Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
Nat Commun ; 15(1): 5910, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39003273

RESUMEN

Lymphoid specification in human hematopoietic progenitors is not fully understood. To better associate lymphoid identity with protein-level cell features, we conduct a highly multiplexed single-cell proteomic screen on human bone marrow progenitors. This screen identifies terminal deoxynucleotidyl transferase (TdT), a specialized DNA polymerase intrinsic to VDJ recombination, broadly expressed within CD34+ progenitors prior to B/T cell emergence. While these TdT+ cells coincide with granulocyte-monocyte progenitor (GMP) immunophenotype, their accessible chromatin regions show enrichment for lymphoid-associated transcription factor (TF) motifs. TdT expression on GMPs is inversely related to the SLAM family member CD84. Prospective isolation of CD84lo GMPs demonstrates robust lymphoid potentials ex vivo, while still retaining significant myeloid differentiation capacity, akin to LMPPs. This multi-omic study identifies human bone marrow lymphoid-primed progenitors, further defining the lympho-myeloid axis in human hematopoiesis.


Asunto(s)
ADN Nucleotidilexotransferasa , Células Progenitoras Linfoides , Humanos , ADN Nucleotidilexotransferasa/metabolismo , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/citología , Diferenciación Celular , Análisis de la Célula Individual , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Hematopoyesis , Proteómica/métodos , Antígenos CD/metabolismo , Antígenos CD/genética , Antígenos CD34/metabolismo
2.
Nat Immunol ; 25(7): 1183-1192, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38872000

RESUMEN

Natural killer (NK) cells function by eliminating virus-infected or tumor cells. Here we identified an NK-lineage-biased progenitor population, referred to as early NK progenitors (ENKPs), which developed into NK cells independently of common precursors for innate lymphoid cells (ILCPs). ENKP-derived NK cells (ENKP_NK cells) and ILCP-derived NK cells (ILCP_NK cells) were transcriptionally different. We devised combinations of surface markers that identified highly enriched ENKP_NK and ILCP_NK cell populations in wild-type mice. Furthermore, Ly49H+ NK cells that responded to mouse cytomegalovirus infection primarily developed from ENKPs, whereas ILCP_NK cells were better IFNγ producers after infection with Salmonella and herpes simplex virus. Human CD56dim and CD56bright NK cells were transcriptionally similar to ENKP_NK cells and ILCP_NK cells, respectively. Our findings establish the existence of two pathways of NK cell development that generate functionally distinct NK cell subsets in mice and further suggest these pathways may be conserved in humans.


Asunto(s)
Diferenciación Celular , Células Asesinas Naturales , Células Asesinas Naturales/inmunología , Animales , Ratones , Humanos , Diferenciación Celular/inmunología , Ratones Endogámicos C57BL , Inmunidad Innata , Antígeno CD56/metabolismo , Muromegalovirus/inmunología , Linaje de la Célula/inmunología , Interferón gamma/metabolismo , Interferón gamma/inmunología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Ratones Noqueados , Células Cultivadas
3.
Trends Immunol ; 45(7): 495-510, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38908962

RESUMEN

Over the past decade our research has implemented a multimodal approach to human lymphopoiesis, combining clonal-scale mapping of lymphoid developmental architecture with the monitoring of dynamic changes in the pattern of lymphocyte generation across ontogeny. We propose that lymphopoiesis stems from founder populations of CD127/interleukin (IL)7R- or CD127/IL7R+ early lymphoid progenitors (ELPs) polarized respectively toward the T-natural killer (NK)/innate lymphoid cell (ILC) or B lineages, arising from newly characterized CD117lo multi-lymphoid progenitors (MLPs). Recent data on the lifelong lymphocyte dynamics of healthy donors suggest that, after birth, lymphopoiesis may become increasingly oriented toward the production of B lymphocytes. Stemming from this, we posit that there are three major developmental transitions, the first occurring during the neonatal period, the next at puberty, and the last during aging.


Asunto(s)
Envejecimiento , Linfopoyesis , Humanos , Envejecimiento/inmunología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/inmunología , Linfocitos B/inmunología , Animales , Diferenciación Celular , Células Asesinas Naturales/inmunología
4.
Cell Rep ; 43(5): 114200, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38717905

RESUMEN

Innate lymphoid cells (ILCs), strategically positioned throughout the body, undergo population declines over time. A solution to counteract this problem is timely mobilization of multipotential progenitors from the bone marrow. It remains unknown what triggers the mobilization of bone marrow ILC progenitors (ILCPs). We report that ILCPs are regulated by the circadian clock to emigrate and generate mature ILCs in the periphery. We found that circadian-clock-defective ILCPs fail to normally emigrate and generate ILCs. We identified circadian-clock-controlled endocrine and cytokine cues that, respectively, regulate the retention and emigration of ILCPs at distinct times of each day. Activation of the stress-hormone-sensing glucocorticoid receptor upregulates CXCR4 on ILCPs for their retention in the bone marrow, while the interleukin-18 (IL-18) and RORα signals upregulate S1PR1 on ILCPs for their mobilization to the periphery. Our findings establish important roles of circadian signals for the homeostatic efflux of bone marrow ILCPs.


Asunto(s)
Relojes Circadianos , Animales , Ratones , Citocinas/metabolismo , Ratones Endogámicos C57BL , Médula Ósea/metabolismo , Transducción de Señal , Receptores CXCR4/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Células Progenitoras Linfoides/metabolismo , Células Progenitoras Linfoides/citología , Inmunidad Innata , Movimiento Celular , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Receptores de Glucocorticoides/metabolismo , Linfocitos/metabolismo , Linfocitos/inmunología
5.
Methods Mol Biol ; 2580: 211-232, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36374460

RESUMEN

T cells and innate lymphoid cells (ILCs) share expression of many key transcription factors during development and at mature stage, resulting in striking functional similarities between these lineages. Taking into account ILC contribution is thus necessary to appreciate T cell functions during immune responses. Furthermore, understanding ILC development and functions helps to understand T cells. Here we provide methods and protocols to isolate pure populations of multipotent precursors to T cells and innate lymphoid cells (ILCs) from adult mouse bone marrow, using flow cytometric sorting. These include precursors to all lymphocytes (viz., LMPPs and ALPs) and multipotent precursors to ILCs that have been recently refined (viz., specified EILPs, committed EILPs, and ILCPs).


Asunto(s)
Linfocitos , Linfocitos T , Ratones , Animales , Inmunidad Innata , Médula Ósea , Células Progenitoras Linfoides/metabolismo , Diferenciación Celular
6.
Cell Rep ; 41(5): 111569, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36323259

RESUMEN

Innate lymphoid cells (ILCs) play important roles in regulating tissue homeostasis and innate immune responses. Generation of ILCs after engraftment of pluripotent stem cell (PSC)-derived hematopoietic progenitors (iHPCs) has not yet been reported. Here, we document that ILCs exist in Rag2-/-Il2rg-/- recipients engrafted with PSC-derived iHPCs guided by Runx1 and Hoxa9 expression. Upon transplantation, iHPCs immediately give rise to ILC-related progenitors containing common helper ILC progenitors in the bone marrow, followed by a more restricted population named ILC progenitors, which are able to further differentiate into mature ILCs in the primary and secondary immunodeficient recipients. The PSC-derived ILCs exhibit multiple tissue distributions and normal immunological functions. Single-cell transcriptomics illustrates the developmental trajectory of PSC-derived ILCs in vivo, which is consistent with that of natural ILCs. Our study provides insights into the generation of ILCs in animals transplanted with PSC-derived iHPCs as a cell source.


Asunto(s)
Inmunidad Innata , Células Madre Pluripotentes , Animales , Linfocitos/metabolismo , Diferenciación Celular , Células Progenitoras Linfoides/metabolismo
7.
Front Immunol ; 13: 880668, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35603175

RESUMEN

The development of B cells relies on an intricate network of transcription factors critical for developmental progression and lineage commitment. In the B cell developmental trajectory, a temporal switch from predominant Foxo3 to Foxo1 expression occurs at the CLP stage. Utilizing VAV-iCre mediated conditional deletion, we found that the loss of FOXO3 impaired B cell development from LMPP down to B cell precursors, while the loss of FOXO1 impaired B cell commitment and resulted in a complete developmental block at the CD25 negative proB cell stage. Strikingly, the combined loss of FOXO1 and FOXO3 resulted in the failure to restrict the myeloid potential of CLPs and the complete loss of the B cell lineage. This is underpinned by the failure to enforce the early B-lineage gene regulatory circuitry upon a predominantly pre-established open chromatin landscape. Altogether, this demonstrates that FOXO3 and FOXO1 cooperatively govern early lineage restriction and initiation of B-lineage commitment in CLPs.


Asunto(s)
Hematopoyesis , Células Progenitoras Linfoides , Linfocitos B/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Hematopoyesis/genética , Células Progenitoras Linfoides/metabolismo , Células Precursoras de Linfocitos B/metabolismo
8.
J Exp Med ; 219(2)2022 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-34958351

RESUMEN

During dendritic cell (DC) development, Myc expression in progenitors is replaced by Mycl in mature DCs, but when and how this transition occurs is unknown. We evaluated DC development using reporters for MYC, MYCL, and cell cycle proteins Geminin and CDT1 in wild-type and various mutant mice. For classical type 1 dendritic cells (cDC1s) and plasmacytoid DCs (pDCs), the transition occurred upon their initial specification from common dendritic cell progenitors (CDPs) or common lymphoid progenitors (CLPs), respectively. This transition required high levels of IRF8 and interaction with PU.1, suggesting the use of EICEs within Mycl enhancers. In pDCs, maximal MYCL induction also required the +41kb Irf8 enhancer that controls pDC IRF8 expression. IRF8 also contributed to repression of MYC. While MYC is expressed only in rapidly dividing DC progenitors, MYCL is most highly expressed in DCs that have exited the cell cycle. Thus, IRF8 levels coordinate the Myc-Mycl transition during DC development.


Asunto(s)
Diferenciación Celular/genética , Células Dendríticas/citología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica , Genes myc , Factores Reguladores del Interferón/genética , Animales , Proteínas de Ciclo Celular/genética , Elementos de Facilitación Genéticos , Genes Reporteros , Inmunofenotipificación , Factores Reguladores del Interferón/metabolismo , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/metabolismo , Ratones , Ratones Noqueados , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo
9.
J Exp Med ; 219(3)2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-34928315

RESUMEN

In the mouse, the first hematopoietic cells are generated in the yolk sac from the primitive, erythro-myeloid progenitor (EMP) and lymphoid programs that are specified before the emergence of hematopoietic stem cells. While many of the yolk sac-derived populations are transient, specific immune cell progeny seed developing tissues, where they function into adult life. To access the human equivalent of these lineages, we modeled yolk sac hematopoietic development using pluripotent stem cell differentiation. Here, we show that the combination of Activin A, BMP4, and FGF2 induces a population of KDR+CD235a/b+ mesoderm that gives rise to the spectrum of erythroid, myeloid, and T lymphoid lineages characteristic of the mouse yolk sac hematopoietic programs, including the Vδ2+ subset of γ/δ T cells that develops early in the human embryo. Through clonal analyses, we identified a multipotent hematopoietic progenitor with erythroid, myeloid, and T lymphoid potential, suggesting that the yolk sac EMP and lymphoid lineages may develop from a common progenitor.


