boost. Single-cell RNA-sequencing revealed that intratumoral monocytes expressing an immunoregulatory gene signature (Chil3, Anxa2, Wfdc17) were reduced after SNP-IV boost. In humans, the Chil3+ monocyte gene signature is enriched in CD16- monocytes and associated with worse outcomes. Our results show that the generation of tumor-specific CD8+ T cells combined with remodeling of the TME is a promising approach for tumor immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Microambiente Tumoral , Humanos , Ratones , Animales , Linfocitos T CD8-positivos , Línea Celular Tumoral , Inmunoterapia/métodos , Antígenos de Neoplasias , Vacunación/métodos , Adyuvantes Inmunológicos
2.
Cell ; 185(7): 1189-1207.e25, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35325594

RESUMEN

Macrophage infiltration is a hallmark of solid cancers, and overall macrophage infiltration correlates with lower patient survival and resistance to therapy. Tumor-associated macrophages, however, are phenotypically and functionally heterogeneous. Specific subsets of tumor-associated macrophage might be endowed with distinct roles on cancer progression and antitumor immunity. Here, we identify a discrete population of FOLR2+ tissue-resident macrophages in healthy mammary gland and breast cancer primary tumors. FOLR2+ macrophages localize in perivascular areas in the tumor stroma, where they interact with CD8+ T cells. FOLR2+ macrophages efficiently prime effector CD8+ T cells ex vivo. The density of FOLR2+ macrophages in tumors positively correlates with better patient survival. This study highlights specific roles for tumor-associated macrophage subsets and paves the way for subset-targeted therapeutic interventions in macrophages-based cancer therapies.


Asunto(s)
Neoplasias de la Mama , Macrófagos , Mama/inmunología , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos , Femenino , Receptor 2 de Folato , Humanos , Linfocitos Infiltrantes de Tumor , Pronóstico
3.
Cell ; 184(13): 3394-3409.e20, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34077752

RESUMEN

The human fetal immune system begins to develop early during gestation; however, factors responsible for fetal immune-priming remain elusive. We explored potential exposure to microbial agents in utero and their contribution toward activation of memory T cells in fetal tissues. We profiled microbes across fetal organs using 16S rRNA gene sequencing and detected low but consistent microbial signal in fetal gut, skin, placenta, and lungs in the 2nd trimester of gestation. We identified several live bacterial strains including Staphylococcus and Lactobacillus in fetal tissues, which induced in vitro activation of memory T cells in fetal mesenteric lymph node, supporting the role of microbial exposure in fetal immune-priming. Finally, using SEM and RNA-ISH, we visualized discrete localization of bacteria-like structures and eubacterial-RNA within 14th weeks fetal gut lumen. These findings indicate selective presence of live microbes in fetal organs during the 2nd trimester of gestation and have broader implications toward the establishment of immune competency and priming before birth.


Asunto(s)
Bacterias/metabolismo , Desarrollo Embrionario , Feto/citología , Feto/microbiología , Leucocitos/citología , Adulto , Bacterias/genética , Bacterias/ultraestructura , Proliferación Celular , Células Dendríticas/metabolismo , Femenino , Feto/ultraestructura , Tracto Gastrointestinal/embriología , Tracto Gastrointestinal/ultraestructura , Humanos , Memoria Inmunológica , Activación de Linfocitos/inmunología , Viabilidad Microbiana , Embarazo , Segundo Trimestre del Embarazo , ARN Bacteriano/genética , ARN Ribosómico 16S/genética , Reproducibilidad de los Resultados , Linfocitos T/citología
4.
Cell ; 183(2): 377-394.e21, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32976798

RESUMEN

We employed scRNA sequencing to extensively characterize the cellular landscape of human liver from development to disease. Analysis of ∼212,000 cells representing human fetal, hepatocellular carcinoma (HCC), and mouse liver revealed remarkable fetal-like reprogramming of the tumor microenvironment. Specifically, the HCC ecosystem displayed features reminiscent of fetal development, including re-emergence of fetal-associated endothelial cells (PLVAP/VEGFR2) and fetal-like (FOLR2) tumor-associated macrophages. In a cross-species comparative analysis, we discovered remarkable similarity between mouse embryonic, fetal-liver, and tumor macrophages. Spatial transcriptomics further revealed a shared onco-fetal ecosystem between fetal liver and HCC. Furthermore, gene regulatory analysis, spatial transcriptomics, and in vitro functional assays implicated VEGF and NOTCH signaling in maintaining onco-fetal ecosystem. Taken together, we report a shared immunosuppressive onco-fetal ecosystem in fetal liver and HCC. Our results unravel a previously unexplored onco-fetal reprogramming of the tumor ecosystem, provide novel targets for therapeutic interventions in HCC, and open avenues for identifying similar paradigms in other cancers and disease.


Asunto(s)
Carcinoma Hepatocelular/patología , Células Endoteliales/metabolismo , Microambiente Tumoral/genética , Adulto , Animales , Carcinoma Hepatocelular/genética , Línea Celular , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Receptor 2 de Folato/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Hígado/patología , Neoplasias Hepáticas/genética , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal/genética , Transcriptoma/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
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
6.
Cell ; 179(7): 1609-1622.e16, 2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31835035

RESUMEN

Microglia, the brain-resident immune cells, are critically involved in many physiological and pathological brain processes, including neurodegeneration. Here we characterize microglia morphology and transcriptional programs across ten species spanning more than 450 million years of evolution. We find that microglia express a conserved core gene program of orthologous genes from rodents to humans, including ligands and receptors associated with interactions between glia and neurons. In most species, microglia show a single dominant transcriptional state, whereas human microglia display significant heterogeneity. In addition, we observed notable differences in several gene modules of rodents compared with primate microglia, including complement, phagocytic, and susceptibility genes to neurodegeneration, such as Alzheimer's and Parkinson's disease. Our study provides an essential resource of conserved and divergent microglia pathways across evolution, with important implications for future development of microglia-based therapies in humans.


Asunto(s)
Evolución Molecular , Redes Reguladoras de Genes , Microglía/metabolismo , Enfermedades Neurodegenerativas/genética , Análisis de la Célula Individual , Transcriptoma , Animales , Pollos , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Primates , Reptiles , Roedores , Ovinos , Porcinos , Pez Cebra
7.
Nat Immunol ; 22(1): 41-52, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33139915

RESUMEN

Personalized cancer vaccines are a promising approach for inducing T cell immunity to tumor neoantigens. Using a self-assembling nanoparticle vaccine that links neoantigen peptides to a Toll-like receptor 7/8 agonist (SNP-7/8a), we show how the route and dose alter the magnitude and quality of neoantigen-specific CD8+ T cells. Intravenous vaccination (SNP-IV) induced a higher proportion of TCF1+PD-1+CD8+ T cells as compared to subcutaneous immunization (SNP-SC). Single-cell RNA sequencing showed that SNP-IV induced stem-like genes (Tcf7, Slamf6, Xcl1) whereas SNP-SC enriched for effector genes (Gzmb, Klrg1, Cx3cr1). Stem-like cells generated by SNP-IV proliferated and differentiated into effector cells upon checkpoint blockade, leading to superior antitumor response as compared to SNP-SC in a therapeutic model. The duration of antigen presentation by dendritic cells controlled the magnitude and quality of CD8+ T cells. These data demonstrate how to optimize antitumor immunity by modulating vaccine parameters for specific generation of effector or stem-like CD8+ T cells.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Factor Nuclear 1-alfa del Hepatocito/análisis , Nanopartículas , Animales , Presentación de Antígeno , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Femenino , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Vacunación
8.
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
9.
Nat Immunol ; 25(3): 383-384, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38351323

Asunto(s)
Animales , Ratones
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 ; 55(3): 393-404, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35263567

RESUMEN

The immune system is a complex, dynamic, and plastic ecosystem composed of multiple cell types that constantly sense and interact with their local microenvironment to protect from infection and maintain homeostasis. For over a century, great efforts and ingenuity have been applied to the characterization of immune cells and their microenvironments, but traditional marker-based and bulk technologies left key questions unanswered. In the past decade, the advent of single-cell genomic approaches has revolutionized our knowledge of the cellular and molecular makeup of the immune system. In this perspective, we outline the past, present, and future applications of single-cell genomics in immunology and discuss how the integration of multiomics at the single-cell level will pave the way for future advances in immunology research and clinical translation.


Asunto(s)
Ecosistema , Análisis de la Célula Individual , Genómica , Sistema Inmunológico
12.
Immunity ; 55(11): 2187-2205.e5, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36351376

RESUMEN

Bats are reservoir hosts of many zoonotic viruses with pandemic potential. We utilized single-cell transcriptome sequencing (scRNA-seq) to analyze the immune response in bat lungs upon in vivo infection with a double-stranded RNA virus, Pteropine orthoreovirus PRV3M. Bat neutrophils were distinguished by high basal IDO1 expression. NK cells and T cells were the most abundant immune cells in lung tissue. Three distinct CD8+ effector T cell populations could be delineated by differential expression of KLRB1, GFRA2, and DPP4. Select NK and T clusters increased expression of genes involved in T cell activation and effector function early after viral infection. Alveolar macrophages and classical monocytes drove antiviral interferon signaling. Infection expanded a CSF1R+ population expressing collagen-like genes, which became the predominant myeloid cell type post-infection. This work uncovers features relevant to viral disease tolerance in bats, lays a foundation for future experimental work, and serves as a resource for comparative immunology studies.


Asunto(s)
Quirópteros , Virosis , Animales , Quirópteros/genética , Néctar de las Plantas , Transcriptoma , Análisis de la Célula Individual , Perfilación de la Expresión Génica
13.
Immunity ; 55(8): 1448-1465.e6, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35931085

RESUMEN

Brain macrophage populations include parenchymal microglia, border-associated macrophages, and recruited monocyte-derived cells; together, they control brain development and homeostasis but are also implicated in aging pathogenesis and neurodegeneration. The phenotypes, localization, and functions of each population in different contexts have yet to be resolved. We generated a murine brain myeloid scRNA-seq integration to systematically delineate brain macrophage populations. We show that the previously identified disease-associated microglia (DAM) population detected in murine Alzheimer's disease models actually comprises two ontogenetically and functionally distinct cell lineages: embryonically derived triggering receptor expressed on myeloid cells 2 (TREM2)-dependent DAM expressing a neuroprotective signature and monocyte-derived TREM2-expressing disease inflammatory macrophages (DIMs) accumulating in the brain during aging. These two distinct populations appear to also be conserved in the human brain. Herein, we generate an ontogeny-resolved model of brain myeloid cell heterogeneity in development, homeostasis, and disease and identify cellular targets for the treatment of neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Envejecimiento , Enfermedad de Alzheimer/genética , Animales , Encéfalo/patología , Humanos , Macrófagos/patología , Glicoproteínas de Membrana , Ratones , Microglía/patología , Receptores Inmunológicos
14.
Immunity ; 54(8): 1825-1840.e7, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34270940

RESUMEN

Hepatocellular carcinoma (HCC) often develops following chronic hepatitis B virus (HBV) infection and responds poorly to immune checkpoint blockade. Here, we examined the antigen specificities of HCC-infiltrating T cells and their relevance to tumor control. Using highly multiplexed peptide-MHC tetramer staining of unexpanded cells from blood, liver, and tumor tissues from 46 HCC patients, we detected 91 different antigen-specific CD8+ T cell populations targeting HBV, neoantigen, tumor-associated, and disease-unrelated antigens. Parallel high-dimensional analysis delineated five distinct antigen-specific tissue-resident memory T (Trm) cell populations. Intratumoral and intrahepatic HBV-specific T cells were enriched for two Trm cell subsets that were PD-1loTOXlo, despite being clonally expanded. High frequencies of intratumoral terminally exhausted T cells were uncommon. Patients with tumor-infiltrating HBV-specific CD8+ Trm cells exhibited longer-term relapse-free survival. Thus, non-terminally exhausted HBV-specific CD8+ Trm cells show hallmarks of active involvement and effective antitumor response, implying that these cells could be harnessed for therapeutic purposes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma Hepatocelular/inmunología , Memoria Inmunológica/inmunología , Neoplasias Hepáticas/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Antígenos de Neoplasias/inmunología , Carcinoma Hepatocelular/patología , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/inmunología , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Neoplasias Hepáticas/patología , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/prevención & control , Receptor de Muerte Celular Programada 1/metabolismo , Células Tumorales Cultivadas
15.
Immunity ; 54(8): 1883-1900.e5, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34331874

