Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(7): 114365, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-38909363

RESUMO

Here, we examine how prenatal inflammation shapes tissue function and immunity in the lung by reprogramming tissue-resident immune cells from early development. Maternal, but not fetal, type I interferon-mediated inflammation provokes expansion and hyperactivation of group 2 innate lymphoid cells (ILC2s) seeding the developing lung. Hyperactivated ILC2s produce increased IL-5 and IL-13 and are associated with acute Th2 bias, decreased Tregs, and persistent lung eosinophilia into adulthood. ILC2 hyperactivation is recapitulated by adoptive transfer of fetal liver precursors following prenatal inflammation, indicative of developmental programming at the fetal progenitor level. Reprogrammed ILC2 hyperactivation and subsequent lung immune remodeling, including persistent eosinophilia, is concomitant with worsened histopathology and increased airway dysfunction equivalent to papain exposure, indicating increased asthma susceptibility in offspring. Our data elucidate a mechanism by which early-life inflammation results in increased asthma susceptibility in the presence of hyperactivated ILC2s that drive persistent changes to lung immunity during perinatal development.


Assuntos
Imunidade Inata , Inflamação , Pulmão , Linfócitos , Animais , Pulmão/imunologia , Pulmão/patologia , Feminino , Linfócitos/imunologia , Inflamação/patologia , Inflamação/imunologia , Gravidez , Camundongos , Asma/imunologia , Asma/patologia , Camundongos Endogâmicos C57BL , Células Th2/imunologia
2.
Cell ; 187(12): 3090-3107.e21, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38749423

RESUMO

Platelet dysregulation is drastically increased with advanced age and contributes to making cardiovascular disorders the leading cause of death of elderly humans. Here, we reveal a direct differentiation pathway from hematopoietic stem cells into platelets that is progressively propagated upon aging. Remarkably, the aging-enriched platelet path is decoupled from all other hematopoietic lineages, including erythropoiesis, and operates as an additional layer in parallel with canonical platelet production. This results in two molecularly and functionally distinct populations of megakaryocyte progenitors. The age-induced megakaryocyte progenitors have a profoundly enhanced capacity to engraft, expand, restore, and reconstitute platelets in situ and upon transplantation and produce an additional platelet population in old mice. The two pools of co-existing platelets cause age-related thrombocytosis and dramatically increased thrombosis in vivo. Strikingly, aging-enriched platelets are functionally hyper-reactive compared with the canonical platelet populations. These findings reveal stem cell-based aging as a mechanism for platelet dysregulation and age-induced thrombosis.


Assuntos
Envelhecimento , Plaquetas , Diferenciação Celular , Células-Tronco Hematopoéticas , Trombose , Animais , Células-Tronco Hematopoéticas/metabolismo , Plaquetas/metabolismo , Trombose/patologia , Trombose/metabolismo , Camundongos , Humanos , Megacariócitos/metabolismo , Camundongos Endogâmicos C57BL , Células Progenitoras de Megacariócitos/metabolismo , Masculino
3.
EMBO J ; 42(14): e112693, 2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37259639

RESUMO

Infection directly influences adult hematopoietic stem cell (HSC) function and differentiation, but the fetal hematopoietic response to infection during pregnancy is not well-studied. Here, we investigated the fetal hematopoietic response to maternal infection with Toxoplasma gondii (T. gondii), an intracellular parasite that elicits Type II IFNγ-mediated maternal immunity. While it is known that maternal infection without direct pathogen transmission can affect fetal immune development, the effects of maternal IFNγ on developing HSCs and the signals that mediate these interactions have not been investigated. Our investigation reveals that the fetal HSCs respond to T. gondii infection with virulence-dependent changes in proliferation, self-renewal potential, and lineage output. Furthermore, maternal IFNγ crosses the fetal-maternal interface, where it is perceived by fetal HSCs. By comparing the effects of maternal IFNγ injection with maternal T. gondii infection, we reveal that the effects of IFNγ treatment mimic some aspects of the fetal HSC response to infection. Moreover, our findings illuminate that the fetal HSC response to prenatal infection is distinct from the adult HSC response to IFNγ-induced inflammation. Altogether, our data disentangle the role of infection-induced inflammatory cytokines in driving the expansion of downstream hematopoietic progenitors.