Asunto(s)
Hematopoyesis , Modelos Biológicos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Saco Vitelino/citología , Animales , Biomarcadores , Diferenciación Celular/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunofenotipificación , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Linfopoyesis/genética , Ratones , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
10.
Science ; 371(6536)2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33766856

RESUMEN

The pathways that lead to the development of tissue-resident lymphocytes, including liver type 1 innate lymphoid cells (ILC1s), remain unclear. We show here that the adult mouse liver contains Lin-Sca-1+Mac-1+ hematopoietic stem cells derived from the fetal liver. This population includes Lin-CD122+CD49a+ progenitors that can generate liver ILC1s but not conventional natural killer cells. Interferon-γ (IFN-γ) production by the liver ILC1s themselves promotes the development of these cells in situ, through effects on their IFN-γR+ liver progenitors. Thus, an IFN-γ-dependent loop drives liver ILC1 development in situ, highlighting the contribution of extramedullary hematopoiesis to regional immune composition within the liver.


Asunto(s)
Interferón gamma/metabolismo , Hígado/citología , Hígado/inmunología , Linfocitos/citología , Linfocitos/inmunología , Animales , Hematopoyesis Extramedular , Inmunidad Innata , Interferón gamma/genética , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Linfopoyesis , Ratones , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/metabolismo , Receptor de Interferón gamma
11.
Cell Rep ; 34(12): 108894, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33761361

RESUMEN

The process of hematopoiesis is subject to substantial ontogenic remodeling that is accompanied by alterations in cellular fate during both development and disease. We combine state-of-the-art mass spectrometry with extensive functional assays to gain insight into ontogeny-specific proteomic mechanisms regulating hematopoiesis. Through deep coverage of the cellular proteome of fetal and adult lympho-myeloid multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs), and granulocyte-monocyte progenitors (GMPs), we establish that features traditionally attributed to adult hematopoiesis are conserved across lymphoid and myeloid lineages, whereas generic fetal features are suppressed in GMPs. We reveal molecular and functional evidence for a diminished granulocyte differentiation capacity in fetal LMPPs and GMPs relative to their adult counterparts. Our data indicate an ontogeny-specific requirement of myosin activity for myelopoiesis in LMPPs. Finally, we uncover an ontogenic shift in the monocytic differentiation capacity of GMPs, partially driven by a differential expression of Irf8 during fetal and adult life.


Asunto(s)
Linaje de la Célula , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteómica , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular , Feto/citología , Granulocitos/citología , Células HEK293 , Humanos , Inmunofenotipificación , Factores Reguladores del Interferón/metabolismo , Cinética , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Ratones Endogámicos C57BL , Monocitos/citología , Monocitos/metabolismo , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Proteoma/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
12.
Blood ; 137(8): 1024-1036, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33025012

RESUMEN

During embryonic development, multiple waves of hematopoietic progenitors with distinct lineage potential are differentially regulated in time and space. Two different waves of thymic progenitors colonize the fetal thymus where they contribute to thymic organogenesis and homeostasis. The origin, the lineage differentiation potential of the first wave, and their relative contribution in shaping the thymus architecture, remained, however, unclear. Here, we show that the first wave of thymic progenitors comprises a unique population of bipotent T and innatel lymphoid cells (T/ILC), generating a lymphoid tissue inducer cells (LTi's), in addition to invariant Vγ5+ T cells. Transcriptional analysis revealed that innate lymphoid gene signatures and, more precisely, the LTi-associated transcripts were expressed in the first, but not in the second, wave of thymic progenitors. Depletion of early thymic progenitors in a temporally controlled manner showed that the progeny of the first wave is indispensable for the differentiation of autoimmune regulator-expressing medullary thymic epithelial cells (mTECs). We further show that these progenitors are of strict hematopoietic stem cell origin, despite the overlap between lymphopoiesis initiation and the transient expression of lymphoid-associated transcripts in yolk sac (YS) erythromyeloid-restricted precursors. Our work highlights the relevance of the developmental timing on the emergence of different lymphoid subsets, required for the establishment of a functionally diverse immune system.


Asunto(s)
Células Progenitoras Linfoides/citología , Linfocitos T/citología , Timo/citología , Timo/embriología , Animales , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Células Progenitoras Linfoides/metabolismo , Linfopoyesis , Ratones Endogámicos C57BL , Linfocitos T/metabolismo , Timo/metabolismo , Transcriptoma
13.
Front Immunol ; 12: 825813, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095929

RESUMEN

Protection against pathogen re-infection is mediated, in large part, by two humoral cellular compartments, namely, long-lived plasma cells and memory B cells. Recent data have reinforced the importance of memory B cells, particularly in response to re-infection of different viral subtypes or in response with viral escape mutants. In regard to memory B cell generation, considerable advancements have been made in recent years in elucidating its basic mechanism, which seems to well explain why the memory B cells pool can deal with variant viruses. Despite such progress, efforts to develop vaccines that induce broadly protective memory B cells to fight against rapidly mutating pathogens such as influenza virus and HIV have not yet been successful. Here, we discuss recent advances regarding the key signals and factors regulating germinal center-derived memory B cell development and activation and highlight the challenges for successful vaccine development.


Asunto(s)
Memoria Inmunológica , Células B de Memoria/inmunología , Células B de Memoria/metabolismo , Anticuerpos Neutralizantes/inmunología , Formación de Anticuerpos/genética , Formación de Anticuerpos/inmunología , Comunicación Celular/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Selección Clonal Mediada por Antígenos , Femenino , Centro Germinal/citología , Centro Germinal/inmunología , Centro Germinal/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/metabolismo , Masculino , Células B de Memoria/citología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
14.
PLoS One ; 15(12): e0244161, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33332417

RESUMEN

C/EBPα is required for formation of granulocyte-monocyte progenitors (GMP) and also participates in B lymphopoiesis. The common lymphoid progenitor (CLP) and preproB populations but not proB cells express Cebpa, and pan-hematopoietic deletion of the +37 kb Cebpa enhancer using Mx1-Cre leads not only to reduced GMP but also to 2-fold reduced marrow preproB and >15-fold reduced proB and preB cells. We now show that IL7Rα-Cre-mediated deletion of the +37 kb Cebpa enhancer, which occurs in 89% of Ly6D+ and 65% of upstream Ly6D- CLP, leads to a 2-fold reduction of both preproB and proB cells, and a 3-fold reduction in preB cells, with no impact on GMP numbers. These data support a direct role for C/EBPα during B lineage development, with reduced enhancer deletion in Ly6D- CLP mediated by IL7Rα-Cre diminishing the effect on B lymphopoiesis compared to that seen with Mx1-Cre. Amongst mRNAs encoding key transcriptional regulators that initiate B lymphoid specification (PU.1, E2A, IKAROS, EBF1, FOXO1, and BACH2), only Ebf1 levels are altered in CLP upon Mx1-Cre-mediated Cebpa enhancer deletion, with Ebf1 reduced ~40-fold in Flt3+Sca-1intc-kitintIL7Rα+ CLP. In addition, Cebpa and Ebf1 RNAs were 4- and 14-fold higher in hCD4+ versus hCD4- CLP from Cebpa-hCD4 transgenic mice. Histone modification ChIP-Seq data for CLP indicate the presence of active, intronic Ebf1 enhancers located 270 and 280 kb upstream of the transcription start sites. We identified a cis element in this region that strongly binds C/EBPα using the electrophoretic mobility shift assay. Mutation of this C/EBPα-binding site in an Ebf1 enhancer-TK-luciferase reporter leads to a 4-fold reduction in C/EBPα-mediated trans-activation. These findings support a model of B lymphopoiesis in which induction of Ebf1 by C/EBPα in a subset of CLP contributes to initiation of B lymphopoiesis.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Células Progenitoras Linfoides/metabolismo , Transactivadores/genética , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Linaje de la Célula , Células Cultivadas , Elementos de Facilitación Genéticos , Femenino , Células HEK293 , Humanos , Células Progenitoras Linfoides/citología , Linfopoyesis , Masculino , Ratones , Ratones Endogámicos C57BL , Transactivadores/metabolismo
15.
Sci Immunol ; 5(54)2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33277375

RESUMEN

Early hematopoietic progenitors undergo sophisticated developmental processes to become committed innate lymphoid cell (ILC) progenitors and ultimately mature ILC subsets in the periphery. Basic leucine zipper ATF-like transcription factor (Batf) plays important roles in lymphocyte biology. We report here that Batf regulates the production of bone marrow ILC progenitors and maintenance of peripheral ILCs. The expression of Batf is induced during ILC development at the α-lymphoid progenitor stage in response to the cytokine IL-7. As a potential mechanism, up-regulated Batf binds and activates transcription of the Nfil3 gene to promote ILC hematopoiesis. Batf is necessary to maintain normal numbers of early and late ILC progenitors in the bone marrow and mature ILC1, ILC2, ILC3, and NK cells in most peripheral tissues. Batf deficiency causes ILC lymphopenia, leading to defective ILC responses to inflammatory cytokines and defective immunity to enteric bacterial infections. Thus, Batf plays critical roles in bone marrow hematopoiesis, peripheral homeostasis, and effector functions of ILCs.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Hematopoyesis/fisiología , Homeostasis , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Inmunofenotipificación , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Ratones , Ratones Noqueados , Especificidad de Órganos , Transducción de Señal
16.
Nat Immunol ; 21(12): 1574-1584, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33077975

RESUMEN

A classical view of blood cell development is that multipotent hematopoietic stem and progenitor cells (HSPCs) become lineage-restricted at defined stages. Lin-c-Kit+Sca-1+Flt3+ cells, termed lymphoid-primed multipotent progenitors (LMPPs), have lost megakaryocyte and erythroid potential but are heterogeneous in their fate. Here, through single-cell RNA sequencing, we identify the expression of Dach1 and associated genes in this fraction as being coexpressed with myeloid/stem genes but inversely correlated with lymphoid genes. Through generation of Dach1-GFP reporter mice, we identify a transcriptionally and functionally unique Dach1-GFP- subpopulation within LMPPs with lymphoid potential with low to negligible classic myeloid potential. We term these 'lymphoid-primed progenitors' (LPPs). These findings define an early definitive branch point of lymphoid development in hematopoiesis and a means for prospective isolation of LPPs.


Asunto(s)
Biomarcadores , Proteínas del Ojo/metabolismo , Genómica , Células Progenitoras Linfoides/metabolismo , Análisis de la Célula Individual , Animales , Células Cultivadas , Biología Computacional/métodos , Proteínas del Ojo/genética , Perfilación de la Expresión Génica , Genómica/métodos , Hematopoyesis/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteómica , Análisis de la Célula Individual/métodos
17.
Nat Immunol ; 21(12): 1552-1562, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046887

RESUMEN

T cell memory relies on the generation of antigen-specific progenitors with stem-like properties. However, the identity of these progenitors has remained unclear, precluding a full understanding of the differentiation trajectories that underpin the heterogeneity of antigen-experienced T cells. We used a systematic approach guided by single-cell RNA-sequencing data to map the organizational structure of the human CD8+ memory T cell pool under physiological conditions. We identified two previously unrecognized subsets of clonally, epigenetically, functionally, phenotypically and transcriptionally distinct stem-like CD8+ memory T cells. Progenitors lacking the inhibitory receptors programmed death-1 (PD-1) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) were committed to a functional lineage, whereas progenitors expressing PD-1 and TIGIT were committed to a dysfunctional, exhausted-like lineage. Collectively, these data reveal the existence of parallel differentiation programs in the human CD8+ memory T cell pool, with potentially broad implications for the development of immunotherapies and vaccines.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Memoria Inmunológica , Células Progenitoras Linfoides/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Biomarcadores , Diferenciación Celular/inmunología , Biología Computacional/métodos , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunofenotipificación , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Ratones , Homeostasis del Telómero
18.
Immunity ; 52(6): 1088-1104.e6, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32304633

RESUMEN

During postnatal life, thymopoiesis depends on the continuous colonization of the thymus by bone-marrow-derived hematopoietic progenitors that migrate through the bloodstream. The current understanding of the nature of thymic immigrants is largely based on data from pre-clinical models. Here, we employed single-cell RNA sequencing (scRNA-seq) to examine the immature postnatal thymocyte population in humans. Integration of bone marrow and peripheral blood precursor datasets identified two putative thymus seeding progenitors that varied in expression of CD7; CD10; and the homing receptors CCR7, CCR9, and ITGB7. Whereas both precursors supported T cell development, only one contributed to intrathymic dendritic cell (DC) differentiation, predominantly of plasmacytoid dendritic cells. Trajectory inference delineated the transcriptional dynamics underlying early human T lineage development, enabling prediction of transcription factor (TF) modules that drive stage-specific steps of human T cell development. This comprehensive dataset defines the expression signature of immature human thymocytes and provides a resource for the further study of human thymopoiesis.


Asunto(s)
Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , ARN Citoplasmático Pequeño/genética , Timocitos/citología , Timocitos/metabolismo , Biomarcadores , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunofenotipificación , Análisis de la Célula Individual , Timocitos/inmunología , Transcriptoma
19.
J Immunol ; 204(9): 2447-2454, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32198141

RESUMEN

The caudal hematopoietic tissue in zebrafish, the equivalent to the fetal liver in mammals, is an intermediate hematopoietic niche for the maintenance and differentiation of hematopoietic stem and progenitor cells before homing to the thymus and kidney marrow. As one of the ultimate hematopoietic organs, the thymus sustains T lymphopoiesis, which is essential for adaptive immune system. However, the mechanism of prethymic T lymphoid progenitors migrating to the thymus remains elusive. In this study, we identify an Rho GTPase Rac2 as a modulator of T lymphoid progenitor homing to the thymus in zebrafish. rac2-Deficient embryos show the inability of T lymphoid progenitors homing to the thymus because of defective cell-autonomous motility. Mechanistically, we demonstrate that Rac2 regulates homing of T lymphoid progenitor through Pak1-mediated AKT pathway. Taken together, our work reveals an important function of Rac2 in directing T lymphoid progenitor migration to the thymus during zebrafish embryogenesis.


Asunto(s)
Movimiento Celular/inmunología , Desarrollo Embrionario/inmunología , Células Progenitoras Linfoides/metabolismo , Timo/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Animales , Animales Modificados Genéticamente/inmunología , Animales Modificados Genéticamente/metabolismo , Médula Ósea/inmunología , Médula Ósea/metabolismo , Diferenciación Celular/inmunología , Células Progenitoras Linfoides/inmunología , Linfopoyesis/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/inmunología , Timo/inmunología , Pez Cebra/inmunología , Proteínas de Pez Cebra/inmunología , Quinasas p21 Activadas/inmunología , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/inmunología , Proteínas de Unión al GTP rho/inmunología , Proteínas de Unión al GTP rho/metabolismo
20.
Methods Mol Biol ; 2121: 7-22, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32147782

RESUMEN

Understanding the origins and developmental trajectory of innate lymphoid cell (ILC) progenitors has been of substantial interest to the fields of ILC biology and immunology. While mature ILC are rare lymphocytes, ILC progenitors represent an even smaller fraction of cells, providing additional challenges in studying them. Moreover, though the approaches to studying these cells are conceptually straightforward, the technical nuances that underlie them can substantially affect the quality of the data. Herein, we provide a detailed protocol for assessing the frequency of ILC progenitors in the bone marrow, their phenotype, and their potential to develop into mature ILC. These methods make up the foundation of in vivo investigations into ILC development, and we hope these thorough protocols and associated notes facilitate additional, high-quality inquiries into this fascinating field.


Asunto(s)
Traslado Adoptivo/métodos , Células de la Médula Ósea , Células Asesinas Naturales/citología , Hígado/citología , Linfocitos/citología , Células Progenitoras Linfoides/citología , Linfopoyesis/inmunología , Animales , Médula Ósea , Células de la Médula Ósea/citología , Linaje de la Célula , Femenino , Citometría de Flujo , Células Asesinas Naturales/inmunología , Hígado/inmunología , Linfocitos/inmunología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/metabolismo , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...