Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Cell ; 187(12): 3090-3107.e21, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38749423

RESUMEN

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.


Asunto(s)
Envejecimiento , Plaquetas , Diferenciación Celular , Células Madre Hematopoyéticas , Trombosis , Animales , Células Madre Hematopoyéticas/metabolismo , Plaquetas/metabolismo , Trombosis/patología , Trombosis/metabolismo , Ratones , Humanos , Megacariocitos/metabolismo , Ratones Endogámicos C57BL , Células Progenitoras de Megacariocitos/metabolismo , Masculino
2.
EMBO J ; 42(14): e112693, 2023 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-37259639

RESUMEN

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.


Asunto(s)
Toxoplasma , Toxoplasmosis , Embarazo , Femenino , Humanos , Células Madre Hematopoyéticas , Diferenciación Celular , Toxoplasmosis/metabolismo , Inflamación
3.
Development ; 149(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35072209

RESUMEN

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.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Linfopoyesis/genética , Receptores de Interleucina-7/genética , Tirosina Quinasa 3 Similar a fms/genética , Inmunidad Adaptativa/genética , Animales , Linfocitos B/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Regulación del Desarrollo de la Expresión Génica/genética , Células Madre Hematopoyéticas/citología , Inmunidad Innata/genética , Linfocitos/citología , Linfocitos/inmunología , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Linfopoyesis/inmunología , Ratones , Especificidad de Órganos/genética , Linfocitos T Reguladores/inmunología
4.
Immunity ; 42(4): 665-78, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25902481

RESUMEN

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


Asunto(s)
Envejecimiento/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Células Progenitoras Mieloides/inmunología , Proteínas Proto-Oncogénicas c-myb/inmunología , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula/inmunología , Rastreo Celular , Embrión de Mamíferos , Femenino , Feto , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Riñón/citología , Riñón/inmunología , Hígado/citología , Hígado/inmunología , Pulmón/citología , Pulmón/inmunología , Macrófagos/citología , Ratones , Microglía/citología , Microglía/inmunología , Monocitos/citología , Células Progenitoras Mieloides/citología , Embarazo , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-myb/metabolismo , Piel/citología , Piel/inmunología , Saco Vitelino/citología , Saco Vitelino/inmunología
5.
Curr Opin Hematol ; 30(4): 130-136, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37254855

RESUMEN

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.


Asunto(s)
Médula Ósea , Células Madre Hematopoyéticas , Humanos , Médula Ósea/patología , Células Madre Hematopoyéticas/patología , Hematopoyesis , Inflamación/patología
6.
Immunity ; 40(1): 91-104, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439267

RESUMEN

Cardiac macrophages are crucial for tissue repair after cardiac injury but are not well characterized. Here we identify four populations of cardiac macrophages. At steady state, resident macrophages were primarily maintained through local proliferation. However, after macrophage depletion or during cardiac inflammation, Ly6c(hi) monocytes contributed to all four macrophage populations, whereas resident macrophages also expanded numerically through proliferation. Genetic fate mapping revealed that yolk-sac and fetal monocyte progenitors gave rise to the majority of cardiac macrophages, and the heart was among a minority of organs in which substantial numbers of yolk-sac macrophages persisted in adulthood. CCR2 expression and dependence distinguished cardiac macrophages of adult monocyte versus embryonic origin. Transcriptional and functional data revealed that monocyte-derived macrophages coordinate cardiac inflammation, while playing redundant but lesser roles in antigen sampling and efferocytosis. These data highlight the presence of multiple cardiac macrophage subsets, with different functions, origins, and strategies to regulate compartment size.


Asunto(s)
Macrófagos/inmunología , Monocitos/fisiología , Miocarditis/inmunología , Miocardio/inmunología , Animales , Presentación de Antígeno , Antígenos Ly/metabolismo , Muerte Celular , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Desarrollo Fetal , Corazón/embriología , Homeostasis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/inmunología , Fagocitosis , Receptores CCR2/metabolismo , Transcriptoma , Saco Vitelino/citología
7.
Development ; 146(14)2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-31332039

RESUMEN

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.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Macrófagos/fisiología , Mielopoyesis/genética , Receptores de Interleucina-7/fisiología , Animales , Embrión de Mamíferos , Femenino , Feto/metabolismo , Hematopoyesis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo
8.
J Immunol ; 201(1): 31-40, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29743314

RESUMEN

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.


Asunto(s)
Anemia Hemolítica Autoinmune/inmunología , Anemia Hemolítica Autoinmune/patología , Linfocitos T CD8-positivos/inmunología , Cambio de Clase de Inmunoglobulina/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Autoinmunidad/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Eritrocitos/inmunología , Femenino , Interleucina-2/genética , Interleucinas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Receptores CXCR5/metabolismo
9.
Nucleic Acids Res ; 46(10): e62, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29548006

RESUMEN

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.


Asunto(s)
Linfocitos B/citología , Análisis de Secuencia de ARN/métodos , Sitio de Iniciación de la Transcripción , Transposasas/genética , ADP-Ribosil Ciclasa 1/genética , Adulto , Animales , Linfocitos B/fisiología , Línea Celular , Perfilación de la Expresión Génica , Biblioteca de Genes , Humanos , Glicoproteínas de Membrana/genética , Ratones Endogámicos C57BL , Análisis de la Célula Individual/métodos , Transposasas/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
10.
Blood ; 137(8): 1003-1004, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33630049
11.
Blood ; 128(24): 2765-2769, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27799163

RESUMEN

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.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Linfocitos/citología , Animales , Linaje de la Célula , Feto/citología , Humanos , Hígado/citología , Hígado/embriología , Modelos Biológicos
12.
Cell Rep ; 43(7): 114365, 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38909363

RESUMEN

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.

13.
J Nutr ; 143(7): 1028-35, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23700346

RESUMEN

Impaired folate-mediated one-carbon metabolism (OCM) has emerged as a risk factor for several diseases associated with age-related cognitive decline, but the underlying mechanisms remain unknown and thus hinder the identification of subpopulations most vulnerable to OCM disruption. Here we investigated the role of serine hydroxymethyltransferase 1 (SHMT1), a folate-dependent enzyme regulating de novo thymidylate biosynthesis, in influencing neuronal and cognitive function in the adult mouse. We observed Shmt1 expression in the hippocampus, including the granule cell layer of the dentate gyrus (DG), and examined hippocampal neurogenesis and hippocampal-dependent fear conditioning in mice deficient for Shmt1. We used a 3 × 3 design in which adult male Shmt1(+/+), Shmt1(+/-), and Shmt1(-/-) mice were fed folic acid control (2 mg/kg), folic acid-deficient (0 mg/kg), or folic acid-supplemented (8 mg/kg) diets from weaning through the duration of the study. Proliferation within the DG was elevated by 70% in Shmt1(+/-) mice, yet the number of newborn mature neurons was reduced by 98% compared with that in Shmt1(+/+) mice. Concomitant with these alterations, Shmt1(+/-) mice showed a 45% reduction in mnemonic recall during trace fear conditioning. Dietary folate manipulations alone did not influence neural outcomes. Together, these data identify SHMT1 as one of the first enzymes within the OCM pathway to regulate neuronal and cognitive profiles and implicate impaired thymidylate biosynthesis in the etiology of folate-related neuropathogenesis.


Asunto(s)
Glicina Hidroximetiltransferasa/genética , Hipocampo/patología , Memoria/fisiología , Neurogénesis/genética , Animales , Proliferación Celular , Giro Dentado/metabolismo , Miedo/fisiología , Ácido Fólico/administración & dosificación , Ácido Fólico/sangre , Deficiencia de Ácido Fólico/patología , Regulación de la Expresión Génica , Glicina Hidroximetiltransferasa/metabolismo , Hipocampo/metabolismo , Homocisteína/metabolismo , Hibridación in Situ , Masculino , Ratones , Ratones Noqueados , Neuronas/citología , Neuronas/metabolismo , Factores de Riesgo , Timidina Monofosfato/biosíntesis
14.
Methods Mol Biol ; 2567: 99-112, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36255697

RESUMEN

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.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , Humanos , Adulto , Ratones , Animales , Células Madre Hematopoyéticas/metabolismo , Médula Ósea , Diferenciación Celular , Hígado/metabolismo
15.
bioRxiv ; 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-38045298

RESUMEN

Allergic asthma is a chronic respiratory disease that initiates in early life, but causal mechanisms are poorly understood. Here we examined how prenatal inflammation shapes allergic asthma susceptibility by reprogramming lung immunity from early development. Induction of Type I interferon-mediated inflammation during development provoked expansion and hyperactivation of group 2 innate lymphoid cells (ILC2s) seeding the developing lung. Hyperactivated ILC2s produced increased IL-5 and IL-13, and were associated with acute Th2 bias, eosinophilia, and decreased Tregs in the lung. The hyperactive ILC2 phenotype was recapitulated by adoptive transfer of a fetal liver precursor following exposure to prenatal inflammation, indicative of developmental programming. Programming of ILC2 function and subsequent lung immune remodeling by prenatal inflammation led to airway dysfunction at baseline and in response to papain, indicating increased asthma susceptibility. Our data provide a link by which developmental programming of progenitors by early-life inflammation drives lung immune remodeling and asthma susceptibility through hyperactivation of lung-resident ILC2s. One Sentence Summary: Prenatal inflammation programs asthma susceptibility by inducing the production of hyperactivated ILC2s in the developing lung.

16.
J Exp Med ; 220(4)2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36811605

RESUMEN

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.


Asunto(s)
Subgrupos de Linfocitos B , Ratones , Animales , Linfocitos B , Inmunoglobulina M , Linfocitos T CD4-Positivos , Células Plasmáticas
17.
Cell Rep ; 41(8): 111677, 2022 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-36417858

RESUMEN

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.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas , Embarazo , Femenino , Ratones , Animales , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Feto , Inflamación/metabolismo , Desarrollo Fetal
18.
Nat Cardiovasc Res ; 1(1): 67-84, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35599984

RESUMEN

Leukocytes and endothelial cells frequently cooperate to resolve inflammatory events. In most cases, these interactions are transient in nature and triggered by immunological insults. Here, we report that in areas of disturbed blood flow, aortic endothelial cells permanently and intimately associate with a population of specialized macrophages that are recruited at birth from the closing ductus arteriosus and share the luminal surface with the endothelium becoming interwoven in the tunica intima. Anatomical changes that affect hemodynamics, like in patent ductus arteriosus, alter macrophage seeding to coincide with regions of disturbed flow. Aortic resident macrophages expand in situ via direct cell renewal. Induced-depletion of intimal macrophages led to thrombin-mediated endothelial cell contraction, progressive fibrin accumulation and formation of microthrombi that, once dislodged, caused blockade of vessels in several organs. Together the findings revealed that intravascular resident macrophages are essential to regulate thrombin activity and clear fibrin deposits in regions of disturbed blood flow.

19.
J Exp Med ; 218(10)2021 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-34529750

RESUMEN

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.


Asunto(s)
Linfoma de Células del Manto , Animales , Ciclina D1 , Modelos Animales de Enfermedad , Ratones
20.
Cells ; 10(12)2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34944105

RESUMEN

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.


Asunto(s)
Cóclea/patología , Hematopoyesis , Sistema Inmunológico/crecimiento & desarrollo , Inflamación/patología , Animales , Feto/inmunología , Pérdida Auditiva Sensorineural/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA