Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.758
Filtrar
Mais filtros

Coleção Fiocruz
Intervalo de ano de publicação
1.
Nat Immunol ; 23(7): 1109-1120, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35761081

RESUMO

Nonimmune cells can have immunomodulatory roles that contribute to healthy development. However, the molecular and cellular mechanisms underlying the immunomodulatory functions of erythroid cells during human ontogenesis remain elusive. Here, integrated, single-cell transcriptomic studies of erythroid cells from the human yolk sac, fetal liver, preterm umbilical cord blood (UCB), term UCB and adult bone marrow (BM) identified classical and immune subsets of erythroid precursors with divergent differentiation trajectories. Immune-erythroid cells were present from the yolk sac to the adult BM throughout human ontogenesis but failed to be generated in vitro from human embryonic stem cells. Compared with classical-erythroid precursors, these immune-erythroid cells possessed dual erythroid and immune regulatory networks, showed immunomodulatory functions and interacted more frequently with various innate and adaptive immune cells. Our findings provide important insights into the nature of immune-erythroid cells and their roles during development and diseases.


Assuntos
Células Precursoras Eritroides , Transcriptoma , Adulto , Diferenciação Celular/genética , Células Eritroides , Sangue Fetal , Humanos , Recém-Nascido , Saco Vitelino
2.
Cell ; 179(4): 937-952.e18, 2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31675500

RESUMO

Cell-cell junctions respond to mechanical forces by changing their organization and function. To gain insight into the mechanochemical basis underlying junction mechanosensitivity, we analyzed tight junction (TJ) formation between the enveloping cell layer (EVL) and the yolk syncytial layer (YSL) in the gastrulating zebrafish embryo. We found that the accumulation of Zonula Occludens-1 (ZO-1) at TJs closely scales with tension of the adjacent actomyosin network, revealing that these junctions are mechanosensitive. Actomyosin tension triggers ZO-1 junctional accumulation by driving retrograde actomyosin flow within the YSL, which transports non-junctional ZO-1 clusters toward the TJ. Non-junctional ZO-1 clusters form by phase separation, and direct actin binding of ZO-1 is required for stable incorporation of retrogradely flowing ZO-1 clusters into TJs. If the formation and/or junctional incorporation of ZO-1 clusters is impaired, then TJs lose their mechanosensitivity, and consequently, EVL-YSL movement is delayed. Thus, phase separation and flow of non-junctional ZO-1 confer mechanosensitivity to TJs.


Assuntos
Desenvolvimento Embrionário/genética , Mecanotransdução Celular/genética , Junções Íntimas/genética , Proteína da Zônula de Oclusão-1/genética , Citoesqueleto de Actina/genética , Actomiosina/genética , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/crescimento & desenvolvimento , Embrião não Mamífero/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Proteínas de Membrana/genética , Camundongos , Fosfoproteínas/genética , Ligação Proteica , Junções Íntimas/fisiologia , Saco Vitelino/crescimento & desenvolvimento , Saco Vitelino/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
3.
Immunity ; 54(7): 1433-1446.e5, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34062116

RESUMO

The extra-embryonic yolk sac contains the first definitive multipotent hematopoietic cells, denominated erythromyeloid progenitors. They originate in situ prior to the emergence of hematopoietic stem cells and give rise to erythroid, monocytes, granulocytes, mast cells and macrophages, the latter in a Myb transcription factor-independent manner. We uncovered here the heterogeneity of yolk sac erythromyeloid progenitors, at the single cell level, and discriminated multipotent from committed progenitors, prior to fetal liver colonization. We identified two temporally distinct megakaryocyte differentiation pathways. The first occurs in the yolk sac, bypasses intermediate bipotent megakaryocyte-erythroid progenitors and, similar to the differentiation of macrophages, is Myb-independent. By contrast, the second originates later, from Myb-dependent bipotent progenitors expressing Csf2rb and colonize the fetal liver, where they give rise to megakaryocytes and to large numbers of erythrocytes. Understanding megakaryocyte development is crucial as they play key functions during vascular development, in particular in separating blood and lymphatic networks.


Assuntos
Diferenciação Celular/fisiologia , Eritrócitos/citologia , Megacariócitos/citologia , Células Mieloides/citologia , Células-Tronco/citologia , Saco Vitelino/citologia , Animais , Linhagem da Célula/fisiologia , Células Cultivadas , Embrião de Mamíferos/citologia , Feminino , Granulócitos/citologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Células-Tronco Multipotentes/citologia , Gravidez
4.
Nature ; 626(7998): 367-376, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38092041

RESUMO

Implantation of the human embryo begins a critical developmental stage that comprises profound events including axis formation, gastrulation and the emergence of haematopoietic system1,2. Our mechanistic knowledge of this window of human life remains limited due to restricted access to in vivo samples for both technical and ethical reasons3-5. Stem cell models of human embryo have emerged to help unlock the mysteries of this stage6-16. Here we present a genetically inducible stem cell-derived embryoid model of early post-implantation human embryogenesis that captures the reciprocal codevelopment of embryonic tissue and the extra-embryonic endoderm and mesoderm niche with early haematopoiesis. This model is produced from induced pluripotent stem cells and shows unanticipated self-organizing cellular programmes similar to those that occur in embryogenesis, including the formation of amniotic cavity and bilaminar disc morphologies as well as the generation of an anterior hypoblast pole and posterior domain. The extra-embryonic layer in these embryoids lacks trophoblast and shows advanced multilineage yolk sac tissue-like morphogenesis that harbours a process similar to distinct waves of haematopoiesis, including the emergence of erythroid-, megakaryocyte-, myeloid- and lymphoid-like cells. This model presents an easy-to-use, high-throughput, reproducible and scalable platform to probe multifaceted aspects of human development and blood formation at the early post-implantation stage. It will provide a tractable human-based model for drug testing and disease modelling.


Assuntos
Desenvolvimento Embrionário , Camadas Germinativas , Hematopoese , Saco Vitelino , Humanos , Implantação do Embrião , Endoderma/citologia , Endoderma/embriologia , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Saco Vitelino/citologia , Saco Vitelino/embriologia , Mesoderma/citologia , Mesoderma/embriologia , Células-Tronco Pluripotentes Induzidas/citologia , Âmnio/citologia , Âmnio/embriologia , Corpos Embrioides/citologia , Linhagem da Célula , Biologia do Desenvolvimento/métodos , Biologia do Desenvolvimento/tendências
5.
Nature ; 622(7983): 562-573, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37673118

RESUMO

The ability to study human post-implantation development remains limited owing to ethical and technical challenges associated with intrauterine development after implantation1. Embryo-like models with spatially organized morphogenesis and structure of all defining embryonic and extra-embryonic tissues of the post-implantation human conceptus (that is, the embryonic disc, the bilaminar disc, the yolk sac, the chorionic sac and the surrounding trophoblast layer) remain lacking1,2. Mouse naive embryonic stem cells have recently been shown to give rise to embryonic and extra-embryonic stem cells capable of self-assembling into post-gastrulation structured stem-cell-based embryo models with spatially organized morphogenesis (called SEMs)3. Here we extend those findings to humans using only genetically unmodified human naive embryonic stem cells (cultured in human enhanced naive stem cell medium conditions)4. Such human fully integrated and complete SEMs recapitulate the organization of nearly all known lineages and compartments of post-implantation human embryos, including the epiblast, the hypoblast, the extra-embryonic mesoderm and the trophoblast layer surrounding the latter compartments. These human complete SEMs demonstrated developmental growth dynamics that resemble key hallmarks of post-implantation stage embryogenesis up to 13-14 days after fertilization (Carnegie stage 6a). These include embryonic disc and bilaminar disc formation, epiblast lumenogenesis, polarized amniogenesis, anterior-posterior symmetry breaking, primordial germ-cell specification, polarized yolk sac with visceral and parietal endoderm formation, extra-embryonic mesoderm expansion that defines a chorionic cavity and a connecting stalk, and a trophoblast-surrounding compartment demonstrating syncytium and lacunae formation. This SEM platform will probably enable the experimental investigation of previously inaccessible windows of human early post implantation up to peri-gastrulation development.


Assuntos
Implantação do Embrião , Embrião de Mamíferos , Desenvolvimento Embrionário , Células-Tronco Embrionárias Humanas , Humanos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Fertilização , Gastrulação , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Células-Tronco Embrionárias Humanas/citologia , Trofoblastos/citologia , Saco Vitelino/citologia , Saco Vitelino/embriologia , Células Gigantes/citologia
6.
Nature ; 622(7983): 574-583, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37369348

RESUMO

Investigating human development is a substantial scientific challenge due to the technical and ethical limitations of working with embryonic samples. In the face of these difficulties, stem cells have provided an alternative to experimentally model inaccessible stages of human development in vitro1-13. Here we show that human pluripotent stem cells can be triggered to self-organize into three-dimensional structures that recapitulate some key spatiotemporal events of early human post-implantation embryonic development. Our system reproducibly captures spontaneous differentiation and co-development of embryonic epiblast-like and extra-embryonic hypoblast-like lineages, establishes key signalling hubs with secreted modulators and undergoes symmetry breaking-like events. Single-cell transcriptomics confirms differentiation into diverse cell states of the perigastrulating human embryo14,15 without establishing placental cell types, including signatures of post-implantation epiblast, amniotic ectoderm, primitive streak, mesoderm, early extra-embryonic endoderm, as well as initial yolk sac induction. Collectively, our system captures key features of human embryonic development spanning from Carnegie stage16 4-7, offering a reproducible, tractable and scalable experimental platform to understand the basic cellular and molecular mechanisms that underlie human development, including new opportunities to dissect congenital pathologies with high throughput.


Assuntos
Linhagem da Célula , Implantação do Embrião , Desenvolvimento Embrionário , Células-Tronco Pluripotentes , Feminino , Humanos , Gravidez , Diferenciação Celular , Camadas Germinativas/citologia , Camadas Germinativas/enzimologia , Células-Tronco Embrionárias Humanas/citologia , Placenta/citologia , Células-Tronco Pluripotentes/citologia , Linha Primitiva/citologia , Linha Primitiva/embriologia , Saco Vitelino/citologia , Saco Vitelino/embriologia
7.
Immunity ; 50(6): 1453-1466.e4, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31053503

RESUMO

In lymph nodes, subcapsular sinus macrophages (SSMs) form an immunological barrier that monitors lymph drained from peripheral tissues. Upon infection, SSMs activate B and natural killer T (NKT) cells while secreting inflammatory mediators. Here, we investigated the mechanisms regulating development and homeostasis of SSMs. Embryonic SSMs originated from yolk sac hematopoiesis and were replaced by a postnatal wave of bone marrow (BM)-derived monocytes that proliferated to establish the adult SSM network. The SSM network self-maintained by proliferation with minimal BM contribution. Upon pathogen-induced transient deletion, BM-derived cells contributed to restoring the SSM network. Lymphatic endothelial cells (LECs) were the main source of CSF-1 within the lymph node and conditional deletion of Csf1 in adult LECs decreased the network of SSMs and medullary sinus macrophages (MSMs). Thus, SSMs have a dual hematopoietic origin, and LECs are essential to the niche supporting these macrophages.


Assuntos
Células Endoteliais/metabolismo , Macrófagos/metabolismo , Animais , Biomarcadores , Comunicação Celular , Diferenciação Celular , Expressão Gênica , Genes Reporter , Hematopoese/genética , Hematopoese/imunologia , Homeostase , Linfonodos/citologia , Linfonodos/imunologia , Vasos Linfáticos , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Monócitos/citologia , Monócitos/metabolismo , Saco Vitelino
8.
Nature ; 604(7907): 740-748, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35444273

RESUMO

All tissue-resident macrophages of the central nervous system (CNS)-including parenchymal microglia, as well as CNS-associated macrophages (CAMs1) such as meningeal and perivascular macrophages2-7-are part of the CNS endogenous innate immune system that acts as the first line of defence during infections or trauma2,8-10. It has been suggested that microglia and all subsets of CAMs are derived from prenatal cellular sources in the yolk sac that were defined as early erythromyeloid progenitors11-15. However, the precise ontogenetic relationships, the underlying transcriptional programs and the molecular signals that drive the development of distinct CAM subsets in situ are poorly understood. Here we show, using fate-mapping systems, single-cell profiling and cell-specific mutants, that only meningeal macrophages and microglia share a common prenatal progenitor. By contrast, perivascular macrophages originate from perinatal meningeal macrophages only after birth in an integrin-dependent manner. The establishment of perivascular macrophages critically requires the presence of arterial vascular smooth muscle cells. Together, our data reveal a precisely timed process in distinct anatomical niches for the establishment of macrophage subsets in the CNS.


Assuntos
Linhagem da Célula , Sistema Nervoso Central , Macrófagos , Sistema Nervoso Central/imunologia , Feminino , Humanos , Imunidade Inata , Macrófagos/citologia , Microglia , Gravidez , Saco Vitelino
9.
Immunity ; 48(6): 1160-1171.e5, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29858009

RESUMO

Hematopoiesis occurs in distinct waves. "Definitive" hematopoietic stem cells (HSCs) with the potential for all blood lineages emerge in the aorta-gonado-mesonephros, while "primitive" progenitors, whose potential is thought to be limited to erythrocytes, megakaryocytes, and macrophages, arise earlier in the yolk sac (YS). Here, we questioned whether other YS lineages exist that have not been identified, partially owing to limitations of current lineage tracing models. We established the use of Cdh5-CreERT2 for hematopoietic fate mapping, which revealed the YS origin of mast cells (MCs). YS-derived MCs were replaced by definitive MCs, which maintained themselves independently from the bone marrow in the adult. Replacement occurred with tissue-specific kinetics. MCs in the embryonic skin, but not other organs, remained largely YS derived prenatally and were phenotypically and transcriptomically distinct from definite adult MCs. We conclude that within myeloid lineages, dual hematopoietic origin is shared between macrophages and MCs.


Assuntos
Linhagem da Célula/imunologia , Hematopoese/fisiologia , Mastócitos/citologia , Animais , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/citologia , Macrófagos/citologia , Macrófagos/imunologia , Mastócitos/imunologia , Camundongos , Pele/citologia , Pele/imunologia , Saco Vitelino/citologia , Saco Vitelino/embriologia
10.
Cell ; 150(3): 590-605, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22863011

RESUMO

Endothelium in embryonic hematopoietic tissues generates hematopoietic stem/progenitor cells; however, it is unknown how its unique potential is specified. We show that transcription factor Scl/Tal1 is essential for both establishing the hematopoietic transcriptional program in hemogenic endothelium and preventing its misspecification to a cardiomyogenic fate. Scl(-/-) embryos activated a cardiac transcriptional program in yolk sac endothelium, leading to the emergence of CD31+Pdgfrα+ cardiogenic precursors that generated spontaneously beating cardiomyocytes. Ectopic cardiogenesis was also observed in Scl(-/-) hearts, where the disorganized endocardium precociously differentiated into cardiomyocytes. Induction of mosaic deletion of Scl in Scl(fl/fl)Rosa26Cre-ER(T2) embryos revealed a cell-intrinsic, temporal requirement for Scl to prevent cardiomyogenesis from endothelium. Scl(-/-) endothelium also upregulated the expression of Wnt antagonists, which promoted rapid cardiomyocyte differentiation of ectopic cardiogenic cells. These results reveal unexpected plasticity in embryonic endothelium such that loss of a single master regulator can induce ectopic cardiomyogenesis from endothelial cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Endotélio Vascular/embriologia , Coração/embriologia , Proteínas Proto-Oncogênicas/metabolismo , Animais , Caderinas/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hemangioblastos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Mesoderma/metabolismo , Camundongos , Miócitos Cardíacos/citologia , Placenta/irrigação sanguínea , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Gravidez , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Transcrição/metabolismo , Saco Vitelino/irrigação sanguínea
11.
Immunity ; 47(2): 217-218, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28813653

RESUMO

In this issue of Immunity, Zhu et al. (2017) report that tumor-associated macrophages in a mouse model of pancreatic ductal adenocarcinoma (PDAC) originate from both the yolk sac (YS) and bone marrow. Differential ablation of these populations indicates that only the YS-derived macrophages promote PDAC progression and growth.


Assuntos
Carcinoma Ductal Pancreático , Saco Vitelino , Animais , Medula Óssea , Macrófagos , Pâncreas
12.
Immunity ; 47(1): 10-12, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28723544

RESUMO

In this issue of Immunity, Takata et al. (2017) describe a novel method to differentiate macrophages from iPSCs. These cells, which they call iMacs, are similar to yolk-sac-derived macrophages and are capable of undergoing terminal differentiation into tissue-resident-like macrophages in vitro and in vivo.


Assuntos
Diferenciação Celular , Macrófagos/citologia , Feto/citologia , Humanos , Saco Vitelino/citologia
13.
Nature ; 577(7791): 537-542, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31830756

RESUMO

Our understanding of how human embryos develop before gastrulation, including spatial self-organization and cell type ontogeny, remains limited by available two-dimensional technological platforms1,2 that do not recapitulate the in vivo conditions3-5. Here we report a three-dimensional (3D) blastocyst-culture system that enables human blastocyst development up to the primitive streak anlage stage. These 3D embryos mimic developmental landmarks and 3D architectures in vivo, including the embryonic disc, amnion, basement membrane, primary and primate unique secondary yolk sac, formation of anterior-posterior polarity and primitive streak anlage. Using single-cell transcriptome profiling, we delineate ontology and regulatory networks that underlie the segregation of epiblast, primitive endoderm and trophoblast. Compared with epiblasts, the amniotic epithelium shows unique and characteristic phenotypes. After implantation, specific pathways and transcription factors trigger the differentiation of cytotrophoblasts, extravillous cytotrophoblasts and syncytiotrophoblasts. Epiblasts undergo a transition to pluripotency upon implantation, and the transcriptome of these cells is maintained until the generation of the primitive streak anlage. These developmental processes are driven by different pluripotency factors. Together, findings from our 3D-culture approach help to determine the molecular and morphogenetic developmental landscape that occurs during human embryogenesis.


Assuntos
Técnicas de Cultura de Células/métodos , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário , Linha Primitiva/citologia , Linha Primitiva/embriologia , Âmnio/citologia , Âmnio/embriologia , Blastocisto/citologia , Diferenciação Celular , Linhagem da Célula , Polaridade Celular , Colágeno , Combinação de Medicamentos , Epitélio/embriologia , Gastrulação , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Humanos , Laminina , Proteoglicanas , RNA-Seq , Análise de Célula Única , Fatores de Transcrição/metabolismo , Transcriptoma , Trofoblastos/citologia , Saco Vitelino/citologia , Saco Vitelino/embriologia
14.
Nature ; 587(7834): 443-447, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32968278

RESUMO

Current understandings of cell specification in early mammalian pre-implantation development are based mainly on mouse studies. The first lineage differentiation event occurs at the morula stage, with outer cells initiating a trophectoderm (TE) placental progenitor program. The inner cell mass arises from inner cells during subsequent developmental stages and comprises precursor cells of the embryo proper and yolk sac1. Recent gene-expression analyses suggest that the mechanisms that regulate early lineage specification in the mouse may differ in other mammals, including human2-5 and cow6. Here we show the evolutionary conservation of a molecular cascade that initiates TE segregation in human, cow and mouse embryos. At the morula stage, outer cells acquire an apical-basal cell polarity, with expression of atypical protein kinase C (aPKC) at the contact-free domain, nuclear expression of Hippo signalling pathway effectors and restricted expression of TE-associated factors such as GATA3, which suggests initiation of a TE program. Furthermore, we demonstrate that inhibition of aPKC by small-molecule pharmacological modulation or Trim-Away protein depletion impairs TE initiation at the morula stage. Our comparative embryology analysis provides insights into early lineage specification and suggests that a similar mechanism initiates a TE program in human, cow and mouse embryos.


Assuntos
Evolução Biológica , Ectoderma/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Transcrição Gênica , Trofoblastos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Massa Celular Interna do Blastocisto/citologia , Massa Celular Interna do Blastocisto/metabolismo , Bovinos , Linhagem da Célula , Polaridade Celular , Ectoderma/citologia , Embrião de Mamíferos/enzimologia , Feminino , Fator de Transcrição GATA3/metabolismo , Via de Sinalização Hippo , Humanos , Camundongos , Mórula/citologia , Mórula/enzimologia , Mórula/metabolismo , Placenta/citologia , Placenta/metabolismo , Gravidez , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Trofoblastos/citologia , Proteínas de Sinalização YAP , Saco Vitelino/citologia , Saco Vitelino/metabolismo
15.
Nature ; 582(7813): 571-576, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32499656

RESUMO

Macrophages are the first cells of the nascent immune system to emerge during embryonic development. In mice, embryonic macrophages infiltrate developing organs, where they differentiate symbiotically into tissue-resident macrophages (TRMs)1. However, our understanding of the origins and specialization of macrophages in human embryos is limited. Here we isolated CD45+ haematopoietic cells from human embryos at Carnegie stages 11 to 23 and subjected them to transcriptomic profiling by single-cell RNA sequencing, followed by functional characterization of a population of CD45+CD34+CD44+ yolk sac-derived myeloid-biased progenitors (YSMPs) by single-cell culture. We also mapped macrophage heterogeneity across multiple anatomical sites and identified diverse subsets, including various types of embryonic TRM (in the head, liver, lung and skin). We further traced the specification trajectories of TRMs from either yolk sac-derived primitive macrophages or YSMP-derived embryonic liver monocytes using both transcriptomic and developmental staging information, with a focus on microglia. Finally, we evaluated the molecular similarities between embryonic TRMs and their adult counterparts. Our data represent a comprehensive characterization of the spatiotemporal dynamics of early macrophage development during human embryogenesis, providing a reference for future studies of the development and function of human TRMs.


Assuntos
Macrófagos/citologia , Análise de Célula Única , Linhagem da Célula , Embrião de Mamíferos/citologia , Cabeça , Hematopoese , Humanos , Antígenos Comuns de Leucócito/metabolismo , Fígado/citologia , Fígado/embriologia , Pulmão/citologia , Macrófagos/metabolismo , Microglia/citologia , Células Progenitoras Mieloides/citologia , RNA-Seq , Pele/citologia , Análise Espaço-Temporal , Transcriptoma , Saco Vitelino/citologia
16.
Development ; 149(18)2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36162815

RESUMO

Viral-derived genes have had a huge impact during mammalian evolution, with many of the exapted genes being expressed in the placenta. Now, new research published in Development describes the importance of two genes with retroviral origins in microglia, the innate immune cells of the brain, which are derived from another extra-embryonic tissue, the yolk sac. We caught up with the first author, Masahito Irie, and the corresponding authors, Fumitoshi Ishino, Professor at Tokyo Medical and Dental University, and Tomoko Kaneko-Ishino, Professor at Tokai University, to hear about more about their research.


Assuntos
Placenta , Saco Vitelino , Animais , Feminino , Humanos , Mamíferos , Microglia , Gravidez
17.
Development ; 149(8)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35452096

RESUMO

Previously, we have demonstrated that a subpopulation of microglia, known as Hoxb8 microglia, is derived from the Hoxb8 lineage during the second wave (E8.5) of yolk sac hematopoiesis, whereas canonical non-Hoxb8 microglia arise from the first wave (E7.5). Hoxb8 microglia have an ontogeny distinct from non-Hoxb8 microglia. Dysfunctional Hoxb8 microglia cause the acquisition of chronic anxiety and an obsessive-compulsive spectrum-like behavior, trichotillomania, in mice. The nature and fate of the progenitors generated during E8.5 yolk sac hematopoiesis have been controversial. Herein, we use the Hoxb8 cell lineage reporter to define the ontogeny of hematopoietic cells arising during the definitive waves of hematopoiesis initiated in the E8.5 yolk sac and aorta-gonad-mesonephros (AGM) region. Our murine cell lineage analysis shows that the Hoxb8 cell lineage reporter robustly marks erythromyeloid progenitors, hematopoietic stem cells and their progeny, particularly monocytes. Hoxb8 progenitors and microglia require Myb function, a hallmark transcription factor for definitive hematopoiesis, for propagation and maturation. During adulthood, all immune lineages and, interestingly, resident macrophages in only hematopoietic/lymphoid tissues are derived from Hoxb8 precursors. These results illustrate that the Hoxb8 lineage exclusively mirrors murine definitive hematopoiesis.


Assuntos
Hematopoese , Saco Vitelino , Animais , Linhagem da Célula , Células-Tronco Hematopoéticas , Proteínas de Homeodomínio/genética , Mesonefro , Camundongos
18.
Development ; 149(8)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35438172

RESUMO

Hofbauer cells (HBCs) are tissue macrophages of the placenta thought to be important for fetoplacental vascular development and innate immune protection. The developmental origins of HBCs remain unresolved and could implicate functional diversity of HBCs in placenta development and disease. In this study, we used flow cytometry and paternally inherited reporters to phenotype placenta macrophages and to identify fetal-derived HBCs and placenta-associated maternal macrophages in the mouse. In vivo pulse-labeling traced the ontogeny of HBCs from yolk sac-derived erythro-myeloid progenitors, with a minor contribution from fetal hematopoietic stem cells later on. Single-cell RNA-sequencing revealed transcriptional similarities between placenta macrophages and erythro-myeloid progenitor-derived fetal liver macrophages and microglia. As with other fetal tissue macrophages, HBCs were dependent on the transcription factor Pu.1, the loss-of-function of which in embryos disrupted fetoplacental labyrinth morphology, supporting a role for HBC in labyrinth angiogenesis and/or remodeling. HBC were also sensitive to Pu.1 (Spi1) haploinsufficiency, which caused an initial deficiency in the numbers of macrophages in the early mouse placenta. These results provide groundwork for future investigation into the relationship between HBC ontogeny and function in placenta pathophysiology.


Assuntos
Macrófagos , Placenta , Animais , Feminino , Células-Tronco Hematopoéticas , Camundongos , Células Progenitoras Mieloides , Gravidez , Saco Vitelino
19.
Development ; 149(7)2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35297995

RESUMO

Establishing a functional circulatory system is required for post-implantation development during murine embryogenesis. Previous studies in loss-of-function mouse models showed that FOXO1, a Forkhead family transcription factor, is required for yolk sac (YS) vascular remodeling and survival beyond embryonic day (E) 11. Here, we demonstrate that at E8.25, loss of Foxo1 in Tie2-cre expressing cells resulted in increased sprouty 2 (Spry2) and Spry4 expression, reduced arterial gene expression and reduced Kdr (also known as Vegfr2 and Flk1) transcripts without affecting overall endothelial cell identity, survival or proliferation. Using a Dll4-BAC-nlacZ reporter line, we found that one of the earliest expressed arterial genes, delta like 4, is significantly reduced in Foxo1 mutant YS without being substantially affected in the embryo proper. We show that FOXO1 binds directly to previously identified Spry2 gene regulatory elements (GREs) and newly identified, evolutionarily conserved Spry4 GREs to repress their expression. Furthermore, overexpression of Spry4 in transient transgenic embryos largely recapitulates the reduced expression of arterial genes seen in conditional Foxo1 mutants. Together, these data reveal a novel role for FOXO1 as a key transcriptional repressor regulating both pre-flow arterial specification and subsequent vessel remodeling within the murine YS.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Remodelação Vascular , Saco Vitelino , Animais , Artérias , Embrião de Mamíferos/metabolismo , Células Endoteliais/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Camundongos , Remodelação Vascular/genética , Saco Vitelino/metabolismo
20.
Immunity ; 44(4): 755-68, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-26992565

RESUMO

Tissue-resident macrophages can derive from yolk sac macrophages (YS-Macs), fetal liver monocytes (FL-MOs), or adult bone-marrow monocytes (BM-MOs). The relative capacity of these precursors to colonize a niche, self-maintain, and perform tissue-specific functions is unknown. We simultaneously transferred traceable YS-Macs, FL-MOs, and BM-MOs into the empty alveolar macrophage (AM) niche of neonatal Csf2rb(-/-) mice. All subsets produced AMs, but in competition preferential outgrowth of FL-MOs was observed, correlating with their superior granulocyte macrophage-colony stimulating factor (GM-CSF) reactivity and proliferation capacity. When transferred separately, however, all precursors efficiently colonized the alveolar niche and generated AMs that were transcriptionally almost identical, self-maintained, and durably prevented alveolar proteinosis. Mature liver, peritoneal, or colon macrophages could not efficiently colonize the empty AM niche, whereas mature AMs could. Thus, precursor origin does not affect the development of functional self-maintaining tissue-resident macrophages and the plasticity of the mononuclear phagocyte system is largest at the precursor stage.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fígado/citologia , Macrófagos Alveolares/citologia , Saco Vitelino/citologia , Animais , Proliferação de Células , Subunidade beta Comum dos Receptores de Citocinas/genética , Fígado/embriologia , Fígado/imunologia , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transcriptoma/imunologia , Saco Vitelino/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA