Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Res ; 33(6): 448-463, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37016019

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) are considered as a heterogeneous population, but precisely when, where and how HSPC heterogeneity arises remain largely unclear. Here, using a combination of single-cell multi-omics, lineage tracing and functional assays, we show that embryonic HSPCs originate from heterogeneous hemogenic endothelial cells (HECs) during zebrafish embryogenesis. Integrated single-cell transcriptome and chromatin accessibility analysis demonstrates transcriptional heterogeneity and regulatory programs that prime lymphoid/myeloid fates at the HEC level. Importantly, spi2+ HECs give rise to lymphoid/myeloid-primed HSPCs (L/M-HSPCs) and display a stress-responsive function under acute inflammation. Moreover, we uncover that Spi2 is required for the formation of L/M-HSPCs through tightly controlling the endothelial-to-hematopoietic transition program. Finally, single-cell transcriptional comparison of zebrafish and human HECs and human induced pluripotent stem cell-based hematopoietic differentiation results support the evolutionary conservation of L/M-HECs and a conserved role of SPI1 (spi2 homolog in mammals) in humans. These results unveil the lineage origin, biological function and molecular determinant of HSPC heterogeneity and lay the foundation for new strategies for induction of transplantable lineage-primed HSPCs in vitro.


Assuntos
Hemangioblastos , Células-Tronco Pluripotentes Induzidas , Animais , Humanos , Peixe-Zebra , Hematopoese/fisiologia , Células-Tronco Hematopoéticas , Diferenciação Celular , Linhagem da Célula , Mamíferos
2.
Proc Natl Acad Sci U S A ; 120(7): e2212212120, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36745802

RESUMO

During vertebrate embryogenesis, hematopoietic stem and progenitor cell (HSPC) production through endothelial-to-hematopoietic transition requires suitable developmental signals, but how these signals are accurately regulated remains incompletely understood. Cytoplasmic polyadenylation, which is one of the posttranscriptional regulations, plays a crucial role in RNA metabolism. Here, we report that Cpeb1b-mediated cytoplasmic polyadenylation is important for HSPC specification by translational control of Hedgehog (Hh) signaling during zebrafish early development. Cpeb1b is highly expressed in notochord and its deficiency results in defective HSPC production. Mechanistically, Cpeb1b regulates hemogenic endothelium specification by the Hedgehog-Vegf-Notch axis. We demonstrate that the cytoplasmic polyadenylation element motif-dependent interaction between Cpeb1b and shha messenger RNA (mRNA) in the liquid-like condensates, which are induced by Pabpc1b phase separation, is required for cytoplasmic polyadenylation of shha mRNA. Intriguingly, the cytoplasmic polyadenylation regulates translation but not stability of shha mRNA, which further enhances the Shha protein level and Hh signal transduction. Taken together, our findings uncover the role of Cpeb1b-mediated cytoplasmic polyadenylation in HSPC development and provide insights into how posttranscriptional regulation can direct developmental signals with high fidelity to translate them into cell fate transition.


Assuntos
Poliadenilação , Peixe-Zebra , Animais , Peixe-Zebra/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Hedgehog/metabolismo , Hematopoese/genética
3.
Cell Res ; 32(1): 38-53, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34341490

RESUMO

Limited knowledge of cellular and molecular mechanisms underlying hematopoietic stem cell and multipotent progenitor (HSC/MPP) expansion within their native niche has impeded the application of stem cell-based therapies for hematological malignancies. Here, we constructed a spatiotemporal transcriptome map of mouse fetal liver (FL) as a platform for hypothesis generation and subsequent experimental validation of novel regulatory mechanisms. Single-cell transcriptomics revealed three transcriptionally heterogeneous HSC/MPP subsets, among which a CD93-enriched subset exhibited enhanced stem cell properties. Moreover, by employing integrative analysis of single-cell and spatial transcriptomics, we identified novel HSC/MPP 'pocket-like' units (HSC PLUS), composed of niche cells (hepatoblasts, stromal cells, endothelial cells, and macrophages) and enriched with growth factors. Unexpectedly, macrophages showed an 11-fold enrichment in the HSC PLUS. Functionally, macrophage-HSC/MPP co-culture assay and candidate molecule testing, respectively, validated the supportive role of macrophages and growth factors (MDK, PTN, and IGFBP5) in HSC/MPP expansion. Finally, cross-species analysis and functional validation showed conserved cell-cell interactions and expansion mechanisms but divergent transcriptome signatures between mouse and human FL HSCs/MPPs. Taken together, these results provide an essential resource for understanding HSC/MPP development in FL, and novel insight into functional HSC/MPP expansion ex vivo.


Assuntos
Células Endoteliais , Transcriptoma , Animais , Hematopoese/genética , Células-Tronco Hematopoéticas , Fígado , Camundongos
4.
Dev Cell ; 56(14): 2121-2133.e6, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34197725

RESUMO

Macrophages play pivotal roles in immunity, hematopoiesis, and tissue homeostasis. In mammals, macrophages have been shown to originate from yolk-sac-derived erythro-myeloid progenitors and aorta-gonad-mesonephros (AGM)-derived hematopoietic stem cells. However, whether macrophages can arise from other embryonic sites remains unclear. Here, using single-cell RNA sequencing, we profile the transcriptional landscape of mouse fetal placental hematopoiesis. We uncover and experimentally validate that a CD44+ subpopulation of placental endothelial cells (ECs) exhibits hemogenic potential. Importantly, lineage tracing using the newly generated Hoxa13 reporter line shows that Hoxa13-labeled ECs can produce placental macrophages, named Hofbauer cell (HBC)-like cells. Furthermore, we identify two subtypes of HBC-like cells, and cell-cell interaction analysis identifies their potential roles in angiogenesis and antigen presentation, separately. Our study provides a comprehensive understanding of placental hematopoiesis and highlights the placenta as a source of macrophages, which has important implications for both basic and translational research.


Assuntos
Linhagem da Célula , Hemangioblastos/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Macrófagos/citologia , Placenta/citologia , Animais , Feminino , Hemangioblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Placenta/metabolismo , Gravidez , Análise de Célula Única , Transcriptoma
5.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33785593

RESUMO

During vertebrate embryogenesis, fetal hematopoietic stem and progenitor cells (HSPCs) exhibit expansion and differentiation properties in a supportive hematopoietic niche. To profile the developmental landscape of fetal HSPCs and their local niche, here, using single-cell RNA-sequencing, we deciphered a dynamic atlas covering 28,777 cells and 9 major cell types (23 clusters) of zebrafish caudal hematopoietic tissue (CHT). We characterized four heterogeneous HSPCs with distinct lineage priming and metabolic gene signatures. Furthermore, we investigated the regulatory mechanism of CHT niche components for HSPC development, with a focus on the transcription factors and ligand-receptor networks involved in HSPC expansion. Importantly, we identified an endothelial cell-specific G protein-coupled receptor 182, followed by in vivo and in vitro functional validation of its evolutionally conserved role in supporting HSPC expansion in zebrafish and mice. Finally, comparison between zebrafish CHT and human fetal liver highlighted the conservation and divergence across evolution. These findings enhance our understanding of the regulatory mechanism underlying hematopoietic niche for HSPC expansion in vivo and provide insights into improving protocols for HSPC expansion in vitro.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas/fisiologia , Nicho de Células-Tronco , Animais , Linhagem da Célula , Feto/metabolismo , Perfilação da Expressão Gênica , Humanos , Fígado/metabolismo , Camundongos , Análise de Célula Única , Peixe-Zebra
6.
Blood ; 137(2): 190-202, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-32756943

RESUMO

Nascent hematopoietic stem and progenitor cells (HSPCs) acquire definitive hematopoietic characteristics only when they develop into fetal HSPCs; however, the mechanisms underlying fetal HSPC development are poorly understood. Here, we profiled the chromatin accessibility and transcriptional features of zebrafish nascent and fetal HSPCs using ATAC-seq and RNA-seq and revealed dynamic changes during HSPC transition. Functional assays demonstrated that chromatin remodeler-mediated epigenetic programming facilitates fetal HSPC development in vertebrates. Systematical screening of chromatin remodeler-related genes identified that smarca5 is responsible for the maintenance of chromatin accessibility at promoters of hematopoiesis-related genes in fetal HSPCs. Mechanistically, Smarca5 interacts with nucleolin to promote chromatin remodeling, thereby facilitating genomic binding of transcription factors to regulate expression of hematopoietic regulators such as bcl11ab. Our results unravel a new role of epigenetic regulation and reveal that Smarca5-mediated epigenetic programming is responsible for fetal HSPC development, which will provide new insights into the generation of functional HSPCs both in vivo and in vitro.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Epigênese Genética/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Proteínas de Peixe-Zebra/metabolismo , Adenosina Trifosfatases/genética , Animais , Proteínas Cromossômicas não Histona/genética , Camundongos , Camundongos Endogâmicos C57BL , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
7.
Stem Cell Reports ; 15(3): 749-760, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32822590

RESUMO

Cell transplantation into immunodeficient recipients is a widely used approach to study stem cell and cancer biology; however, studying cell states post transplantation in vivo is inconvenient in mammals. Here, we generated a foxn1/Casper mutant zebrafish that is transparent and exhibits T cell deficiency. By employing the line for hematopoietic stem cell (HSC) transplantation (HSCT), we could achieve nonconditioned transplantation. Meanwhile, we found that fetal HSCs from 3 days post fertilization zebrafish embryos produce a better transplant outcome in foxn1/Casper mutants, compared with adult HSCs. In addition to HSCT, the foxn1/Casper mutant is feasible for allografts of myelodysplastic syndrome-like and muscle cells, as well as xenografts of medaka muscle cells. In summary, foxn1/Casper mutants permit the nonconditioned engraftment of multiple cell types and visualized characterization of transplanted cells in vivo.


Assuntos
Aloenxertos/transplante , Fatores de Transcrição Forkhead/genética , Xenoenxertos/transplante , Mutação/genética , Neoplasias/patologia , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Sequência de Bases , Células-Tronco Fetais/citologia , Fatores de Transcrição Forkhead/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Resultado do Tratamento , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/metabolismo
8.
PLoS Biol ; 18(4): e3000696, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32275659

RESUMO

It is well known that various developmental signals play diverse roles in hematopoietic stem and progenitor cell (HSPC) production; however, how these signaling pathways are orchestrated remains incompletely understood. Here, we report that Rab5c is essential for HSPC specification by endocytic trafficking of Notch and AKT signaling in zebrafish embryos. Rab5c deficiency leads to defects in HSPC production. Mechanistically, Rab5c regulates hemogenic endothelium (HE) specification by endocytic trafficking of Notch ligands and receptor. We further show that the interaction between Rab5c and Appl1 in the endosome is required for the survival of HE in the ventral wall of the dorsal aorta through AKT signaling. Interestingly, Rab5c overactivation can also lead to defects in HSPC production, which is attributed to excessive endolysosomal trafficking inducing Notch signaling defect. Taken together, our findings establish a previously unrecognized role of Rab5c-mediated endocytic trafficking in HSPC development and provide new insights into how spatiotemporal signals are orchestrated to accurately execute cell fate transition.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Notch/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Animais , Animais Geneticamente Modificados , Embrião não Mamífero , Endocitose , Endotélio/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Receptores Notch/genética , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Proteínas rab5 de Ligação ao GTP/química , Proteínas rab5 de Ligação ao GTP/genética
9.
Nat Commun ; 10(1): 1839, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31015398

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) are capable of producing all mature blood lineages, as well as maintaining the self-renewal ability throughout life. The hairy-like organelle, cilium, is present in most types of vertebrate cells, and plays important roles in various biological processes. However, it is unclear whether and how cilia regulate HSPC development in vertebrates. Here, we show that cilia-specific genes, involved in primary cilia formation and function, are required for HSPC development, especially in hemogenic endothelium (HE) specification in zebrafish embryos. Blocking primary cilia formation or function by genetic or chemical manipulations impairs HSPC development. Mechanistically, we uncover that primary cilia in endothelial cells transduce Notch signal to the earliest HE for proper HSPC specification during embryogenesis. Altogether, our findings reveal a pivotal role of endothelial primary cilia in HSPC development, and may shed lights into in vitro directed differentiation of HSPCs.


Assuntos
Cílios/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Cílios/genética , Embrião não Mamífero , Desenvolvimento Embrionário/fisiologia , Hemangioblastos/citologia , Hemangioblastos/metabolismo , Hematopoese/fisiologia , Modelos Animais , Peixe-Zebra/fisiologia
10.
Nature ; 549(7671): 273-276, 2017 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-28869969

RESUMO

N6-methyladenosine (m6A) has been identified as the most abundant modification on eukaryote messenger RNA (mRNA). Although the rapid development of high-throughput sequencing technologies has enabled insight into the biological functions of m6A modification, the function of m6A during vertebrate embryogenesis remains poorly understood. Here we show that m6A determines cell fate during the endothelial-to-haematopoietic transition (EHT) to specify the earliest haematopoietic stem/progenitor cells (HSPCs) during zebrafish embryogenesis. m6A-specific methylated RNA immunoprecipitation combined with high-throughput sequencing (MeRIP-seq) and m6A individual-nucleotide-resolution cross-linking and immunoprecipitation with sequencing (miCLIP-seq) analyses reveal conserved features on zebrafish m6A methylome and preferential distribution of m6A peaks near the stop codon with a consensus RRACH motif. In mettl3-deficient embryos, levels of m6A are significantly decreased and emergence of HSPCs is blocked. Mechanistically, we identify that the delayed YTHDF2-mediated mRNA decay of the arterial endothelial genes notch1a and rhoca contributes to this deleterious effect. The continuous activation of Notch signalling in arterial endothelial cells of mettl3-deficient embryos blocks EHT, thereby repressing the generation of the earliest HSPCs. Furthermore, knockdown of Mettl3 in mice confers a similar phenotype. Collectively, our findings demonstrate the critical function of m6A modification in the fate determination of HSPCs during vertebrate embryogenesis.


Assuntos
Adenosina/análogos & derivados , Diferenciação Celular , Células Endoteliais/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , RNA Mensageiro/metabolismo , Peixe-Zebra/embriologia , Adenosina/metabolismo , Animais , Diferenciação Celular/genética , Códon de Terminação/genética , Sequência Consenso , Células Endoteliais/metabolismo , Técnicas de Silenciamento de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Proteínas de Homeodomínio/genética , Imunoprecipitação , Metilação , Metiltransferases/deficiência , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Estabilidade de RNA , RNA Mensageiro/química , RNA Mensageiro/genética , Receptor Notch1/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
11.
EMBO J ; 36(20): 2987-2997, 2017 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-28882847

RESUMO

DNA methylation is a major epigenetic modification; however, the precise role of DNA methylation in vertebrate development is still not fully understood. Here, we show that DNA methylation is essential for the establishment of the left-right (LR) asymmetric body plan during vertebrate embryogenesis. Perturbation of DNA methylation by depletion of DNA methyltransferase 1 (dnmt1) or dnmt3bb.1 in zebrafish embryos leads to defects in dorsal forerunner cell (DFC) specification or collective migration, laterality organ malformation, and disruption of LR patterning. Knockdown of dnmt1 in Xenopus embryos also causes similar defects. Mechanistically, loss of dnmt1 function induces hypomethylation of the lefty2 gene enhancer and promotes lefty2 expression, which consequently represses Nodal signaling in zebrafish embryos. We also show that Dnmt3bb.1 regulates collective DFC migration through cadherin 1 (Cdh1). Taken together, our data uncover dynamic DNA methylation as an epigenetic mechanism to control LR determination during early embryogenesis in vertebrates.


Assuntos
Metilação de DNA , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Xenopus/embriologia , Peixe-Zebra/embriologia , Animais , Metiltransferases/metabolismo
12.
Elife ; 62017 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-28945193

RESUMO

SUN (Sad1 and UNC84 domain containing)-domain proteins are reported to reside on the nuclear membrane playing distinct roles in nuclear dynamics. SUN5 is a new member of the SUN family, with little knowledge regarding its function. Here, we generated Sun5-/- mice and found that male mice were infertile. Most Sun5-null spermatozoa displayed a globozoospermia-like phenotype but they were actually acephalic spermatozoa. Additional studies revealed that SUN5 was located in the neck of the spermatozoa, anchoring sperm head to the tail, and without functional SUN5 the sperm head to tail coupling apparatus was detached from nucleus during spermatid elongation. Finally, we found that healthy heterozygous offspring could be obtained via intracytoplasmic injection of Sun5-mutated sperm heads for both male mice and patients. Our studies reveal the essential role of SUN5 in anchoring sperm head to the tail and provide a promising way to treat this kind of acephalic spermatozoa-associated male infertility.


Assuntos
Proteínas de Membrana/metabolismo , Cabeça do Espermatozoide/fisiologia , Cauda do Espermatozoide/fisiologia , Espermatogênese , Animais , Masculino , Proteínas de Membrana/deficiência , Camundongos Knockout , Membrana Nuclear/metabolismo
13.
Dev Cell ; 42(4): 349-362.e4, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28803829

RESUMO

In mammals, hematopoietic stem and progenitor cells (HSPCs) rapidly expand in the fetal liver (FL), but the underlying mechanism remains unclear. Here, we characterize zebrafish caudal hematopoietic tissue (CHT) and identify an important cellular and molecular mechanism of HSPC expansion. Time-lapse imaging showed that HSPCs localize adjacent to vascular endothelial cells (ECs), and their migration and expansion display caudal vein-specific orientation in the CHT. RNA sequencing and functional analysis identified that an EC-expressed transcription factor, Krüppel-like factor 6a (Klf6a), is essential for the CHT niche. We further demonstrated that Klf6a directly regulates the expression of the chemokine (C-C motif) ligand 25b to modulate HSPC lodgment and proliferation. Ex vivo culture results support the conserved role of Ccl21/Ccr7 signaling in promoting HSPC expansion in mammals. Together, we identify the Klf6a-Ccl25b/Ccr7 axis in controlling the complex HSPC-CHT niche interaction, which may be applicable to in vitro expansion or engraftment of HSPCs after transplantation.


Assuntos
Quimiocinas CC/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nicho de Células-Tronco , Proteínas de Peixe-Zebra/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Quimiocinas CC/genética , Células Endoteliais/citologia , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Receptores CCR7/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
15.
Sci Rep ; 5: 15605, 2015 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-26499367

RESUMO

The photosynthetic rate of virus-infected plants is always reduced. However, the molecular mechanism underlying this phenomenon remains unclear. The helper component-proteinase (HC-Pro) of Potato virus Y (PVY) was found in the chloroplasts of PVY-infected tobacco, indicating some new function of HC-Pro in the chloroplasts. We generated HC-Pro transgenic plants with a transit peptide to target the protein to chloroplast. The HC-Pro transgenic tobacco showed a decreased photosynthetic rate by 25% at the light intensity of 600 µmol m(-2) s(-1). Using a yeast two-hybrid screening assay to search for chloroplast proteins interacting with HC-Pro, we identified that PVY HC-Pro can interact with the chloroplast ATP synthase NtCF1ß-subunit. This interaction was confirmed by GST pull-down and co-immunoprecipitation assays. HC-Pro didn't interfere with the activity of assembled ATP synthase in vitro. The HC-Pro/NtCF1ß-subunit interaction might affect the assembly of ATP synthase complex. Quantitative western blot and immunogold labeling of the ATP synthase indicated that the amount of ATP synthase complex was decreased in both the HC-Pro transgenic and the PVY-infected tobacco. These results demonstrate that HC-Pro plays an important role in reducing the photosynthetic rate of PVY-infected plants, which is a completely new role of HC-Pro besides its multiple known functions.


Assuntos
Arabidopsis/virologia , ATPases de Cloroplastos Translocadoras de Prótons/genética , Cloroplastos/virologia , Cisteína Endopeptidases/genética , Nicotiana/virologia , Fotossíntese/fisiologia , Proteínas Virais/genética , Arabidopsis/genética , Arabidopsis/metabolismo , Western Blotting , ATPases de Cloroplastos Translocadoras de Prótons/metabolismo , Cisteína Endopeptidases/metabolismo , Imuno-Histoquímica , Doenças das Plantas/virologia , Plantas Geneticamente Modificadas/virologia , Potyvirus/patogenicidade , Saccharomyces cerevisiae/genética , Nicotiana/genética , Nicotiana/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Proteínas Virais/metabolismo
16.
Cell Res ; 25(10): 1093-107, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26358189

RESUMO

In vertebrates, embryonic hematopoietic stem and progenitor cells (HSPCs) are derived from a subset of endothelial cells, the hemogenic endothelium (HE), through the endothelial-to-hematopoietic transition (EHT). Notch signaling is essential for HSPC development during embryogenesis across vertebrates. However, whether and how it regulates EHT remains unclear. Here, we show that G protein-coupled receptor 183 (Gpr183) signaling serves as an indispensable switch for HSPC emergence by repressing Notch signaling before the onset of EHT. Inhibition of Gpr183 significantly upregulates Notch signaling and abolishes HSPC emergence. Upon activation by its ligand 7α-25-OHC, Gpr183 recruits ß-arrestin1 and the E3 ligase Nedd4 to degrade Notch1 in specified HE cells and then facilitates the subsequent EHT. Importantly, 7α-25-OHC stimulation promotes HSPC emergence in vivo and in vitro, providing an attractive strategy for enhancing the in vitro generation of functional HSPCs.


Assuntos
Hemangioblastos/citologia , Células-Tronco Hematopoéticas/citologia , Receptor Notch1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/fisiologia , Animais , Arrestinas/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Hemangioblastos/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Ubiquitina-Proteína Ligases Nedd4 , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , beta-Arrestinas
17.
Dev Cell ; 34(6): 621-31, 2015 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-26300447

RESUMO

T lymphoid-primed progenitors are hematopoietic progenitors destined to enter the thymus. The in vivo characterization of these embryonic progenitors is challenging, however, due to the intrauterine development of mouse embryos. Thus, how the fate of these cells is determined has not been fully defined in mammals. Here we use zebrafish embryos to show that the homing of T lymphoid-primed progenitors to the thymus is impaired, concomitant with a decrease in ccr9a expression, in the absence of irf4a. Strikingly, fate mapping assays at the single-cell level showed a fate change of irf4a-deficient T lymphoid-primed progenitors to myeloid cells, accompanied by an increase in Pu.1 expression. These data indicate that in addition to regulating ccr9a expression, Irf4a is essential in T lymphoid-primed progenitors for repressing Pu.1 expression to prevent an alternate fate. Our findings provide insight into the fate determination mechanism of T lymphoid-primed progenitors.


Assuntos
Linhagem da Célula , Embrião não Mamífero/citologia , Embrião não Mamífero/fisiologia , Epitopos de Linfócito T/fisiologia , Fatores Reguladores de Interferon/metabolismo , Células Mieloides/citologia , Células-Tronco/citologia , Peixe-Zebra/crescimento & desenvolvimento , Animais , Western Blotting , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas Imunoenzimáticas , Fatores Reguladores de Interferon/genética , Camundongos , Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/metabolismo , Transativadores/genética , Transativadores/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
18.
Plant Cell Environ ; 38(10): 2023-34, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25736930

