Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 307
Filtrar
1.
Nat Commun ; 15(1): 5547, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38956047

RESUMO

The meninges are critical for the brain functions, but the diversity of meningeal cell types and intercellular interactions have yet to be thoroughly examined. Here we identify a population of meningeal lymphatic supporting cells (mLSCs) in the zebrafish leptomeninges, which are specifically labeled by ependymin. Morphologically, mLSCs form membranous structures that enwrap the majority of leptomeningeal blood vessels and all the mural lymphatic endothelial cells (muLECs). Based on its unique cellular morphologies and transcriptional profile, mLSC is characterized as a unique cell type different from all the currently known meningeal cell types. Because of the formation of supportive structures and production of pro-lymphangiogenic factors, mLSCs not only promote muLEC development and maintain the dispersed distributions of muLECs in the leptomeninges, but also are required for muLEC regeneration after ablation. This study characterizes a newly identified cell type in leptomeninges, mLSC, which is required for muLEC development, maintenance, and regeneration.


Assuntos
Células Endoteliais , Meninges , Peixe-Zebra , Animais , Meninges/citologia , Meninges/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/citologia , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Vasos Linfáticos/citologia , Vasos Linfáticos/metabolismo , Animais Geneticamente Modificados , Linfangiogênese/fisiologia , Regeneração/fisiologia
2.
PLoS One ; 19(5): e0302926, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38718095

RESUMO

Zinc Finger MIZ-Type Containing 1 (Zmiz1), also known as ZIMP10 or RAI17, is a transcription cofactor and member of the Protein Inhibitor of Activated STAT (PIAS) family of proteins. Zmiz1 is critical for a variety of biological processes including vascular development. However, its role in the lymphatic vasculature is unknown. In this study, we utilized human dermal lymphatic endothelial cells (HDLECs) and an inducible, lymphatic endothelial cell (LEC)-specific Zmiz1 knockout mouse model to investigate the role of Zmiz1 in LECs. Transcriptional profiling of ZMIZ1-deficient HDLECs revealed downregulation of genes crucial for lymphatic vessel development. Additionally, our findings demonstrated that loss of Zmiz1 results in reduced expression of proliferation and migration genes in HDLECs and reduced proliferation and migration in vitro. We also presented evidence that Zmiz1 regulates Prox1 expression in vitro and in vivo by modulating chromatin accessibility at Prox1 regulatory regions. Furthermore, we observed that loss of Zmiz1 in mesenteric lymphatic vessels significantly reduced valve density. Collectively, our results highlight a novel role of Zmiz1 in LECs and as a transcriptional regulator of Prox1, shedding light on a previously unknown regulatory factor in lymphatic vascular biology.


Assuntos
Proliferação de Células , Células Endoteliais , Proteínas de Homeodomínio , Vasos Linfáticos , Fatores de Transcrição , Proteínas Supressoras de Tumor , Animais , Humanos , Camundongos , Movimento Celular/genética , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Linfangiogênese/genética , Vasos Linfáticos/metabolismo , Vasos Linfáticos/citologia , Camundongos Knockout , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
3.
Dev Cell ; 59(9): 1159-1174.e5, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38537630

RESUMO

Inside the finger-like intestinal projections called villi, strands of smooth muscle cells contract to propel absorbed dietary fats through the adjacent lymphatic capillary, the lacteal, sending fats into the systemic blood circulation for energy production. Despite this vital function, mechanisms of formation, assembly alongside lacteals, and maintenance of villus smooth muscle are unknown. By combining single-cell RNA sequencing and quantitative lineage tracing of the mouse intestine, we identified a local hierarchy of subepithelial fibroblast progenitors that differentiate into mature smooth muscle fibers via intermediate contractile myofibroblasts. This continuum persists as the major mechanism for villus musculature renewal throughout adult life. The NOTCH3-DLL4 signaling axis governs the assembly of smooth muscle fibers alongside their adjacent lacteals and is required for fat absorption. Our studies identify the ontogeny and maintenance of a poorly defined class of intestinal smooth muscle, with implications for accelerated repair and recovery of digestive function following injury.


Assuntos
Diferenciação Celular , Miofibroblastos , Animais , Miofibroblastos/metabolismo , Miofibroblastos/citologia , Camundongos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/citologia , Transdução de Sinais , Vasos Linfáticos/metabolismo , Vasos Linfáticos/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/citologia , Intestinos/citologia , Músculo Liso/metabolismo , Músculo Liso/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Receptor Notch3/metabolismo , Receptor Notch3/genética , Camundongos Endogâmicos C57BL
4.
J Virol ; 97(11): e0048023, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37877715

RESUMO

IMPORTANCE: Viruses are able to mimic the physiological or pathological mechanism of the host to favor their infection and replication. Virus-mock basement membrane (VMBM) is a Megalocytivirus-induced extracellular structure formed on the surface of infected cells and structurally and functionally mimics the basement membrane of the host. VMBM provides specific support for lymphatic endothelial cells (LECs) rather than blood endothelial cells to adhere to the surface of infected cells, which constitutes a unique phenomenon of Megalocytivirus infection. Here, the structure of VMBM and the interactions between VMBM components and LECs have been analyzed at the molecular level. The regulatory effect of VMBM components on the proliferation and migration of LECs has also been explored. This study helps to understand the mechanism of LEC-specific attachment to VMBM and to address the issue of where the LECs come from in the context of Megalocytivirus infection.


Assuntos
Membrana Basal , Células Endoteliais , Iridoviridae , Vasos Linfáticos , Membrana Basal/metabolismo , Membrana Basal/virologia , Células Endoteliais/citologia , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Iridoviridae/fisiologia , Vasos Linfáticos/citologia , Proliferação de Células , Movimento Celular , Vasos Sanguíneos/citologia , Interações entre Hospedeiro e Microrganismos
5.
J Biol Chem ; 299(6): 104775, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37142226

RESUMO

The vascular and lymphatic systems both comprise a series of structurally distinct vessels lined with an inner layer of endothelial cells that function to provide a semipermeable barrier to blood and lymph. Regulation of the endothelial barrier is critical for maintaining vascular and lymphatic barrier homeostasis. One of the regulators of endothelial barrier function and integrity is sphingosine-1-phosphate (S1P), a bioactive sphingolipid metabolite secreted into the blood by erythrocytes, platelets, and endothelial cells and into the lymph by lymph endothelial cells. Binding of S1P to its G protein-coupled receptors, known as S1PR1-5, regulates its pleiotropic functions. This review outlines the structural and functional differences between vascular and lymphatic endothelium and describes current understanding of the importance of S1P/S1PR signaling in regulation of barrier functions. Most studies thus far have been primarily focused on the role of the S1P/S1PR1 axis in vasculature and have been summarized in several excellent reviews, and thus, we will only discuss new perspectives on the molecular mechanisms of action of S1P and its receptors. Much less is known about the responses of the lymphatic endothelium to S1P and the functions of S1PRs in lymph endothelial cells, and this is the major focus of this review. We also discuss current knowledge related to signaling pathways and factors regulated by the S1P/S1PR axis that control lymphatic endothelial cell junctional integrity. Gaps and limitations in current knowledge are highlighted together with the need to further understand the role of S1P receptors in the lymphatic system.


Assuntos
Endotélio Vascular , Vasos Linfáticos , Lisofosfolipídeos , Receptores de Lisoesfingolipídeo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Lisofosfolipídeos/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Humanos , Animais , Junções Intercelulares , Transdução de Sinais , Vasos Linfáticos/citologia , Vasos Linfáticos/metabolismo
6.
Nature ; 614(7947): 343-348, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36697821

RESUMO

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.


Assuntos
Células Endoteliais , Elementos Facilitadores Genéticos , Hematopoese , Vasos Linfáticos , Animais , Camundongos , Células Endoteliais/metabolismo , Elementos Facilitadores Genéticos/genética , Hematopoese/genética , Proteínas de Homeodomínio/metabolismo , Vasos Linfáticos/citologia , Vasos Linfáticos/metabolismo , Fatores de Transcrição/metabolismo
7.
Nature ; 606(7914): 570-575, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35614218

RESUMO

The lineage and developmental trajectory of a cell are key determinants of cellular identity. In the vascular system, endothelial cells (ECs) of blood and lymphatic vessels differentiate and specialize to cater to the unique physiological demands of each organ1,2. Although lymphatic vessels were shown to derive from multiple cellular origins, lymphatic ECs (LECs) are not known to generate other cell types3,4. Here we use recurrent imaging and lineage-tracing of ECs in zebrafish anal fins, from early development to adulthood, to uncover a mechanism of specialized blood vessel formation through the transdifferentiation of LECs. Moreover, we demonstrate that deriving anal-fin vessels from lymphatic versus blood ECs results in functional differences in the adult organism, uncovering a link between cell ontogeny and functionality. We further use single-cell RNA-sequencing analysis to characterize the different cellular populations and transition states involved in the transdifferentiation process. Finally, we show that, similar to normal development, the vasculature is rederived from lymphatics during anal-fin regeneration, demonstrating that LECs in adult fish retain both potency and plasticity for generating blood ECs. Overall, our research highlights an innate mechanism of blood vessel formation through LEC transdifferentiation, and provides in vivo evidence for a link between cell ontogeny and functionality in ECs.


Assuntos
Vasos Sanguíneos , Transdiferenciação Celular , Vasos Linfáticos , Nadadeiras de Animais/citologia , Animais , Vasos Sanguíneos/citologia , Linhagem da Célula , Células Endoteliais/citologia , Vasos Linfáticos/citologia , Peixe-Zebra
8.
PLoS One ; 16(9): e0249256, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34543279

RESUMO

Lymphatic vessels are indispensable for tissue fluid homeostasis, transport of solutes and dietary lipids and immune cell trafficking. In contrast to blood vessels, which are easily visible by their erythrocyte cargo, lymphatic vessels are not readily detected in the tissue context. Their invisibility interferes with the analysis of the three-dimensional lymph vessel structure in large tissue volumes and hampers dynamic intravital studies on lymphatic function and pathofunction. An approach to overcome these limitations are mouse models, which express transgenic fluorescent proteins under the control of tissue-specific promotor elements. We introduce here the BAC-transgenic mouse reporter strain Vegfr3-tdTomato that expresses a membrane-tagged version of tdTomato under control of Flt4 regulatory elements. Vegfr3-tdTomato mice inherited the reporter in a mendelian fashion and showed selective and stable fluorescence in the lymphatic vessels of multiple organs tested, including lung, kidney, heart, diaphragm, intestine, mesentery, liver and dermis. In this model, tdTomato expression was sufficient for direct visualisation of lymphatic vessels by epifluorescence microscopy. Furthermore, lymph vessels were readily visualized using a number of microscopic modalities including confocal laser scanning, light sheet fluorescence and two-photon microscopy. Due to the early onset of VEGFR-3 expression in venous embryonic vessels and the short maturation time of tdTomato, this reporter offers an interesting alternative to Prox1-promoter driven lymphatic reporter mice for instance to study the developmental differentiation of venous to lymphatic endothelial cells.


Assuntos
Proteínas Luminescentes/genética , Vasos Linfáticos/citologia , Camundongos Transgênicos , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Células Endoteliais , Genes Reporter , Proteínas Luminescentes/metabolismo , Vasos Linfáticos/fisiologia , Microscopia Confocal , Microscopia de Fluorescência/métodos , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteína Vermelha Fluorescente
9.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34326257

RESUMO

The lymphatic system is involved in various biological processes, including fluid transport from the interstitium into the venous circulation, lipid absorption, and immune cell trafficking. Despite its critical role in homeostasis, lymphangiogenesis (lymphatic vessel formation) is less widely studied than its counterpart, angiogenesis (blood vessel formation). Although the incorporation of lymphatic vasculature in engineered tissues or organoids would enable more precise mimicry of native tissue, few studies have focused on creating engineered tissues containing lymphatic vessels. Here, we populated thick collagen sheets with human lymphatic endothelial cells, combined with supporting cells and blood endothelial cells, and examined lymphangiogenesis within the resulting constructs. Our model required just a few days to develop a functional lymphatic vessel network, in contrast to other reported models requiring several weeks. Coculture of lymphatic endothelial cells with the appropriate supporting cells and intact PDGFR-ß signaling proved essential for the lymphangiogenesis process. Additionally, subjecting the constructs to cyclic stretch enabled the creation of complex muscle tissue aligned with the lymphatic and blood vessel networks, more precisely biomimicking native tissue. Interestingly, the response of developing lymphatic vessels to tensile forces was different from that of blood vessels; while blood vessels oriented perpendicularly to the stretch direction, lymphatic vessels mostly oriented in parallel to the stretch direction. Implantation of the engineered lymphatic constructs into a mouse abdominal wall muscle resulted in anastomosis between host and implant lymphatic vasculatures, demonstrating the engineered construct's potential functionality in vivo. Overall, this model provides a potential platform for investigating lymphangiogenesis and lymphatic disease mechanisms.


Assuntos
Polpa Dentária/fisiologia , Células Endoteliais/fisiologia , Linfangiogênese/fisiologia , Vasos Linfáticos/fisiologia , Engenharia Tecidual , Técnicas de Cocultura , Humanos , Vasos Linfáticos/citologia , Neovascularização Fisiológica , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Células-Tronco/fisiologia
10.
Cell Rep ; 35(11): 109255, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34133928

RESUMO

The formation of new vessels requires a tight synchronization between proliferation, differentiation, and sprouting. However, how these processes are differentially activated, often by neighboring endothelial cells (ECs), remains unclear. Here, we identify cell cycle progression as a regulator of EC sprouting and differentiation. Using transgenic zebrafish illuminating cell cycle stages, we show that venous and lymphatic precursors sprout from the cardinal vein exclusively in G1 and reveal that cell-cycle arrest is induced in these ECs by overexpression of p53 and the cyclin-dependent kinase (CDK) inhibitors p27 and p21. We further demonstrate that, in vivo, forcing G1 cell-cycle arrest results in enhanced vascular sprouting. Mechanistically, we identify the mitogenic VEGFC/VEGFR3/ERK axis as a direct inducer of cell-cycle arrest in ECs and characterize the cascade of events that render "sprouting-competent" ECs. Overall, our results uncover a mechanism whereby mitogen-controlled cell-cycle arrest boosts sprouting, raising important questions about the use of cell cycle inhibitors in pathological angiogenesis and lymphangiogenesis.


Assuntos
Pontos de Checagem do Ciclo Celular , Células Endoteliais , Vasos Linfáticos , Neovascularização Fisiológica , Fator C de Crescimento do Endotélio Vascular , Veias , Proteínas de Peixe-Zebra , Animais , Animais Geneticamente Modificados , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Diferenciação Celular , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fase G1 , Vasos Linfáticos/citologia , Sistema de Sinalização das MAP Quinases , Neovascularização Fisiológica/efeitos dos fármacos , Roscovitina/farmacologia , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Veias/citologia , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
11.
Development ; 148(11)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34080610

RESUMO

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.


Assuntos
Sistema Cardiovascular/crescimento & desenvolvimento , Linfangiogênese/fisiologia , Vasos Linfáticos/fisiologia , Animais , Sistema Cardiovascular/citologia , Sistema Cardiovascular/embriologia , Células Endoteliais/fisiologia , Homeostase , Humanos , Vasos Linfáticos/citologia , Vasos Linfáticos/embriologia , Transdução de Sinais
12.
Cell Metab ; 33(7): 1449-1465.e6, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34038712

RESUMO

The lymphatic vasculature plays important roles in the physiology of the organs in which it resides, though a clear mechanistic understanding of how this crosstalk is mediated is lacking. Here, we performed single-cell transcriptional profiling of human and mouse adipose tissue and found that lymphatic endothelial cells highly express neurotensin (NTS/Nts). Nts expression is reduced by cold and norepinephrine in an α-adrenergic-dependent manner, suggesting a role in adipose thermogenesis. Indeed, NTS treatment of brown adipose tissue explants reduced expression of thermogenic genes. Furthermore, adenoviral-mediated overexpression and knockdown or knockout of NTS in vivo reduced and enhanced cold tolerance, respectively, an effect that is mediated by NTSR2 and ERK signaling. Inhibition of NTSR2 promoted energy expenditure and improved metabolic function in obese mice. These data establish a link between adipose tissue lymphatics and adipocytes with potential therapeutic implications.


Assuntos
Células Endoteliais/metabolismo , Vasos Linfáticos/citologia , Neurotensina/fisiologia , Termogênese , Animais , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Vasos Linfáticos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Neurotensina/genética , Neurotensina/metabolismo , Neurotensina/farmacologia , Transdução de Sinais/genética , Termogênese/efeitos dos fármacos , Termogênese/genética
13.
Cancer Immunol Res ; 9(7): 748-764, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33952631

RESUMO

Several solid malignancies trigger lymphangiogenesis, facilitating metastasis. Tumor-associated lymphatic vessels significantly contribute to the generation of an immunosuppressive tumor microenvironment (TME). Here, we have investigated the ability of tumoral lymphatic endothelial cells (LEC) to function as MHC class II-restricted antigen-presenting cells in the regulation of antitumor immunity. Using murine models of lymphangiogenic tumors engrafted under the skin, we have shown that tumoral LECs upregulate MHC class II and the MHC class II antigen-processing machinery, and that they promote regulatory T-cell (Treg) expansion ex vivo. In mice with LEC-restricted lack of MHC class II expression, tumor growth was severely impaired, whereas tumor-infiltrating effector T cells were increased. Reduction of tumor growth and reinvigoration of tumor-specific T-cell responses both resulted from alterations of the tumor-infiltrating Treg transcriptome and phenotype. Treg-suppressive functions were profoundly altered in tumors lacking MHC class II in LECs. No difference in effector T-cell responses or Treg phenotype and functions was observed in tumor-draining lymph nodes, indicating that MHC class II-restricted antigen presentation by LECs was required locally in the TME to confer potent suppressive functions to Tregs. Altogether, our study suggests that MHC class II-restricted antigen-presenting tumoral LECs function as a local brake, dampening T cell-mediated antitumor immunity and promoting intratumoral Treg-suppressive functions.


Assuntos
Células Endoteliais/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Evasão Tumoral , Animais , Apresentação de Antígeno , Comunicação Celular/imunologia , Técnicas de Cocultura , Modelos Animais de Doenças , Células Endoteliais/imunologia , Feminino , Humanos , Linfonodos/citologia , Linfonodos/imunologia , Vasos Linfáticos/citologia , Vasos Linfáticos/imunologia , Camundongos , Cultura Primária de Células , Células Tumorais Cultivadas , Microambiente Tumoral/imunologia
14.
Int J Mol Sci ; 22(8)2021 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-33921847

RESUMO

The abnormal development or disruption of the lymphatic vasculature has been implicated in metabolic and hypertensive diseases. Recent evidence suggests that the offspring exposed to preeclampsia (PE) in utero are at higher risk of long-term health problems, such as cardiovascular and metabolic diseases in adulthood, owing to in utero fetal programming. We aimed to investigate lymphangiogenic activities in the lymphatic endothelial progenitor cells (LEPCs) of the offspring of PE. Human umbilical cord blood LEPCs from pregnant women with severe PE (n = 10) and gestationally matched normal pregnancies (n = 10) were purified with anti-vascular endothelial growth factor receptor 3 (VEGFR3)/podoplanin/CD11b microbeads using a magnetic cell sorter device. LEPCs from PE displayed significantly delayed differentiation and reduced formation of lymphatic endothelial cell (LEC) colonies compared with the LEPCs from normal pregnancies. LECs differentiated from PE-derived LEPCs exhibited decreased tube formation, migration, proliferation, adhesion, wound healing, and 3D-sprouting activities as well as increased lymphatic permeability through the disorganization of VE-cadherin junctions, compared with the normal pregnancy-derived LECs. In vivo, LEPCs from PE showed significantly reduced lymphatic vessel formation compared to the LEPCs of the normal pregnancy. Gene expression analysis revealed that compared to the normal pregnancy-derived LECs, the PE-derived LECs showed a significant decrease in the expression of pro-lymphangiogenic genes (GREM1, EPHB3, VEGFA, AMOT, THSD7A, ANGPTL4, SEMA5A, FGF2, and GBX2). Collectively, our findings demonstrate, for the first time, that LEPCs from PE have reduced lymphangiogenic activities in vitro and in vivo and show the decreased expression of pro-lymphangiogenic genes. This study opens a new avenue for investigation of the molecular mechanism of LEPC differentiation and lymphangiogenesis in the offspring of PE and subsequently may impact the treatment of long-term health problems such as cardiovascular and metabolic disorders of offspring with abnormal development of lymphatic vasculature.


Assuntos
Antígeno CD11b/metabolismo , Células Progenitoras Endoteliais/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto , Animais , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Feminino , Humanos , Vasos Linfáticos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pré-Eclâmpsia/metabolismo , Gravidez , Cicatrização/fisiologia
15.
PLoS One ; 16(4): e0249068, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33857173

RESUMO

Atypical chemokine receptor ACKR3 (formerly CXCR7) is a scavenging receptor that has recently been implicated in murine lymphatic development. Specifically, ACKR3-deficiency was shown to result in lymphatic hyperplasia and lymphedema, in addition to cardiac hyperplasia and cardiac valve defects leading to embryonic lethality. The lymphatic phenotype was attributed to a lymphatic endothelial cell (LEC)-intrinsic scavenging function of ACKR3 for the vascular peptide hormone adrenomedullin (AM), which is also important during postnatal lymphangiogenesis. In this study, we investigated the expression of ACKR3 in the lymphatic vasculature of adult mice and its function in postnatal lymphatic development and function. We show that ACKR3 is widely expressed in mature lymphatics and that it exerts chemokine-scavenging activity in cultured murine skin-derived LECs. To investigate the role of LEC-expressed ACKR3 in postnatal lymphangiogenesis and function during adulthood, we generated and validated a lymphatic-specific, inducible ACKR3 knockout mouse. Surprisingly, in contrast to the reported involvement of ACKR3 in lymphatic development, our analyses revealed no contribution of LEC-expressed ACKR3 to postnatal lymphangiogenesis, lymphatic morphology and drainage function.


Assuntos
Células Endoteliais/metabolismo , Linfangiogênese , Vasos Linfáticos/citologia , Receptores CXCR/metabolismo , Animais , Vasos Linfáticos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores CXCR/genética
16.
Cells ; 10(1)2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33467729

RESUMO

Junctional adhesion proteins play important roles in controlling angiogenesis, vascular permeability and leukocyte trafficking. CD112 (nectin-2) belongs to the immunoglobulin superfamily and was shown to engage in homophilic and heterophilic interactions with a variety of binding partners expressed on endothelial cells and on leukocytes. Recent in vitro studies suggested that CD112 regulates human endothelial cell migration and proliferation as well as transendothelial migration of leukocytes. However, so far, the role of CD112 in endothelial cell biology and in leukocyte trafficking has not been elucidated in vivo. We found CD112 to be expressed by lymphatic and blood endothelial cells in different murine tissues. In CD112-deficient mice, the blood vessel coverage in the retina and spleen was significantly enhanced. In functional in vitro studies, a blockade of CD112 modulated endothelial cell migration and significantly enhanced endothelial tube formation. An antibody-based blockade of CD112 also significantly reduced T cell transmigration across endothelial monolayers in vitro. Moreover, T cell homing to the spleen was significantly reduced in CD112-deficient mice. Overall, our results identify CD112 as a regulator of angiogenic processes in vivo and demonstrate a novel role for CD112 in T cell entry into the spleen.


Assuntos
Nectinas/metabolismo , Neovascularização Patológica , Baço/metabolismo , Linfócitos T/metabolismo , Animais , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Permeabilidade Capilar , Movimento Celular , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Leucócitos/citologia , Vasos Linfáticos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Neutrófilos/metabolismo , Permeabilidade , Ligação Proteica , Linfócitos T/citologia , Internalização do Vírus
17.
Angiogenesis ; 24(1): 67-82, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32918672

RESUMO

Lymphatic and blood vascular endothelial cells (ECs) share several molecular and developmental features. However, these two cell types possess distinct phenotypic signatures, reflecting their different biological functions. Despite significant advances in elucidating how the specification of lymphatic and blood vascular ECs is regulated at the transcriptional level during development, the key molecular mechanisms governing their lineage identity under physiological or pathological conditions remain poorly understood. To explore the epigenomic signatures in the maintenance of EC lineage specificity, we compared the transcriptomic landscapes, histone composition (H3K4me3 and H3K27me3) and DNA methylomes of cultured matched human primary dermal lymphatic and blood vascular ECs. Our findings reveal that blood vascular lineage genes manifest a more 'repressed' histone composition in lymphatic ECs, whereas DNA methylation at promoters is less linked to the differential transcriptomes of lymphatic versus blood vascular ECs. Meta-analyses identified two transcriptional regulators, BCL6 and MEF2C, which potentially govern endothelial lineage specificity. Notably, the blood vascular endothelial lineage markers CD34, ESAM and FLT1 and the lymphatic endothelial lineage markers PROX1, PDPN and FLT4 exhibited highly differential epigenetic profiles and responded in distinct manners to epigenetic drug treatments. The perturbation of histone and DNA methylation selectively promoted the expression of blood vascular endothelial markers in lymphatic endothelial cells, but not vice versa. Overall, our study reveals that the fine regulation of lymphatic and blood vascular endothelial transcriptomes is maintained via several epigenetic mechanisms, which are crucial to the maintenance of endothelial cell identity.


Assuntos
Células Sanguíneas/citologia , Linhagem da Célula/genética , Derme/citologia , Células Endoteliais/citologia , Epigênese Genética , Vasos Linfáticos/citologia , Sequência de Bases , Biomarcadores/metabolismo , Metilação de DNA/genética , Histonas/metabolismo , Humanos , Fatores de Transcrição MEF2/metabolismo , Motivos de Nucleotídeos/genética , Regiões Promotoras Genéticas , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Transcriptoma/genética
18.
Methods Mol Biol ; 2230: 151-165, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33197014

RESUMO

Renal capsule transplantation is a very helpful method to grow embryonic tissues or tumors in a vascular environment, allowing for long-term engraftment and biological analyses. This chapter describes the surgical procedure for the transplantation of embryonic skeletal elements in the renal capsule of adult mice and points out the manipulations that can be applied for assaying the role of angiogenesis during bone development and repair.


Assuntos
Desenvolvimento Ósseo/genética , Transplante de Rim/métodos , Morfogênese/genética , Neovascularização Fisiológica/genética , Túnica Adventícia/crescimento & desenvolvimento , Túnica Adventícia/patologia , Animais , Epitélio/crescimento & desenvolvimento , Epitélio/patologia , Humanos , Rim/crescimento & desenvolvimento , Rim/patologia , Linfangiogênese/genética , Vasos Linfáticos/citologia , Camundongos , Neovascularização Patológica/genética , Organogênese/genética
19.
Dev Dyn ; 250(5): 701-716, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33369805

RESUMO

BACKGROUND: In zebrafish, lymphatic endothelial cells (LECs) originate from multiple/several distinct progenitor populations and generate organ-specific lymphatic vasculatures. Cell fate and tissue specificities were determined using a combination of genetically engineered transgenic lines in which the promoter of a LEC-specific gene drives expression of a fluorescent reporter protein. RESULTS: We established a novel zebrafish transgenic line expressing eGFP under the control of part of the zebrafish batf3 promoter (Basic Leucine Zipper ATF-Like Transcription Factor 3). Spatiotemporal examination of Tg(batf3MIN:eGFP) transgenic fish revealed a typical lymphatic expression pattern, which does not perfectly recapitulate the expression pattern of existing LEC transgenic lines. eGFP+ cells constitute a heterogeneous endothelial cell population, which expressed LEC and/or blood endothelial cells (BEC) markers in different tissues. In addition, we characterize the renal eGFP+ cell as a population of interest to study kidney diseases and regeneration. CONCLUSION: Our Tg(batf3MIN:eGFP) reporter zebrafish line provides a useful system to study LEC populations, of which heterogeneity depends on origin of progenitors, tissue environment and physiological conditions. We further developed a novel fish-adapted tissue clearing method, which allows deep imaging and 3D-visualization of vascular and lymphatic networks in the whole organism.


Assuntos
Células Endoteliais , Genes Reporter , Vasos Linfáticos/citologia , Peixe-Zebra , Animais , Animais Geneticamente Modificados
20.
Cell Mol Gastroenterol Hepatol ; 11(2): 573-595, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32961356

RESUMO

BACKGROUND AND AIMS: As the incidence of nonalcoholic steatohepatitis (NASH) continues to rise, understanding how normal liver functions are affected during disease is required before developing novel therapeutics which could reduce morbidity and mortality. However, very little is understood about how the transport of proteins and cells from the liver by the lymphatic vasculature is affected by inflammatory mediators or during disease. METHODS: To answer these questions, we utilized a well-validated mouse model of NASH and exposure to highly oxidized low density lipoprotein (oxLDL). In addition to single cell sequencing, multiplexed immunofluorescence and metabolomic analysis of liver lymphatic endothelial cells (LEC)s we evaluated lymphatic permeability and transport both in vitro and in vivo. RESULTS: Confirming similarities between human and mouse liver lymphatic vasculature in NASH, we found that the lymphatic vasculature expands as disease progresses and results in the downregulation of genes important to lymphatic identity and function. We also demonstrate, in mice with NASH, that fluorescein isothiocyanate (FITC) dextran does not accumulate in the liver draining lymph node upon intrahepatic injection, a defect that was rescued with therapeutic administration of the lymphatic growth factor, recombinant vascular endothelial growth factor C (rVEGFC). Similarly, exposure to oxLDL reduced the amount of FITC-dextran in the portal draining lymph node and through an LEC monolayer. We provide evidence that the mechanism by which oxLDL impacts lymphatic permeability is via a reduction in Prox1 expression which decreases lymphatic specific gene expression, impedes LEC metabolism and reorganizes the highly permeable lymphatic cell-cell junctions which are a defining feature of lymphatic capillaries. CONCLUSIONS: We identify oxLDL as a major contributor to decreased lymphatic permeability in the liver, a change which is consistent with decreased protein homeostasis and increased inflammation during chronic liver disease.


Assuntos
Lipoproteínas LDL/metabolismo , Fígado/patologia , Vasos Linfáticos/patologia , Hepatopatia Gordurosa não Alcoólica/imunologia , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Proteínas de Homeodomínio/metabolismo , Humanos , Junções Intercelulares/patologia , Fígado/imunologia , Vasos Linfáticos/citologia , Vasos Linfáticos/imunologia , Masculino , Camundongos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Permeabilidade , Proteostase/genética , Proteostase/imunologia , RNA-Seq , Análise de Célula Única , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...