Assuntos
Toxoplasma , Toxoplasmose , Gravidez , Feminino , Humanos , Células-Tronco Hematopoéticas , Diferenciação Celular , Toxoplasmose/metabolismo , Inflamação
4.
Curr Opin Hematol ; 30(4): 130-136, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37254855

RESUMO

PURPOSE OF REVIEW: Inflammation is now recognized as a major regulator of hematopoietic stem cell (HSC) function. Adult hematopoietic stem cells can adaptively modulate hematopoietic output in direct response to acute infection and inflammation. Conversely, prolonged exposure to inflammation can drive impaired HSC function, clonal expansion, and malignant transformation. As compared with adult hematopoiesis, the effects of prenatal inflammation on developing hematopoietic stem cells are understudied. RECENT FINDINGS: Inflammatory cues directly activate adult HSCs in the bone marrow, but the response of fetal HSCs to maternal inflammation is underexplored. Recent evidence demonstrates that maternal inflammation can be detected by fetal hematopoietic stem and progenitor cells (HSPCs) within the fetal liver and that the same inflammatory cues evoke fundamentally distinct responses during development. The responses of developing stem and progenitor cells and the specialized immune cells they produce have important implications for postnatal hematopoietic output and immune function. SUMMARY: We discuss recent insights into the response of fetal hematopoiesis to prenatal inflammation and examine how recent discoveries regarding the contribution of fetal hematopoiesis to the adult hematopoietic system will influence future studies.


Assuntos
Medula Óssea , Células-Tronco Hematopoéticas , Humanos , Medula Óssea/patologia , Células-Tronco Hematopoéticas/patologia , Hematopoese , Inflamação/patologia
5.
J Exp Med ; 220(4)2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36811605

RESUMO

Evolutionarily conserved, "natural" (n)IgM is broadly reactive to both self and foreign antigens. Its selective deficiency leads to increases in autoimmune diseases and infections. In mice, nIgM is secreted independent of microbial exposure to bone marrow (BM) and spleen B-1 cell-derived plasma cells (B-1PC), generating the majority of nIgM, or by B-1 cells that remain non-terminally differentiated (B-1sec). Thus, it has been assumed that the nIgM repertoire is broadly reflective of the repertoire of body cavity B-1 cells. Studies here reveal, however, that B-1PC generate a distinct, oligoclonal nIgM repertoire, characterized by short CDR3 variable immunoglobulin heavy chain regions, 7-8 amino acids in length, some public, many arising from convergent rearrangements, while specificities previously associated with nIgM were generated by a population of IgM-secreting B-1 (B-1sec). BM, but not spleen B-1PC, or B-1sec also required the presence of TCRαß CD4 T cells for their development from fetal precursors. Together, the studies identify important previously unknown characteristics of the nIgM pool.


Assuntos
Subpopulações de Linfócitos B , Camundongos , Animais , Linfócitos B , Imunoglobulina M , Linfócitos T CD4-Positivos , Plasmócitos
6.
Methods Mol Biol ; 2567: 99-112, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36255697

RESUMO

Hematopoietic stem cells (HSCs) are responsible for the generation and maintenance of pools of multipotent precursors that ultimately give rise to all fully differentiated blood and immune cells. Proper identification and isolation of HSCs for functional analysis has greatly facilitated our understanding of both normal and abnormal adult hematopoiesis. Whereas adult hematopoiesis in mice and humans is driven by quiescent HSCs that reside almost exclusively within the bone marrow (BM), developmental hematopoiesis is characterized by a series of transient progenitors driving waves of increasingly mature hematopoietic cell production that occur across multiple anatomical sites. These waves of hematopoietic cell production are also responsible for the generation of distinct immune cell populations during development that persist into adulthood and contribute uniquely to adult immunity. Therefore, methods to properly isolate and characterize fetal progenitors with high purity across development become increasingly important not only for defining developmental hematopoietic pathways, but also for understanding the contribution of developmental hematopoiesis to the immune system. Here, we describe and discuss methods and considerations for the isolation and characterization of HSCs from the fetal liver, the primary hematopoietic organ during fetal development.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Humanos , Adulto , Camundongos , Animais , Células-Tronco Hematopoéticas/metabolismo , Medula Óssea , Diferenciação Celular , Fígado/metabolismo
7.
Cell Rep ; 41(8): 111677, 2022 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-36417858

RESUMO

Adult hematopoietic stem and progenitor cells (HSPCs) respond directly to inflammation and infection, causing both acute and persistent changes to quiescence, mobilization, and differentiation. Here we show that murine fetal HSPCs respond to prenatal inflammation in utero and that the fetal response shapes postnatal hematopoiesis and immune cell function. Heterogeneous fetal HSPCs show divergent responses to maternal immune activation (MIA), including changes in quiescence, expansion, and lineage-biased output. Single-cell transcriptomic analysis of fetal HSPCs in response to MIA reveals specific upregulation of inflammatory gene profiles in discrete, transient hematopoietic stem cell (HSC) populations that propagate expansion of lymphoid-biased progenitors. Beyond fetal development, MIA causes the inappropriate expansion and persistence of fetal lymphoid-biased progenitors postnatally, concomitant with increased cellularity and hyperresponsiveness of fetal-derived innate-like lymphocytes. Our investigation demonstrates how inflammation in utero can direct the output and function of fetal-derived immune cells by reshaping fetal HSC establishment.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Gravidez , Feminino , Camundongos , Animais , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Feto , Inflamação/metabolismo , Desenvolvimento Fetal
8.
Development ; 149(8)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35072209

RESUMO

Tissue-resident lymphoid cells (TLCs) span the spectrum of innate-to-adaptive immune function. Unlike traditional, circulating lymphocytes that are continuously generated from hematopoietic stem cells (HSCs), many TLCs are of fetal origin and poorly generated from adult HSCs. Here, we sought to further understand murine TLC development and the roles of Flk2 and IL7Rα, two cytokine receptors with known function in traditional lymphopoiesis. Using Flk2- and Il7r-Cre lineage tracing, we found that peritoneal B1a cells, splenic marginal zone B (MZB) cells, lung ILC2s and regulatory T cells (Tregs) were highly labeled. Despite high labeling, loss of Flk2 minimally affected the generation of these cells. In contrast, loss of IL7Rα, or combined deletion of Flk2 and IL7Rα, dramatically reduced the number of B1a cells, MZBs, ILC2s and Tregs, both in situ and upon transplantation, indicating an intrinsic and essential role for IL7Rα. Surprisingly, reciprocal transplants of wild-type HSCs showed that an IL7Rα-/- environment selectively impaired reconstitution of TLCs when compared with TLC numbers in situ. Taken together, our data defined Flk2- and IL7Rα-positive TLC differentiation paths, and revealed functional roles of Flk2 and IL7Rα in TLC establishment.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Linfopoese/genética , Receptores de Interleucina-7/genética , Tirosina Quinase 3 Semelhante a fms/genética , Imunidade Adaptativa/genética , Animais , Linfócitos B/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Células-Tronco Hematopoéticas/citologia , Imunidade Inata/genética , Linfócitos/citologia , Linfócitos/imunologia , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Linfopoese/imunologia , Camundongos , Especificidade de Órgãos/genética , Linfócitos T Reguladores/imunologia
9.
Cells ; 10(12)2021 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-34944105

RESUMO

Emerging evidence indicates that perinatal infection and inflammation can influence the developing immune system and may ultimately affect long-term health and disease outcomes in offspring by perturbing tissue and immune homeostasis. We posit that perinatal inflammation influences immune outcomes in offspring by perturbing (1) the development and function of fetal-derived immune cells that regulate tissue development and homeostasis, and (2) the establishment and function of developing hematopoietic stem cells (HSCs) that continually generate immune cells across the lifespan. To disentangle the complexities of these interlinked systems, we propose the cochlea as an ideal model tissue to investigate how perinatal infection affects immune, tissue, and stem cell development. The cochlea contains complex tissue architecture and a rich immune milieu that is established during early life. A wide range of congenital infections cause cochlea dysfunction and sensorineural hearing loss (SNHL), likely attributable to early life inflammation. Furthermore, we show that both immune cells and bone marrow hematopoietic progenitors can be simultaneously analyzed within neonatal cochlear samples. Future work investigating the pathogenesis of SNHL in the context of congenital infection will therefore provide critical information on how perinatal inflammation drives disease susceptibility in offspring.


Assuntos
Cóclea/patologia , Hematopoese , Sistema Imunitário/crescimento & desenvolvimento , Inflamação/patologia , Animais , Feto/imunologia , Perda Auditiva Neurossensorial/imunologia , Humanos
10.
J Exp Med ; 218(10)2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34529750

RESUMO

Therapeutic discovery for mantle cell lymphoma (MCL) has been hindered by a lack of preclinical mouse models that recapitulate human disease. In this issue, Pieters and colleagues (2021. J. Exp. Med.https://doi.org/10.1084/jem.20202280) establish a novel mouse model of MCL driven by overexpression of cyclin D2 and identify fetal-derived B1a cells as putative cell of origin for MCL.


Assuntos
Linfoma de Célula do Manto , Animais , Ciclina D1 , Modelos Animais de Doenças , Camundongos
11.
Cell Stem Cell ; 27(5): 695-697, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33157043

RESUMO

Seeking to define the "switch" from fetal to adult hematopoiesis, Li et al. (2020) performed extensive genomic and epigenomic profiling of hematopoietic stem and progenitor cells across ontogeny (as explored in this issue of Cell Stem Cell). Gradual and stochastic changes in genomic and epigenomic regulation suggest the absence of any specific regulator.


Assuntos
Células-Tronco Hematopoéticas , Análise de Célula Única , Epigenômica , Feto , Hematopoese/genética
12.
Front Immunol ; 11: 123, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117273

RESUMO

Over the last century, the alarming surge in allergy and autoimmune disease has led to the hypothesis that decreasing exposure to microbes, which has accompanied industrialization and modern life in the Western world, has fundamentally altered the immune response. In its current iteration, the "hygiene hypothesis" suggests that reduced microbial exposures during early life restricts the production and differentiation of immune cells suited for immune regulation. Although it is now well-appreciated that the increase in hypersensitivity disorders represents a "perfect storm" of many contributing factors, we argue here that two important considerations have rarely been explored. First, the window of microbial exposure that impacts immune development is not limited to early childhood, but likely extends into the womb. Second, restricted microbial interactions by an expectant mother will bias the fetal immune system toward hypersensitivity. Here, we extend this discussion to hypothesize that the cell types sensing microbial exposures include fetal hematopoietic stem cells, which drive long-lasting changes to immunity.


Assuntos
Feto/imunologia , Hipótese da Higiene , Hipersensibilidade/imunologia , Sistema Imunitário/imunologia , Adulto , Criança , Feminino , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/microbiologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Recém-Nascido , Inflamação/imunologia , Interações Microbianas/imunologia , Gravidez , Linfócitos T Auxiliares-Indutores/imunologia
13.
Development ; 146(14)2019 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-31332039

RESUMO

The discovery of a fetal origin for tissue-resident macrophages (trMacs) has inspired an intense search for the mechanisms underlying their development. Here, we performed in vivo lineage tracing of cells with an expression history of IL7Rα, a marker exclusively associated with the lymphoid lineage in adult hematopoiesis. Surprisingly, we found that Il7r-Cre labeled fetal-derived, adult trMacs. Labeling was almost complete in some tissues and partial in others. The putative progenitors of trMacs, yolk sac (YS) erythromyeloid progenitors, did not express IL7R, and YS hematopoiesis was unperturbed in IL7R-deficient mice. In contrast, tracking of IL7Rα message levels, surface expression, and Il7r-Cre-mediated labeling across fetal development revealed dynamic regulation of Il7r mRNA expression and rapid upregulation of IL7Rα surface protein upon transition from monocyte to macrophage within fetal tissues. Fetal monocyte differentiation in vitro produced IL7R+ macrophages, supporting a direct progenitor-progeny relationship. Additionally, blockade of IL7R function during late gestation specifically impaired the establishment of fetal-derived trMacs in vivo These data provide evidence for a distinct function of IL7Rα in fetal myelopoiesis and identify IL7R as a novel regulator of trMac development.


Assuntos
Diferenciação Celular/genética , Linhagem da Célula/genética , Macrófagos/fisiologia , Mielopoese/genética , Receptores de Interleucina-7/fisiologia , Animais , Embrião de Mamíferos , Feminino , Feto/metabolismo , Hematopoese/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez
14.
Stem Cell Reports ; 12(4): 801-815, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30905737

RESUMO

Hematopoiesis is arguably one of the best understood stem cell systems; however, significant challenges remain to reach a consensus understanding of the lineage potential, heterogeneity, and relationships of hematopoietic stem and progenitor cell populations. To gain new insights, we performed quantitative analyses of mature cell production from hematopoietic stem cells (HSCs) and multiple hematopoietic progenitor populations. Assessment of the absolute numbers of mature cell types produced by each progenitor cell revealed a striking erythroid dominance of all myeloid-competent progenitors assessed, accompanied by strong platelet reconstitution. All populations with myeloid potential also produced robust numbers of red blood cells and platelets in vivo. Clonal analysis by single-cell transplantation and by spleen colony assays revealed that a significant fraction of HSCs and multipotent progenitors have multilineage potential at the single-cell level. These new insights prompt an erythroid-focused model of hematopoietic differentiation.


Assuntos
Diferenciação Celular , Eritropoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Animais , Biomarcadores , Linhagem da Célula , Ensaio de Unidades Formadoras de Colônias , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Imunofenotipagem , Camundongos , Modelos Biológicos
15.
J Immunol ; 201(1): 31-40, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29743314

RESUMO

CD8 T cells can play both a protective and pathogenic role in inflammation and autoimmune development. Recent studies have highlighted the ability of CD8 T cells to function as T follicular helper (Tfh) cells in the germinal center in the context of infection. However, whether this phenomenon occurs in autoimmunity and contributes to autoimmune pathogenesis is largely unexplored. In this study, we show that CD8 T cells acquire a CD4 Tfh profile in the absence of functional regulatory T cells in both the IL-2-deficient and scurfy mouse models. Depletion of CD8 T cells mitigates autoimmune pathogenesis in IL-2-deficient mice. CD8 T cells express the B cell follicle-localizing chemokine receptor CXCR5, a principal Tfh transcription factor Bcl6, and the Tfh effector cytokine IL-21. CD8 T cells localize to the B cell follicle, express B cell costimulatory proteins, and promote B cell differentiation and Ab isotype class switching. These data reveal a novel contribution of autoreactive CD8 T cells to autoimmune disease, in part, through CD4 follicular-like differentiation and functionality.


Assuntos
Anemia Hemolítica Autoimune/imunologia , Anemia Hemolítica Autoimune/patologia , Linfócitos T CD8-Positivos/imunologia , Switching de Imunoglobulina/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Autoimunidade/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Eritrócitos/imunologia , Feminino , Interleucina-2/genética , Interleucinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Receptores CXCR5/metabolismo
16.
Cell Stem Cell ; 22(3): 285-287, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29499144

RESUMO

The inability to derive multipotent hematopoietic stem cells in vitro stems in part from a limited understanding of how multipotency is acquired during development. Recently in Nature,Vo et al. (2018) reveal the epigenetic enzyme Ezh1 as a master regulator of multipotency during hematopoietic stem cell development.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Desenvolvimento Embrionário , Transplante de Células-Tronco Hematopoéticas
17.
Nucleic Acids Res ; 46(10): e62, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29548006

RESUMO

RNA-sequencing (RNA-seq) is a powerful technique to investigate and quantify entire transcriptomes. Recent advances in the field have made it possible to explore the transcriptomes of single cells. However, most widely used RNA-seq protocols fail to provide crucial information regarding transcription start sites. Here we present a protocol, Tn5Prime, that takes advantage of the Tn5 transposase-based Smart-seq2 protocol to create RNA-seq libraries that capture the 5' end of transcripts. The Tn5Prime method dramatically streamlines the 5' capture process and is both cost effective and reliable. By applying Tn5Prime to bulk RNA and single cell samples, we were able to define transcription start sites as well as quantify transcriptomes at high accuracy and reproducibility. Additionally, similar to 3' end-based high-throughput methods like Drop-seq and 10× Genomics Chromium, the 5' capture Tn5Prime method allows the introduction of cellular identifiers during reverse transcription, simplifying the analysis of large numbers of single cells. In contrast to 3' end-based methods, Tn5Prime also enables the assembly of the variable 5' ends of the antibody sequences present in single B-cell data. Therefore, Tn5Prime presents a robust tool for both basic and applied research into the adaptive immune system and beyond.


Assuntos
Linfócitos B/citologia , Análise de Sequência de RNA/métodos , Sítio de Iniciação de Transcrição , Transposases/genética , ADP-Ribosil Ciclase 1/genética , Adulto , Animais , Linfócitos B/fisiologia , Linhagem Celular , Perfilação da Expressão Gênica , Biblioteca Gênica , Humanos , Glicoproteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Análise de Célula Única/métodos , Transposases/metabolismo , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética
18.
Blood ; 128(24): 2765-2769, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27799163

RESUMO

Hematopoietic stem cells (HSCs) have long been considered the continuous source of all hematopoietic cells for the life of an individual. Recent findings have questioned multiple aspects of this view, including the ability of lifelong HSCs to contribute to tissue-resident immune cells. Here we discuss the most recent findings on the source of B1a cells, innatelike lymphocytes that primarily reside in serous cavities. Powerful experimental approaches including bar coding, single cell transplantation, in vivo lineage tracing, and HSC-specific pulse-chase labeling have provided novel insights on B1a-cell generation during ontogeny. We evaluate the evidence for fetal vs adult B1a-cell production capacity and the identity of putative cells of origin. Integrating these most recent findings with previous work, we propose a working model that encapsulates our current understanding of waves of immune development.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Linfócitos/citologia , Animais , Linhagem da Célula , Feto/citologia , Humanos , Fígado/citologia , Fígado/embriologia , Modelos Biológicos
19.
Cell Stem Cell ; 19(6): 768-783, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27666010

RESUMO

The generation of distinct hematopoietic cell types, including tissue-resident immune cells, distinguishes fetal from adult hematopoiesis. However, the mechanisms underlying differential cell production to generate a layered immune system during hematopoietic development are unclear. Using an irreversible lineage-tracing model, we identify a definitive hematopoietic stem cell (HSC) that supports long-term multilineage reconstitution upon transplantation into adult recipients but does not persist into adulthood in situ. These HSCs are fully multipotent, yet they display both higher lymphoid cell production and greater capacity to generate innate-like B and T lymphocytes as compared to coexisting fetal HSCs and adult HSCs. Thus, these developmentally restricted HSCs (drHSCs) define the origin and generation of early lymphoid cells that play essential roles in establishing self-recognition and tolerance, with important implications for understanding autoimmune disease, allergy, and rejection of transplanted organs.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Desenvolvimento Fetal , Células-Tronco Hematopoéticas/citologia , Imunidade Inata , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Linhagem da Célula , Microambiente Celular , Senescência Celular , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Hematopoéticas/imunologia , Fígado/citologia , Fígado/embriologia , Camundongos , Análise de Sequência de RNA , Timo/citologia
20.
Immunity ; 42(4): 665-78, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25902481

RESUMO

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.


Assuntos
Envelhecimento/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Células Progenitoras Mieloides/imunologia , Proteínas Proto-Oncogênicas c-myb/imunologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula/imunologia , Rastreamento de Células , Embrião de Mamíferos , Feminino , Feto , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Rim/citologia , Rim/imunologia , Fígado/citologia , Fígado/imunologia , Pulmão/citologia , Pulmão/imunologia , Macrófagos/citologia , Camundongos , Microglia/citologia , Microglia/imunologia , Monócitos/citologia , Células Progenitoras Mieloides/citologia , Gravidez , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-myb/metabolismo , Pele/citologia , Pele/imunologia , Saco Vitelino/citologia , Saco Vitelino/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA