Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Development ; 151(9)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38722096

RESUMO

During embryonic development, lymphatic endothelial cell (LEC) precursors are distinguished from blood endothelial cells by the expression of Prospero-related homeobox 1 (Prox1), which is essential for lymphatic vasculature formation in mouse and zebrafish. Prox1 expression initiation precedes LEC sprouting and migration, serving as the marker of specified LECs. Despite its crucial role in lymphatic development, Prox1 upstream regulation in LECs remains to be uncovered. SOX18 and COUP-TFII are thought to regulate Prox1 in mice by binding its promoter region. However, the specific regulation of Prox1 expression in LECs remains to be studied in detail. Here, we used evolutionary conservation and chromatin accessibility to identify enhancers located in the proximity of zebrafish prox1a active in developing LECs. We confirmed the functional role of the identified sequences through CRISPR/Cas9 mutagenesis of a lymphatic valve enhancer. The deletion of this region results in impaired valve morphology and function. Overall, our results reveal an intricate control of prox1a expression through a collection of enhancers. Ray-finned fish-specific distal enhancers drive pan-lymphatic expression, whereas vertebrate-conserved proximal enhancers refine expression in functionally distinct subsets of lymphatic endothelium.


Assuntos
Células Endoteliais , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio , Vasos Linfáticos , Proteínas Supressoras de Tumor , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Peixe-Zebra/genética , Peixe-Zebra/embriologia , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Elementos Facilitadores Genéticos/genética , Vasos Linfáticos/metabolismo , Vasos Linfáticos/embriologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Células Endoteliais/metabolismo , Linfangiogênese/genética , Sistemas CRISPR-Cas/genética , Regiões Promotoras Genéticas/genética , Camundongos
3.
EMBO J ; 42(5): e109032, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36715213

RESUMO

Despite a growing catalog of secreted factors critical for lymphatic network assembly, little is known about the mechanisms that modulate the expression level of these molecular cues in blood vascular endothelial cells (BECs). Here, we show that a BEC-specific transcription factor, SOX7, plays a crucial role in a non-cell-autonomous manner by modulating the transcription of angiocrine signals to pattern lymphatic vessels. While SOX7 is not expressed in lymphatic endothelial cells (LECs), the conditional loss of SOX7 function in mouse embryos causes a dysmorphic dermal lymphatic phenotype. We identify novel distant regulatory regions in mice and humans that contribute to directly repressing the transcription of a major lymphangiogenic growth factor (Vegfc) in a SOX7-dependent manner. Further, we show that SOX7 directly binds HEY1, a canonical repressor of the Notch pathway, suggesting that transcriptional repression may also be modulated by the recruitment of this protein partner at Vegfc genomic regulatory regions. Our work unveils a role for SOX7 in modulating downstream signaling events crucial for lymphatic patterning, at least in part via the transcriptional repression of VEGFC levels in the blood vascular endothelium.


Assuntos
Células Endoteliais , Vasos Linfáticos , Humanos , Camundongos , Animais , Células Endoteliais/metabolismo , Vasos Linfáticos/metabolismo , Regulação da Expressão Gênica , Endotélio Vascular , Fatores de Transcrição/metabolismo , Linfangiogênese/genética , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo
4.
Cell Rep ; 39(12): 110982, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35732122

RESUMO

Lymphangiogenesis, formation of lymphatic vessels from pre-existing vessels, is a dynamic process that requires cell migration. Regardless of location, migrating lymphatic endothelial cell (LEC) progenitors probe their surroundings to form the lymphatic network. Lymphatic-development regulation requires the transcription factor MAFB in different species. Zebrafish Mafba, expressed in LEC progenitors, is essential for their migration in the trunk. However, the transcriptional mechanism that orchestrates LEC migration in different lymphatic endothelial beds remains elusive. Here, we uncover topographically different requirements of the two paralogs, Mafba and Mafbb, for LEC migration. Both mafba and mafbb are necessary for facial lymphatic development, but mafbb is dispensable for trunk lymphatic development. On the molecular level, we demonstrate a regulatory network where Vegfc-Vegfd-SoxF-Mafba-Mafbb is essential in facial lymphangiogenesis. We identify that mafba and mafbb tune the directionality of LEC migration and vessel morphogenesis that is ultimately necessary for lymphatic function.


