Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Development ; 151(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38722096

ABSTRACT

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.


Subject(s)
Endothelial Cells , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins , Lymphatic Vessels , Tumor Suppressor Proteins , Zebrafish Proteins , Zebrafish , Animals , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Zebrafish/genetics , Zebrafish/embryology , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Enhancer Elements, Genetic/genetics , Lymphatic Vessels/metabolism , Lymphatic Vessels/embryology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Endothelial Cells/metabolism , Lymphangiogenesis/genetics , CRISPR-Cas Systems/genetics , Promoter Regions, Genetic/genetics , Mice
2.
Science ; 381(6659): 799-804, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37590348

ABSTRACT

Piezo channels are critical cellular sensors of mechanical forces. Despite their large size, ubiquitous expression, and irreplaceable roles in an ever-growing list of physiological processes, few Piezo channel-binding proteins have emerged. In this work, we found that MyoD (myoblast determination)-family inhibitor proteins (MDFIC and MDFI) are PIEZO1/2 interacting partners. These transcriptional regulators bind to PIEZO1/2 channels, regulating channel inactivation. Using single-particle cryogenic electron microscopy, we mapped the interaction site in MDFIC to a lipidated, C-terminal helix that inserts laterally into the PIEZO1 pore module. These Piezo-interacting proteins fit all the criteria for auxiliary subunits, contribute to explaining the vastly different gating kinetics of endogenous Piezo channels observed in many cell types, and elucidate mechanisms potentially involved in human lymphatic vascular disease.


Subject(s)
Ion Channels , Myogenic Regulatory Factors , Humans , Cryoelectron Microscopy , HEK293 Cells , Ion Channel Gating , Ion Channels/chemistry , Ion Channels/genetics , Ion Channels/metabolism , Kinetics , Lymphatic Diseases/genetics , Mutation , Myogenic Regulatory Factors/chemistry , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Protein Domains , Myoblasts/metabolism , Animals , Mice
3.
Nature ; 614(7947): 343-348, 2023 02.
Article in English | MEDLINE | ID: mdl-36697821

ABSTRACT

Transcriptional enhancer elements are responsible for orchestrating the temporal and spatial control over gene expression that is crucial for programming cell identity during development1-3. Here we describe a novel enhancer element that is important for regulating the expression of Prox1 in lymphatic endothelial cells. This evolutionarily conserved enhancer is bound by key lymphatic transcriptional regulators including GATA2, FOXC2, NFATC1 and PROX1. Genome editing of the enhancer to remove five nucleotides encompassing the GATA2-binding site resulted in perinatal death of homozygous mutant mice due to profound lymphatic vascular defects. Lymphatic endothelial cells in enhancer mutant mice exhibited reduced expression of genes characteristic of lymphatic endothelial cell identity and increased expression of genes characteristic of haemogenic endothelium, and acquired the capacity to generate haematopoietic cells. These data not only reveal a transcriptional enhancer element important for regulating Prox1 expression and lymphatic endothelial cell identity but also demonstrate that the lymphatic endothelium has haemogenic capacity, ordinarily repressed by Prox1.


Subject(s)
Endothelial Cells , Enhancer Elements, Genetic , Hematopoiesis , Lymphatic Vessels , Animals , Mice , Endothelial Cells/metabolism , Enhancer Elements, Genetic/genetics , Hematopoiesis/genetics , Homeodomain Proteins/metabolism , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Transcription Factors/metabolism
4.
EMBO J ; 42(5): e109032, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36715213

ABSTRACT

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.


Subject(s)
Endothelial Cells , Lymphatic Vessels , Humans , Mice , Animals , Endothelial Cells/metabolism , Lymphatic Vessels/metabolism , Gene Expression Regulation , Endothelium, Vascular , Transcription Factors/metabolism , Lymphangiogenesis/genetics , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
5.
Nat Commun ; 13(1): 2018, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440627

ABSTRACT

The ubiquitin ligase NEDD4 promotes neural crest cell (NCC) survival and stem-cell like properties to regulate craniofacial and peripheral nervous system development. However, how ubiquitination and NEDD4 control NCC development remains unknown. Here we combine quantitative analysis of the proteome, transcriptome and ubiquitinome to identify key developmental signalling pathways that are regulated by NEDD4. We report 276 NEDD4 targets in NCCs and show that loss of NEDD4 leads to a pronounced global reduction in specific ubiquitin lysine linkages. We further show that NEDD4 contributes to the regulation of the NCC actin cytoskeleton by controlling ubiquitination and turnover of Profilin 1 to modulate filamentous actin polymerization. Taken together, our data provide insights into how NEDD4-mediated ubiquitination coordinates key regulatory processes during NCC development.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Neural Crest , Actins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Neural Crest/metabolism , Profilins/genetics , Profilins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
6.
Sci Transl Med ; 14(634): eabm4869, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35235341

ABSTRACT

Central conducting lymphatic anomaly (CCLA), characterized by the dysfunction of core collecting lymphatic vessels including the thoracic duct and cisterna chyli, and presenting as chylothorax, pleural effusions, chylous ascites, and lymphedema, is a severe disorder often resulting in fetal or perinatal demise. Although pathogenic variants in RAS/mitogen activated protein kinase (MAPK) signaling pathway components have been documented in some patients with CCLA, the genetic etiology of the disorder remains uncharacterized in most cases. Here, we identified biallelic pathogenic variants in MDFIC, encoding the MyoD family inhibitor domain containing protein, in seven individuals with CCLA from six independent families. Clinical manifestations of affected fetuses and children included nonimmune hydrops fetalis (NIHF), pleural and pericardial effusions, and lymphedema. Generation of a mouse model of human MDFIC truncation variants revealed that homozygous mutant mice died perinatally exhibiting chylothorax. The lymphatic vasculature of homozygous Mdfic mutant mice was profoundly mispatterned and exhibited major defects in lymphatic vessel valve development. Mechanistically, we determined that MDFIC controls collective cell migration, an important early event during the formation of lymphatic vessel valves, by regulating integrin ß1 activation and the interaction between lymphatic endothelial cells and their surrounding extracellular matrix. Our work identifies MDFIC variants underlying human lymphatic disease and reveals a crucial, previously unrecognized role for MDFIC in the lymphatic vasculature. Ultimately, understanding the genetic and mechanistic basis of CCLA will facilitate the development and implementation of new therapeutic approaches to effectively treat this complex disease.


Subject(s)
Chylothorax , Lymphatic Vessels , Lymphedema , Myogenic Regulatory Factors , Animals , Chylothorax/genetics , Chylothorax/metabolism , Endothelial Cells , Female , Humans , Hydrops Fetalis/genetics , Hydrops Fetalis/metabolism , Lymphatic Vessels/pathology , Lymphedema/genetics , Lymphedema/metabolism , Mice , Myogenic Regulatory Factors/genetics , Pregnancy
7.
Cell Death Dis ; 13(1): 75, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075134

ABSTRACT

Gonadogenesis is the process wherein two morphologically distinct organs, the testis and the ovary, arise from a common precursor. In mammals, maleness is driven by the expression of Sry. SRY subsequently upregulates the related family member Sox9 which is responsible for initiating testis differentiation while repressing factors critical to ovarian development such as FOXL2 and ß-catenin. Here, we report a hitherto uncharacterised role for the ubiquitin-protein ligase NEDD4 in this process. XY Nedd4-deficient mice exhibit complete male-to-female gonadal sex reversal shown by the ectopic upregulation of Foxl2 expression at the time of gonadal sex determination as well as insufficient upregulation of Sox9. This sex reversal extends to germ cells with ectopic expression of SYCP3 in XY Nedd4-/- germ cells and significantly higher Sycp3 transcripts in XY and XX Nedd4-deficient mice when compared to both XY and XX controls. Further, Nedd4-/- mice exhibit reduced gonadal precursor cell formation and gonadal size as a result of reduced proliferation within the developing gonad as well as reduced Nr5a1 expression. Together, these results establish an essential role for NEDD4 in XY gonadal sex determination and development and suggest a potential role for NEDD4 in orchestrating these cell fate decisions through the suppression of the female pathway to ensure proper testis differentiation.


Subject(s)
46, XX Testicular Disorders of Sex Development , Gonads , Nedd4 Ubiquitin Protein Ligases , Animals , Cell Differentiation/physiology , Female , Gene Expression Regulation, Developmental , Male , Mammals , Mice , Mice, Knockout , Nedd4 Ubiquitin Protein Ligases/metabolism , Ovary/metabolism , SOX9 Transcription Factor/metabolism , Testis/metabolism
8.
Dev Dyn ; 251(2): 336-349, 2022 02.
Article in English | MEDLINE | ID: mdl-34174014

ABSTRACT

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.


Subject(s)
Lymphatic Vessels , Polycystic Kidney Diseases , Animals , Lymphatic Vessels/metabolism , Mice , Mice, Knockout , Morphogenesis/genetics , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Protein Kinase C , Receptor Protein-Tyrosine Kinases/metabolism , Wnt Signaling Pathway/genetics , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism
9.
Nat Metab ; 3(9): 1175-1188, 2021 09.
Article in English | MEDLINE | ID: mdl-34545251

ABSTRACT

Visceral adipose tissue (VAT) encases mesenteric lymphatic vessels and lymph nodes through which lymph is transported from the intestine and mesentery. Whether mesenteric lymphatics contribute to adipose tissue inflammation and metabolism and insulin resistance is unclear. Here we show that obesity is associated with profound and progressive dysfunction of the mesenteric lymphatic system in mice and humans. We find that lymph from mice and humans consuming a high-fat diet (HFD) stimulates lymphatic vessel growth, leading to the formation of highly branched mesenteric lymphatic vessels that 'leak' HFD-lymph into VAT and, thereby, promote insulin resistance. Mesenteric lymphatic dysfunction is regulated by cyclooxygenase (COX)-2 and vascular endothelial growth factor (VEGF)-C-VEGF receptor (R)3 signalling. Lymph-targeted inhibition of COX-2 using a glyceride prodrug approach reverses mesenteric lymphatic dysfunction, visceral obesity and inflammation and restores glycaemic control in mice. Targeting obesity-associated mesenteric lymphatic dysfunction thus represents a potential therapeutic option to treat metabolic disease.


Subject(s)
Insulin Resistance , Lymphatic Vessels/physiopathology , Mesentery/physiopathology , Obesity, Abdominal/physiopathology , Adult , Aged , Animals , Cyclooxygenase 2/metabolism , Female , Humans , Intra-Abdominal Fat/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Obesity, Abdominal/therapy , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular Endothelial Growth Factor C/metabolism
10.
Hum Mutat ; 42(11): 1399-1421, 2021 11.
Article in English | MEDLINE | ID: mdl-34387894

ABSTRACT

GATA2 deficiency syndrome (G2DS) is a rare autosomal dominant genetic disease predisposing to a range of symptoms, of which myeloid malignancy and immunodeficiency including recurrent infections are most common. In the last decade since it was first reported, there have been over 480 individuals identified carrying a pathogenic or likely pathogenic germline GATA2 variant with symptoms of G2DS, with 240 of these confirmed to be familial and 24 de novo. For those that develop myeloid malignancy (75% of all carriers with G2DS disease symptoms), the median age of onset is 17 years (range 0-78 years) and myelodysplastic syndrome is the first diagnosis in 75% of these cases with acute myeloid leukemia in a further 9%. All variant types appear to predispose to myeloid malignancy and immunodeficiency. Apart from lymphedema in which haploinsufficiency seems necessary, the mutational requirements of the other less common G2DS phenotypes is still unclear. These predominantly loss-of-function variants impact GATA2 expression and function in numerous ways including perturbations to DNA binding, protein structure, protein:protein interactions, and gene transcription, splicing, and expression. In this review, we provide the first expert-curated ACMG/AMP classification with codes of published variants compatible for use in clinical or diagnostic settings.


Subject(s)
GATA2 Deficiency/genetics , GATA2 Transcription Factor/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Cohort Studies , Humans , Infant , Infant, Newborn , Middle Aged , Young Adult
11.
Int J Mol Sci ; 22(14)2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34299378

ABSTRACT

The importance of lymphatic vessels in a myriad of human diseases is rapidly gaining recognition; lymphatic vessel dysfunction is a feature of disorders including congenital lymphatic anomalies, primary lymphoedema and obesity, while improved lymphatic vessel function increases the efficacy of immunotherapy for cancer and neurological disease and promotes cardiac repair following myocardial infarction. Understanding how the growth and function of lymphatic vessels is precisely regulated therefore stands to inform the development of novel therapeutics applicable to a wide range of human diseases. Lymphatic vascular development is initiated during embryogenesis following establishment of the major blood vessels and the onset of blood flow. Lymphatic endothelial progenitor cells arise from a combination of venous and non-venous sources to generate the initial lymphatic vascular structures in the vertebrate embryo, which are then further ramified and remodelled to elaborate an extensive lymphatic vascular network. Signalling mediated via vascular endothelial growth factor (VEGF) family members and vascular endothelial growth factor receptor (VEGFR) tyrosine kinases is crucial for development of both the blood and lymphatic vascular networks, though distinct components are utilised to different degrees in each vascular compartment. Although much is known about the regulation of VEGFA/VEGFR2 signalling in the blood vasculature, less is understood regarding the mechanisms by which VEGFC/VEGFD/VEGFR3 signalling is regulated during lymphatic vascular development. This review will focus on recent advances in our understanding of the cellular and molecular mechanisms regulating VEGFA-, VEGFC- and VEGFD-mediated signalling via VEGFRs which are important for driving the construction of lymphatic vessels during development and disease.


Subject(s)
Lymphatic Vessels/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/physiology , Animals , Endothelial Cells/metabolism , Humans , Lymphangiogenesis/physiology
12.
Development ; 148(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34080610

ABSTRACT

The lymphatic vasculature is an integral component of the cardiovascular system. It is essential to maintain tissue fluid homeostasis, direct immune cell trafficking and absorb dietary lipids from the digestive tract. Major advances in our understanding of the genetic and cellular events important for constructing the lymphatic vasculature during development have recently been made. These include the identification of novel sources of lymphatic endothelial progenitor cells, the recognition of lymphatic endothelial cell specialisation and heterogeneity, and discovery of novel genes and signalling pathways underpinning developmental lymphangiogenesis. Here, we review these advances and discuss how they inform our understanding of lymphatic network formation, function and dysfunction.


Subject(s)
Cardiovascular System/growth & development , Lymphangiogenesis/physiology , Lymphatic Vessels/physiology , Animals , Cardiovascular System/cytology , Cardiovascular System/embryology , Endothelial Cells/physiology , Homeostasis , Humans , Lymphatic Vessels/cytology , Lymphatic Vessels/embryology , Signal Transduction
13.
Cell ; 182(2): 270-296, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32707093

ABSTRACT

Mammals have two specialized vascular circulatory systems: the blood vasculature and the lymphatic vasculature. The lymphatic vasculature is a unidirectional conduit that returns filtered interstitial arterial fluid and tissue metabolites to the blood circulation. It also plays major roles in immune cell trafficking and lipid absorption. As we discuss in this review, the molecular characterization of lymphatic vascular development and our understanding of this vasculature's role in pathophysiological conditions has greatly improved in recent years, changing conventional views about the roles of the lymphatic vasculature in health and disease. Morphological or functional defects in the lymphatic vasculature have now been uncovered in several pathological conditions. We propose that subtle asymptomatic alterations in lymphatic vascular function could underlie the variability seen in the body's response to a wide range of human diseases.


Subject(s)
Lymphatic Vessels/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , History, 21st Century , Humans , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphangiogenesis , Lymphatic Diseases/genetics , Lymphatic Diseases/history , Lymphatic Diseases/pathology , Lymphatic Metastasis , Lymphatic Vessels/anatomy & histology , Lymphatic Vessels/cytology , Neoplasms/metabolism , Neoplasms/pathology , Vascular Endothelial Growth Factor Receptor-3/genetics
14.
J Clin Invest ; 130(6): 3315-3328, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32182215

ABSTRACT

The atypical cadherin FAT4 has established roles in the regulation of planar cell polarity and Hippo pathway signaling that are cell context dependent. The recent identification of FAT4 mutations in Hennekam syndrome, features of which include lymphedema, lymphangiectasia, and mental retardation, uncovered an important role for FAT4 in the lymphatic vasculature. Hennekam syndrome is also caused by mutations in collagen and calcium binding EGF domains 1 (CCBE1) and ADAM metallopeptidase with thrombospondin type 1 motif 3 (ADAMTS3), encoding a matrix protein and protease, respectively, that regulate activity of the key prolymphangiogenic VEGF-C/VEGFR3 signaling axis by facilitating the proteolytic cleavage and activation of VEGF-C. The fact that FAT4, CCBE1, and ADAMTS3 mutations underlie Hennekam syndrome suggested that all 3 genes might function in a common pathway. We identified FAT4 as a target gene of GATA-binding protein 2 (GATA2), a key transcriptional regulator of lymphatic vascular development and, in particular, lymphatic vessel valve development. Here, we demonstrate that FAT4 functions in a lymphatic endothelial cell-autonomous manner to control cell polarity in response to flow and is required for lymphatic vessel morphogenesis throughout development. Our data reveal a crucial role for FAT4 in lymphangiogenesis and shed light on the mechanistic basis by which FAT4 mutations underlie a human lymphedema syndrome.


Subject(s)
Cadherins/metabolism , Cell Polarity , Endothelial Cells/metabolism , Lymphangiogenesis , Lymphatic Vessels/metabolism , Animals , Cadherins/genetics , Endothelial Cells/pathology , Female , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Humans , Lymphatic Vessels/pathology , Lymphedema/genetics , Lymphedema/metabolism , Lymphedema/pathology , Mice , Mice, Transgenic , Syndrome
15.
Science ; 366(6470): 1193-1194, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31806798
16.
Elife ; 82019 04 30.
Article in English | MEDLINE | ID: mdl-31038457

ABSTRACT

Lymphatic vascular development involves specification of lymphatic endothelial progenitors that subsequently undergo sprouting, proliferation and tissue growth to form a complex second vasculature. The Hippo pathway and effectors Yap and Taz control organ growth and regulate morphogenesis and cellular proliferation. Yap and Taz control angiogenesis but a role in lymphangiogenesis remains to be fully elucidated. Here we show that YAP displays dynamic changes in lymphatic progenitors and Yap1 is essential for lymphatic vascular development in zebrafish. Maternal and Zygotic (MZ) yap1 mutants show normal specification of lymphatic progenitors, abnormal cellular sprouting and reduced numbers of lymphatic progenitors emerging from the cardinal vein during lymphangiogenesis. Furthermore, Yap1 is indispensable for Vegfc-induced proliferation in a transgenic model of Vegfc overexpression. Paracrine Vegfc-signalling ultimately increases nuclear YAP in lymphatic progenitors to control lymphatic development. We thus identify a role for Yap in lymphangiogenesis, acting downstream of Vegfc to promote expansion of this vascular lineage.


Subject(s)
Cell Proliferation/drug effects , Lymphatic Vessels/drug effects , Trans-Activators/metabolism , Trans-Activators/pharmacology , Vascular Endothelial Growth Factor C/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/pharmacology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Female , Gene Expression Regulation, Developmental/drug effects , Gene Knockout Techniques , Lymphangiogenesis/drug effects , Lymphatic Vessels/cytology , Male , Morphogenesis/drug effects , Trans-Activators/genetics , YAP-Signaling Proteins , Zebrafish/genetics , Zebrafish Proteins/genetics
17.
Dev Cell ; 49(2): 279-292.e5, 2019 04 22.
Article in English | MEDLINE | ID: mdl-31014480

ABSTRACT

The correct assignment of cell fate within fields of multipotent progenitors is essential for accurate tissue diversification. The first lymphatic vessels arise from pre-existing veins after venous endothelial cells become specified as lymphatic progenitors. Prox1 specifies lymphatic fate and labels these progenitors; however, the mechanisms restricting Prox1 expression and limiting the progenitor pool remain unknown. We identified a zebrafish mutant that displayed premature, expanded, and prolonged lymphatic specification. The gene responsible encodes the regulator of alternative splicing, Nova2. In zebrafish and human endothelial cells, Nova2 selectively regulates pre-mRNA splicing for components of signaling pathways and phosphoproteins. Nova2-deficient endothelial cells display increased Mapk/Erk signaling, and Prox1 expression is dynamically controlled by Erk signaling. We identify a mechanism whereby Nova2-regulated splicing constrains Erk signaling, thus limiting lymphatic progenitor cell specification. This identifies the capacity of a factor that tunes mRNA splicing to control assignment of cell fate during vascular differentiation.


Subject(s)
Lymphatic Vessels/metabolism , MAP Kinase Signaling System , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Alternative Splicing , Animals , Cell Differentiation , Cell Lineage , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Homeodomain Proteins/metabolism , Humans , Lymphangiogenesis , Lymphatic Vessels/cytology , Male , Neuro-Oncological Ventral Antigen , Tumor Suppressor Proteins/metabolism , Veins/cytology , Veins/metabolism , Zebrafish
18.
Methods Mol Biol ; 1846: 19-35, 2018.
Article in English | MEDLINE | ID: mdl-30242750

ABSTRACT

The capacity to visualize the lymphatic vasculature in three-dimensions has revolutionized our understanding of the morphogenetic mechanisms important for constructing the lymphatic vascular network during development. Two complementary approaches are commonly employed to assess the function of genes and signaling pathways important for development of the dermal lymphatic vasculature in the mouse embryo. The first of these is whole-mount immunostaining of embryonic skin to analyze dermal lymphatic vessel network patterning and morphology in two and three dimensions. The second is immunostaining of thin tissue sections to examine lymphatic vessel identity, lumen formation and protein localization within discrete lymphatic endothelial cells in a two-dimensional setting. Here we present detailed protocols for multicolor immunofluorescent immunostaining of embryonic dorsal skin and thin tissue cryosections. Each of these methods generates high-resolution images of the dermal lymphatic vasculature, yielding information integral to in-depth characterization of lymphatic vessel phenotypes in the developing mouse embryo.


Subject(s)
Angiography , Dermis/blood supply , Lymphangiogenesis , Lymphatic Vessels/diagnostic imaging , Animals , Dermis/embryology , Female , Fluorescent Antibody Technique , Mice , Microscopy, Confocal
19.
Development ; 145(10)2018 05 17.
Article in English | MEDLINE | ID: mdl-29773646

ABSTRACT

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.


Subject(s)
Capillaries/cytology , Endothelial Cells/cytology , Homeodomain Proteins/genetics , Lymphangiogenesis/physiology , Lymphatic Vessels/embryology , Stem Cells/cytology , Tumor Suppressor Proteins/genetics , Animals , Calcium-Binding Proteins/genetics , GATA2 Transcription Factor/genetics , Lymphangiogenesis/genetics , Lymphatic Vessels/cytology , Mice , Mice, Transgenic , Vascular Endothelial Growth Factor C/biosynthesis , Vascular Endothelial Growth Factor C/genetics
20.
Nat Commun ; 9(1): 1511, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29666442

ABSTRACT

Tissue and vessel wall stiffening alters endothelial cell properties and contributes to vascular dysfunction. However, whether extracellular matrix (ECM) stiffness impacts vascular development is not known. Here we show that matrix stiffness controls lymphatic vascular morphogenesis. Atomic force microscopy measurements in mouse embryos reveal that venous lymphatic endothelial cell (LEC) progenitors experience a decrease in substrate stiffness upon migration out of the cardinal vein, which induces a GATA2-dependent transcriptional program required to form the first lymphatic vessels. Transcriptome analysis shows that LECs grown on a soft matrix exhibit increased GATA2 expression and a GATA2-dependent upregulation of genes involved in cell migration and lymphangiogenesis, including VEGFR3. Analyses of mouse models demonstrate a cell-autonomous function of GATA2 in regulating LEC responsiveness to VEGF-C and in controlling LEC migration and sprouting in vivo. Our study thus uncovers a mechanism by which ECM stiffness dictates the migratory behavior of LECs during early lymphatic development.


Subject(s)
GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Lymphangiogenesis/genetics , Lymphatic Vessels/physiology , Animals , Cell Movement/genetics , Endothelial Cells/physiology , Female , GATA2 Transcription Factor/genetics , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Lymphatic Vessels/cytology , Male , Mice , Mice, Transgenic , Primary Cell Culture , RNA, Small Interfering/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...