RESUMO

Virus-infected plants show strong morphological and physiological alterations. Many physiological processes in chloroplast are affected, including the plastidic isoprenoid biosynthetic pathway [the 2C-methyl-D-erythritol-4-phosphate (MEP) pathway]; indeed, isoprenoid contents have been demonstrated to be altered in virus-infected plants. In this study, we found that the levels of photosynthetic pigments and abscisic acid (ABA) were altered in Potato virus Y (PVY)-infected tobacco. Using yeast two-hybrid assays, we demonstrated an interaction between virus protein PVY helper component-proteinase (HC-Pro) and tobacco chloroplast protein 1-deoxy-D-xylulose-5-phosphate synthase (NtDXS). This interaction was confirmed using bimolecular fluorescence complementation (BiFC) assays and pull-down assays. The Transket_pyr domain (residues 394-561) of NtDXS was required for interaction with HC-Pro, while the N-terminal region of HC-Pro (residues 1-97) was necessary for interaction with NtDXS. Using in vitro enzyme activity assays, PVY HC-Pro was found to promote the synthase activity of NtDXS. We observed increases in photosynthetic pigment contents and ABA levels in transgenic plants with HC-Pro accumulating in the chloroplasts. During virus infection, the enhancement of plastidic isoprenoid biosynthesis was attributed to the enhancement of DXS activity by HC-Pro. Our study reveals a new role of HC-Pro in the host plant metabolic system and will contribute to the study of host-virus relationships.


Assuntos
Cisteína Endopeptidases/metabolismo , Nicotiana/enzimologia , Doenças das Plantas/virologia , Potyvirus/fisiologia , Terpenos/metabolismo , Transferases/metabolismo , Proteínas Virais/metabolismo , Ácido Abscísico/metabolismo , Sequência de Bases , Proteínas de Cloroplastos/genética , Proteínas de Cloroplastos/metabolismo , Cloroplastos/enzimologia , Biblioteca Gênica , Dados de Sequência Molecular , Fotossíntese , Reguladores de Crescimento de Plantas/metabolismo , Folhas de Planta/enzimologia , Folhas de Planta/genética , Folhas de Planta/metabolismo , Folhas de Planta/virologia , Plantas Geneticamente Modificadas , Plastídeos/metabolismo , Análise de Sequência de DNA , Deleção de Sequência , Nicotiana/genética , Nicotiana/metabolismo , Nicotiana/virologia , Transferases/genética , Técnicas do Sistema de Duplo-Híbrido
19.
Blood ; 125(7): 1098-106, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25540193

RESUMO

Inflammatory signaling has been shown to be essential for stress hematopoiesis in adult bone marrow, either through increasing proliferation or by directing differentiation of hematopoietic stem and progenitor cells (HSPCs) toward myeloid or lymphoid lineages. However, its role in embryonic normal hematopoiesis has been unknown. Here, we demonstrate that in both zebrafish and mouse embryos, inflammatory signaling is necessary and sufficient for HSPC emergence, in the absence of infection or pathological inflammation. Mechanistically, inflammatory signaling regulates hemogenic endothelium-derived HSPC development through a conserved Toll-like receptor 4 (TLR4)-nuclear factor κ-light-chain enhancer of activated B core (NF-κB) signaling, which then promotes Notch activity, a well-known signal required for HSPC specification in vertebrates. Our findings establish a previously unrecognized link between inflammatory signaling and HSPC emergence, and provide new insights into regenerative medicine and novel therapies to treat innate immune-related diseases.


Assuntos
Hematopoese/fisiologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Mediadores da Inflamação/fisiologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular/efeitos dos fármacos , Embrião de Mamíferos , Embrião não Mamífero , Hematopoese/efeitos dos fármacos , Inflamação/metabolismo , Mediadores da Inflamação/farmacologia , Masculino , Camundongos , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/metabolismo , Peixe-Zebra
20.
Genomics Proteomics Bioinformatics ; 13(6): 336-44, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26762955

RESUMO

Technological advances are important for innovative biological research. Development of molecular tools for DNA manipulation, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the clustered regularly-interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas), has revolutionized genome editing. These approaches can be used to develop potential therapeutic strategies to effectively treat heritable diseases. In the last few years, substantial progress has been made in CRISPR/Cas technology, including technical improvements and wide application in many model systems. This review describes recent advancements in genome editing with a particular focus on CRISPR/Cas, covering the underlying principles, technological optimization, and its application in zebrafish and other model organisms, disease modeling, and gene therapy used for personalized medicine.


Assuntos
Sistemas CRISPR-Cas , Engenharia Genética/métodos , Engenharia Genética/estatística & dados numéricos , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Terapia Genética/métodos , Genoma , Humanos , Modelos Biológicos , Mutagênese/fisiologia , Dedos de Zinco/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...