Assuntos
Vasos Linfáticos , Peixe-Zebra , Animais , Movimento Celular , Células Endoteliais , Linfangiogênese , Morfogênese , Transdução de Sinais
5.
Elife ; 112022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35316177

RESUMO

The migration of lymphatic endothelial cells (LECs) is key for the development of the complex and vast lymphatic vascular network that pervades most tissues in an organism. In zebrafish, arterial intersegmental vessels together with chemokines have been shown to promote lymphatic cell migration from the horizontal myoseptum (HM). We observed that emergence of mural cells around the intersegmental arteries coincides with lymphatic departure from HM which raised the possibility that arterial mural cells promote LEC migration. Our live imaging and cell ablation experiments revealed that LECs migrate slower and fail to establish the lymphatic vascular network in the absence of arterial mural cells. We determined that mural cells are a source for the C-X-C motif chemokine 12 (Cxcl12a and Cxcl12b), vascular endothelial growth factor C (Vegfc) and collagen and calcium-binding EGF domain-containing protein 1 (Ccbe1). We showed that chemokine and growth factor signalling function cooperatively to induce robust LEC migration. Specifically, Vegfc-Vegfr3 signalling, but not chemokines, induces extracellular signal-regulated kinase (ERK) activation in LECs, and has an additional pro-survival role in LECs during the migration. Together, the identification of mural cells as a source for signals that guide LEC migration and survival will be important in the future design for rebuilding lymphatic vessels in disease contexts.


Assuntos
Células Endoteliais , Fator C de Crescimento do Endotélio Vascular , Animais , Artérias , Sinais (Psicologia) , Células Endoteliais/fisiologia , Fator C de Crescimento do Endotélio Vascular/fisiologia , Peixe-Zebra
6.
Dev Dyn ; 251(2): 336-349, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34174014

RESUMO

BACKGROUND: Lymphatic vascular development is regulated by well-characterized signaling and transcriptional pathways. These pathways regulate lymphatic endothelial cell (LEC) migration, motility, polarity, and morphogenesis. Canonical and non-canonical WNT signaling pathways are known to control LEC polarity and development of lymphatic vessels and valves. PKD1, encoding Polycystin-1, is the most commonly mutated gene in polycystic kidney disease but has also been shown to be essential in lymphatic vascular morphogenesis. The mechanism by which Pkd1 acts during lymphangiogenesis remains unclear. RESULTS: Here we find that loss of non-canonical WNT signaling components Wnt5a and Ryk phenocopy lymphatic defects seen in Pkd1 knockout mice. To investigate genetic interaction, we generated Pkd1;Wnt5a double knockout mice. Loss of Wnt5a suppressed phenotypes seen in the lymphatic vasculature of Pkd1-/- mice and Pkd1 deletion suppressed phenotypes observed in Wnt5a-/- mice. Thus, we report mutually suppressive roles for Pkd1 and Wnt5a, with developing lymphatic networks restored to a more wild type state in double mutant mice. This genetic interaction between Pkd1 and the non-canonical WNT signaling pathway ultimately controls LEC polarity and the morphogenesis of developing vessel networks. CONCLUSION: Our work suggests that Pkd1 acts at least in part by regulating non-canonical WNT signaling during the formation of lymphatic vascular networks.


Assuntos
Vasos Linfáticos , Doenças Renais Policísticas , Animais , Vasos Linfáticos/metabolismo , Camundongos , Camundongos Knockout , Morfogênese/genética , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Proteína Quinase C , Receptores Proteína Tirosina Quinases/metabolismo , Via de Sinalização Wnt/genética , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo
7.
Nat Cell Biol ; 23(11): 1136-1147, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34750583

RESUMO

The development of a functional vasculature requires the coordinated control of cell fate, lineage differentiation and network growth. Cellular proliferation is spatiotemporally regulated in developing vessels, but how this is orchestrated in different lineages is unknown. Here, using a zebrafish genetic screen for lymphatic-deficient mutants, we uncover a mutant for the RNA helicase Ddx21. Ddx21 cell-autonomously regulates lymphatic vessel development. An established regulator of ribosomal RNA synthesis and ribosome biogenesis, Ddx21 is enriched in sprouting venous endothelial cells in response to Vegfc-Flt4 signalling. Ddx21 function is essential for Vegfc-Flt4-driven endothelial cell proliferation. In the absence of Ddx21, endothelial cells show reduced ribosome biogenesis, p53 and p21 upregulation and cell cycle arrest that blocks lymphangiogenesis. Thus, Ddx21 coordinates the lymphatic endothelial cell response to Vegfc-Flt4 signalling by balancing ribosome biogenesis and p53 function. This mechanism may be targetable in diseases of excessive lymphangiogenesis such as cancer metastasis or lymphatic malformation.


Assuntos
Proliferação de Células , RNA Helicases DEAD-box/metabolismo , Células Endoteliais/enzimologia , Linfangiogênese , Vasos Linfáticos/enzimologia , RNA Ribossômico/biossíntese , Ribossomos/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Pontos de Checagem do Ciclo Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , RNA Helicases DEAD-box/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Vasos Linfáticos/embriologia , RNA Ribossômico/genética , Ribossomos/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Fator C de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
8.
Development ; 148(4)2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33547133

RESUMO

Previous studies have shown that Vasohibin 1 (Vash1) is stimulated by VEGFs in endothelial cells and that its overexpression interferes with angiogenesis in vivo Recently, Vash1 was found to mediate tubulin detyrosination, a post-translational modification that is implicated in many cell functions, such as cell division. Here, we used the zebrafish embryo to investigate the cellular and subcellular mechanisms of Vash1 on endothelial microtubules during formation of the trunk vasculature. We show that microtubules within venous-derived secondary sprouts are strongly and selectively detyrosinated in comparison with other endothelial cells, and that this difference is lost upon vash1 knockdown. Vash1 depletion in zebrafish specifically affected secondary sprouting from the posterior cardinal vein, increasing endothelial cell divisions and cell number in the sprouts. We show that altering secondary sprout numbers and structure upon Vash1 depletion leads to defective lymphatic vessel formation and ectopic lymphatic progenitor specification in the zebrafish trunk.


Assuntos
Proteínas de Ciclo Celular/genética , Desenvolvimento Embrionário/genética , Linfangiogênese/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Sequência Conservada , Evolução Molecular , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Microtúbulos/metabolismo , Modelos Biológicos
9.
Dev Dyn ; 249(10): 1201-1216, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32525258

RESUMO

BACKGROUND: Lymphatic vessels play key roles in tissue fluid homeostasis, immune cell trafficking and in diverse disease settings. Lymphangiogenesis requires lymphatic endothelial cell (LEC) differentiation, proliferation, migration, and co-ordinated network formation, yet the transcriptional regulators underpinning these processes remain to be fully understood. The transcription factor MAFB was recently identified as essential for lymphangiogenesis in zebrafish and in cultured human LECs. MAFB is activated in response to VEGFC-VEGFR3 signaling and acts as a downstream effector. However, it remains unclear if the role of MAFB in lymphatic development is conserved in the mammalian embryo. RESULTS: We generated a Mafb loss-of-function mouse using CRISPR/Cas9 gene editing. Mafb mutant mice presented with perinatal lethality associated with cyanosis. We identify a role for MAFB in modifying lymphatic network morphogenesis in the developing dermis, as well as developing and postnatal diaphragm. Furthermore, mutant vessels displayed excessive smooth muscle cell coverage, suggestive of a defect in the maturation of lymphatic networks. CONCLUSIONS: This work confirms a conserved role for MAFB in murine lymphatics that is subtle and modulatory and may suggest redundancy in MAF family transcription factors during lymphangiogenesis.


Assuntos
Linfangiogênese/fisiologia , Vasos Linfáticos/metabolismo , Fator de Transcrição MafB/fisiologia , Animais , Sistemas CRISPR-Cas , Cruzamentos Genéticos , Genoma , Genótipo , Hibridização In Situ , Camundongos , Camundongos Knockout , Mutação , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Tempo
10.
Development ; 145(10)2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29773646

RESUMO

Despite the essential role of the lymphatic vasculature in tissue homeostasis and disease, knowledge of the organ-specific origins of lymphatic endothelial progenitor cells remains limited. The assumption that most murine embryonic lymphatic endothelial cells (LECs) are venous derived has recently been challenged. Here, we show that the embryonic dermal blood capillary plexus constitutes an additional, local source of LECs that contributes to the formation of the dermal lymphatic vascular network. We describe a novel mechanism whereby rare PROX1-positive endothelial cells exit the capillary plexus in a Ccbe1-dependent manner to establish discrete LEC clusters. As development proceeds, these clusters expand and further contribute to the growing lymphatic system. Lineage tracing and analyses of Gata2-deficient mice confirmed that these clusters are endothelial in origin. Furthermore, ectopic expression of Vegfc in the vasculature increased the number of PROX1-positive progenitors within the capillary bed. Our work reveals a novel source of lymphatic endothelial progenitors employed during construction of the dermal lymphatic vasculature and demonstrates that the blood vasculature is likely to remain an ongoing source of LECs during organogenesis, raising the question of whether a similar mechanism operates during pathological lymphangiogenesis.


Assuntos
Capilares/citologia , Células Endoteliais/citologia , Proteínas de Homeodomínio/genética , Linfangiogênese/fisiologia , Vasos Linfáticos/embriologia , Células-Tronco/citologia , Proteínas Supressoras de Tumor/genética , Animais , Proteínas de Ligação ao Cálcio/genética , Fator de Transcrição GATA2/genética , Linfangiogênese/genética , Vasos Linfáticos/citologia , Camundongos , Camundongos Transgênicos , Fator C de Crescimento do Endotélio Vascular/biossíntese , Fator C de Crescimento do Endotélio Vascular/genética
11.
Cell Rep ; 7(3): 623-33, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24767999

RESUMO

Lymphatic vessels arise during development through sprouting of precursor cells from veins, which is regulated by known signaling and transcriptional mechanisms. The ongoing elaboration of vessels to form a network is less well understood. This involves cell polarization, coordinated migration, adhesion, mixing, regression, and shape rearrangements. We identified a zebrafish mutant, lymphatic and cardiac defects 1 (lyc1), with reduced lymphatic vessel development. A mutation in polycystic kidney disease 1a was responsible for the phenotype. PKD1 is the most frequently mutated gene in autosomal dominant polycystic kidney disease (ADPKD). Initial lymphatic precursor sprouting is normal in lyc1 mutants, but ongoing migration fails. Loss of Pkd1 in mice has no effect on precursor sprouting but leads to failed morphogenesis of the subcutaneous lymphatic network. Individual lymphatic endothelial cells display defective polarity, elongation, and adherens junctions. This work identifies a highly selective and unexpected role for Pkd1 in lymphatic vessel morphogenesis during development.


Assuntos
Linfangiogênese , Vasos Linfáticos/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Animais Geneticamente Modificados , Células Cultivadas , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Junções Intercelulares/metabolismo , Linfonodos/crescimento & desenvolvimento , Linfonodos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Fenótipo , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Rim Policístico Autossômico Dominante/patologia , Canais de Cátion TRPP/antagonistas & inibidores , Canais de Cátion TRPP/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
12.
Dev Biol ; 386(1): 25-33, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24361262

RESUMO

During embryogenesis, lymphatic endothelial progenitor cells first arise from a subset of blood vascular endothelial cells in the dorsolateral aspects of the cardinal veins. The molecular cues responsible for defining the regionalisation of such a discrete pool of progenitors remain uncharacterised. Here we identify a novel function for CYP26B1, an enzyme known to play a role in tissue morphogenesis by fine-tuning retinoic acid (RA) concentration, in regulating lymphangiogenesis. Cyp26b1-null mice, in which RA levels are elevated, exhibited an increased number of lymphatic endothelial progenitor cells in the cardinal veins, together with hyperplastic, blood filled lymph sacs and hyperplastic dermal lymphatic vessels. Conversely, mice over-expressing Cyp26b1 had hypoplastic lymph sacs and lymphatic vessels. Our data suggest that RA clearance by CYP26B1 in the vicinity of lymphatic endothelial progenitor cells is important for determining the position and size of the progenitor pool specified. Our studies identify a genetic pathway that underpins the architecture of the developing lymphatics and define CYP26B1 as a novel modulator of lymphatic vascular patterning.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Linfangiogênese , Sistema Linfático/embriologia , Vasos Linfáticos/metabolismo , Retinoides/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Cruzamentos Genéticos , Células Endoteliais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Fenótipo , Ácido Retinoico 4 Hidroxilase , Transdução de Sinais , Transgenes , Tretinoína/metabolismo
13.
Blood ; 123(7): 1102-12, 2014 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-24269955

RESUMO

Vascular endothelial growth factor-D (VEGFD) is a potent pro-lymphangiogenic molecule during tumor growth and is considered a key therapeutic target to modulate metastasis. Despite roles in pathological neo-lymphangiogenesis, the characterization of an endogenous role for VEGFD in vascular development has remained elusive. Here, we used zebrafish to assay for genetic interactions between the Vegf/Vegf-receptor pathway and SoxF transcription factors and identified a specific interaction between Vegfd and Sox18. Double knockdown zebrafish embryos for Sox18/Vegfd and Sox7/Vegfd exhibit defects in arteriovenous differentiation. Supporting this observation, we found that Sox18/Vegfd double but not single knockout mice displayed dramatic vascular development defects. We find that VEGFD-mitogen-activated protein kinase kinase-extracellular signal-regulated kinase signaling modulates SOX18-mediated transcription, functioning at least in part by enhancing nuclear concentration and transcriptional activity in vascular endothelial cells. This work suggests that VEGFD-mediated pathologies include or involve an underlying dysregulation of SOXF-mediated transcriptional networks.


Assuntos
Vasos Sanguíneos/embriologia , Neovascularização Fisiológica/genética , Fatores de Transcrição SOXF/metabolismo , Fator D de Crescimento do Endotélio Vascular/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Embrião de Mamíferos , Embrião não Mamífero , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição SOXF/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
14.
Blood ; 110(3): 1064-72, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17449800

RESUMO

Although proinflammatory cytokines are key mediators of tissue damage during graft-versus-host disease (GVHD), IFNgamma has previously been attributed with both protective and pathogenic effects. We have resolved this paradox by using wild-type (wt), IFNgamma(-/-), and IFNgammaR(-/-) mice as donors or recipients in well-described models of allogeneic stem cell transplantation (SCT). We show that donor-derived IFNgamma augments acute GVHD via direct effects on (1) the donor T cell to promote T helper 1 (Th1) differentiation and (2) the gastrointestinal (GI) tract to augment inflammatory cytokine generation. However, these detrimental effects are overwhelmed by a protective role of IFNgamma in preventing the development of idiopathic pneumonia syndrome (IPS). This is the result of direct effects on pulmonary parenchyma to prevent donor cell migration and expansion within the lung. Thus, IFNgamma is the key cytokine differentially controlling the development of IPS and gastrointestinal GVHD after allogeneic SCT.


Assuntos
Gastroenteropatias/imunologia , Doença Enxerto-Hospedeiro/imunologia , Interferon gama/imunologia , Pneumonia/imunologia , Transplante de Células-Tronco , Doença Aguda , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Feminino , Gastroenteropatias/complicações , Gastroenteropatias/genética , Gastroenteropatias/patologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/patologia , Doença Enxerto-Hospedeiro/complicações , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/patologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interferon gama/deficiência , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Pneumonia/etiologia , Pneumonia/genética , Pneumonia/patologia , Síndrome , Células Th1/imunologia , Células Th1/patologia , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...