RESUMEN

Mononuclear phagocytes (MNPs) encompass dendritic cells, monocytes, and macrophages (MoMac), which exhibit antimicrobial, homeostatic, and immunoregulatory functions. We integrated 178,651 MNPs from 13 tissues across 41 datasets to generate a MNP single-cell RNA compendium (MNP-VERSE), a publicly available tool to map MNPs and define conserved gene signatures of MNP populations. Next, we generated a MoMac-focused compendium that revealed an array of specialized cell subsets widely distributed across multiple tissues. Specific pathological forms were expanded in cancer and inflammation. All neoplastic tissues contained conserved tumor-associated macrophage populations. In particular, we focused on IL4I1+CD274(PD-L1)+IDO1+ macrophages, which accumulated in the tumor periphery in a T cell-dependent manner via interferon-γ (IFN-γ) and CD40/CD40L-induced maturation from IFN-primed monocytes. IL4I1_Macs exhibited immunosuppressive characteristics through tryptophan degradation and promoted the entry of regulatory T cell into tumors. This integrated analysis provides a robust online-available platform for uniform annotation and dissection of specific macrophage functions in healthy and pathological states.


Asunto(s)
Células Dendríticas/inmunología , Expresión Génica/inmunología , Monocitos/inmunología , Transcriptoma/genética , Macrófagos Asociados a Tumores/inmunología , Artritis Reumatoide/inmunología , COVID-19/inmunología , Expresión Génica/genética , Perfilación de la Expresión Génica , Humanos , Interferón gamma/inmunología , L-Aminoácido Oxidasa/metabolismo , Cirrosis Hepática/inmunología , Macrófagos/inmunología , Neoplasias/inmunología , ARN Citoplasmático Pequeño/genética , Análisis de la Célula Individual , Linfocitos T Reguladores/inmunología , Transcriptoma/inmunología
17.
Immunity ; 53(2): 303-318.e5, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32579887

RESUMEN

Granulocyte-monocyte progenitors (GMPs) have been previously defined for their potential to generate various myeloid progenies such as neutrophils and monocytes. Although studies have proposed lineage heterogeneity within GMPs, it is unclear if committed progenitors already exist among these progenitors and how they may behave differently during inflammation. By combining single-cell transcriptomic and proteomic analyses, we identified the early committed progenitor within the GMPs responsible for the strict production of neutrophils, which we designate as proNeu1. Our dissection of the GMP hierarchy led us to further identify a previously unknown intermediate proNeu2 population. Similar populations could be detected in human samples. proNeu1s, but not proNeu2s, selectively expanded during the early phase of sepsis at the expense of monocytes. Collectively, our findings help shape the neutrophil maturation trajectory roadmap and challenge the current definition of GMPs.


Asunto(s)
Células Precursoras de Granulocitos/citología , Monocitos/citología , Mielopoyesis/fisiología , Neutrófilos/citología , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de la Célula Individual
18.
Nature ; 623(7986): 397-405, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37914940

RESUMEN

Microglia are specialized brain-resident macrophages that arise from primitive macrophages colonizing the embryonic brain1. Microglia contribute to multiple aspects of brain development, but their precise roles in the early human brain remain poorly understood owing to limited access to relevant tissues2-6. The generation of brain organoids from human induced pluripotent stem cells recapitulates some key features of human embryonic brain development7-10. However, current approaches do not incorporate microglia or address their role in organoid maturation11-21. Here we generated microglia-sufficient brain organoids by coculturing brain organoids with primitive-like macrophages generated from the same human induced pluripotent stem cells (iMac)22. In organoid cocultures, iMac differentiated into cells with microglia-like phenotypes and functions (iMicro) and modulated neuronal progenitor cell (NPC) differentiation, limiting NPC proliferation and promoting axonogenesis. Mechanistically, iMicro contained high levels of PLIN2+ lipid droplets that exported cholesterol and its esters, which were taken up by NPCs in the organoids. We also detected PLIN2+ lipid droplet-loaded microglia in mouse and human embryonic brains. Overall, our approach substantially advances current human brain organoid approaches by incorporating microglial cells, as illustrated by the discovery of a key pathway of lipid-mediated crosstalk between microglia and NPCs that leads to improved neurogenesis.


Asunto(s)
Encéfalo , Colesterol , Células Madre Pluripotentes Inducidas , Microglía , Células-Madre Neurales , Neurogénesis , Organoides , Animales , Humanos , Ratones , Encéfalo/citología , Encéfalo/metabolismo , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Microglía/citología , Microglía/metabolismo , Organoides/citología , Organoides/metabolismo , Colesterol/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Axones , Proliferación Celular , Ésteres/metabolismo , Gotas Lipídicas/metabolismo
19.
Immunity ; 51(3): 573-589.e8, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31474513

RESUMEN

Human mononuclear phagocytes comprise phenotypically and functionally overlapping subsets of dendritic cells (DCs) and monocytes, but the extent of their heterogeneity and distinct markers for subset identification remains elusive. By integrating high-dimensional single-cell protein and RNA expression data, we identified distinct markers to delineate monocytes from conventional DC2 (cDC2s). Using CD88 and CD89 for monocytes and HLA-DQ and FcεRIα for cDC2s allowed for their specific identification in blood and tissues. We also showed that cDC2s could be subdivided into phenotypically and functionally distinct subsets based on CD5, CD163, and CD14 expression, including a distinct subset of circulating inflammatory CD5-CD163+CD14+ cells related to previously defined DC3s. These inflammatory DC3s were expanded in systemic lupus erythematosus patients and correlated with disease activity. These findings further unravel the heterogeneity of DC subpopulations in health and disease and may pave the way for the identification of specific DC subset-targeting therapies.


Asunto(s)
Biomarcadores/sangre , Células Dendríticas/inmunología , Inflamación/sangre , Inflamación/inmunología , Leucocitos Mononucleares/inmunología , Fagocitos/inmunología , Antígenos CD/sangre , Antígenos CD/inmunología , Células Cultivadas , Citometría de Flujo/métodos , Humanos , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Monocitos/inmunología , Fenotipo , Análisis de la Célula Individual
20.
Nature ; 603(7899): 152-158, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35173329

RESUMEN

Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system underpinned by partially understood genetic risk factors and environmental triggers and their undefined interactions1,2. Here we investigated the peripheral immune signatures of 61 monozygotic twin pairs discordant for MS to dissect the influence of genetic predisposition and environmental factors. Using complementary multimodal high-throughput and high-dimensional single-cell technologies in conjunction with data-driven computational tools, we identified an inflammatory shift in a monocyte cluster of twins with MS, coupled with the emergence of a population of IL-2 hyper-responsive transitional naive helper T cells as MS-related immune alterations. By integrating data on the immune profiles of healthy monozygotic and dizygotic twin pairs, we estimated the variance in CD25 expression by helper T cells displaying a naive phenotype to be largely driven by genetic and shared early environmental influences. Nonetheless, the expanding helper T cells of twins with MS, which were also elevated in non-twin patients with MS, emerged independent of the individual genetic makeup. These cells expressed central nervous system-homing receptors, exhibited a dysregulated CD25-IL-2 axis, and their proliferative capacity positively correlated with MS severity. Together, our matched-pair analysis of the extended twin approach allowed us to discern genetically and environmentally determined features of an MS-associated immune signature.


Asunto(s)
Esclerosis Múltiple , Predisposición Genética a la Enfermedad/genética , Humanos , Interleucina-2/genética , Ligando OX40 , Gemelos Dicigóticos/genética , Gemelos Monocigóticos/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda