Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.835
Filtrar
1.
Cell Signal ; 123: 111359, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39179089

RESUMO

The placenta is essential organ for oxygen and nutrient exchange between the mother and the developing fetus. Trophoblast lineage differentiation is closely related to the normal function of the placenta. Trophoblast stem cells (TSCs) can differentiate into all placental trophoblast subtypes and are widely used as in vitro stem cell models to study placental development and trophoblast lineage differentiation. Although extensive research has been conducted on the differentiation of TSCs, the possible parallels between trophoblast giant cells (TGCs) that are differentiated from TSCs in vitro and the various subtypes of TGC lineages in vivo are still poorly understood. In this study, mouse TSCs (mTSCs) were induced to differentiate into TGCs, and our mRNA sequencing (RNA-seq) data revealed that mTSCs and TGCs have distinct transcriptional signatures. We conducted a comparison of mTSCs and TGCs transcriptomes with the published transcriptomes of TGC lineages in murine placenta detected by single-cell RNA-seq and found that mTSCs tend to differentiate into maternal blood vessel-associated TGCs in vitro. Moreover, we identified the transcription factor (TF) ZMAT1, which may be responsible for the differentiation of mTSCs into sinusoid TGCs, and the TFs EGR1 and MITF, which are likely involved in the differentiation of mTSCs into spiral artery-associated TGCs. Thus, our findings provide a valuable resource for the mechanisms of trophoblast lineage differentiation and placental deficiency-associated diseases development.


Assuntos
Vasos Sanguíneos , Células-Tronco , Fatores de Transcrição , Transcriptoma , Trofoblastos , Feminino , Masculino , Camundongos , Gravidez , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Diferenciação Celular , Linhagem da Célula , Troca Materno-Fetal , Camundongos Endogâmicos C57BL , Placenta/citologia , Análise da Expressão Gênica de Célula Única , Células-Tronco/citologia , Fatores de Transcrição/metabolismo , Trofoblastos/citologia , Animais
2.
Adv Mater ; 36(36): e2401528, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39092638

RESUMO

Printing human tissues and organs replete with biomimetic vascular networks is of growing interest. While it is possible to embed perfusable channels within acellular and densely cellular matrices, they do not currently possess the biomimetic architectures found in native vessels. Here, coaxial sacrificial writing into functional tissues (co-SWIFT) is developed, an embedded bioprinting method capable of generating hierarchically branching, multilayered vascular networks within both granular hydrogel and densely cellular matrices. Coaxial printheads are designed with an extended core-shell configuration to facilitate robust core-core and shell-shell interconnections between printed branching vessels during embedded bioprinting. Using optimized core-shell ink combinations, biomimetic vessels composed of a smooth muscle cell-laden shell that surrounds perfusable lumens are coaxially printed into granular matrices composed of: 1) transparent alginate microparticles, 2) sacrificial microparticle-laden collagen, or 3) cardiac spheroids derived from human induced pluripotent stem cells. Biomimetic blood vessels that exhibit good barrier function are produced by seeding these interconnected lumens with a confluent layer of endothelial cells. Importantly, it is found that co-SWIFT cardiac tissues mature under perfusion, beat synchronously, and exhibit a cardio-effective drug response in vitro. This advance opens new avenues for the scalable biomanufacturing of vascularized organ-specific tissues for drug testing, disease modeling, and therapeutic use.


Assuntos
Materiais Biomiméticos , Bioimpressão , Engenharia Tecidual , Humanos , Materiais Biomiméticos/química , Bioimpressão/métodos , Engenharia Tecidual/métodos , Alginatos/química , Células-Tronco Pluripotentes Induzidas/citologia , Hidrogéis/química , Alicerces Teciduais/química , Biomimética/métodos , Colágeno/química , Miócitos de Músculo Liso/citologia , Vasos Sanguíneos/citologia , Vasos Sanguíneos/fisiologia , Células Endoteliais da Veia Umbilical Humana , Animais , Esferoides Celulares/citologia
4.
Nat Commun ; 15(1): 7424, 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39198465

RESUMO

In fetal development, tissue interaction such as the interplay between blood vessel (BV) and epithelial tissue is crucial for organogenesis. Here we recapitulate the spatial arrangement between liver epithelial tissue and the portal vein to observe the formation of intrahepatic bile ducts (BDs) from human induced pluripotent stem cells (hiPSC). We co-culture hiPSC-liver progenitors on the artificial BV consisting of immature smooth muscle cells and endothelial cells, both derived from hiPSCs. After 3 weeks, liver progenitors within hiPSC-BV-incorporated liver organoids (BVLO) differentiate to cholangiocytes and acquire epithelial characteristics, including intercellular junctions, microvilli on the apical membrane, and secretory functions. Furthermore, liver surface transplanted-BVLO temporarily attenuates cholestatic injury symptoms. Single cell RNA sequence analysis suggests that BD interact with the BV in BVLO through TGFß and Notch pathways. Knocking out JAG1 in hiPSC-BV significantly attenuates bile duct formation, highlighting BVLO potential as a model for Alagille syndrome, a congenital biliary disease. Overall, we develop a novel 3D co-culture method that successfully establishes functional human BDs by emulating liver epithelial-BV interaction.


Assuntos
Diferenciação Celular , Técnicas de Cocultura , Células-Tronco Pluripotentes Induzidas , Proteína Jagged-1 , Fígado , Organoides , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Organoides/metabolismo , Organoides/citologia , Fígado/citologia , Fígado/metabolismo , Fígado/irrigação sanguínea , Técnicas de Cocultura/métodos , Proteína Jagged-1/metabolismo , Proteína Jagged-1/genética , Síndrome de Alagille/genética , Síndrome de Alagille/metabolismo , Animais , Ductos Biliares Intra-Hepáticos/citologia , Ductos Biliares Intra-Hepáticos/metabolismo , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Camundongos , Receptores Notch/metabolismo , Receptores Notch/genética , Células Endoteliais/metabolismo , Células Endoteliais/citologia , Ductos Biliares/citologia , Ductos Biliares/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/citologia , Fator de Crescimento Transformador beta/metabolismo
5.
Dev Growth Differ ; 66(6): 357-368, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39072708

RESUMO

In recent decades, developmental biologists have come to view vascular development as a series of progressive transitions. Mesoderm differentiates into endothelial cells; arteries, veins and lymphatic endothelial cells are specified from early endothelial cells; and vascular networks diversify and invade developing tissues and organs. Our understanding of this elaborate developmental process has benefitted from detailed studies using the zebrafish as a model system. Here, we review a number of key developmental transitions that occur in zebrafish during the formation of the blood and lymphatic vessel networks.


Assuntos
Células Endoteliais , Peixe-Zebra , Animais , Peixe-Zebra/embriologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Vasos Linfáticos/embriologia , Vasos Linfáticos/metabolismo , Vasos Linfáticos/citologia , Vasos Sanguíneos/citologia , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/crescimento & desenvolvimento , Neovascularização Fisiológica/fisiologia , Diferenciação Celular
6.
Stem Cell Res Ther ; 15(1): 205, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38982541

RESUMO

Vascular tissue engineering is a promising approach for regenerating damaged blood vessels and developing new therapeutic approaches for heart disease treatment. To date, different sources of cells have been recognized that offer assistance within the recovery of heart supply routes and veins with distinctive capacities and are compelling for heart regeneration. However, some challenges still remain that need to be overcome to establish the full potential application of these cells. In this paper, we review the different cell sources used for vascular tissue engineering, focusing on extraembryonic tissue-derived cells (ESCs), and elucidate their roles in cardiovascular disease. In addition, we highlight the intricate interplay between mechanical and biochemical factors in regulating mesenchymal stem cell (MSC) differentiation, offering insights into optimizing their application in vascular tissues.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais , Regeneração , Engenharia Tecidual , Humanos , Engenharia Tecidual/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Regeneração/fisiologia , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/fisiologia , Vasos Sanguíneos/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Doenças Cardiovasculares/terapia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia
7.
Exp Cell Res ; 440(2): 114136, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38909881

RESUMO

Considering the importance of alternative methodologies to animal experimentation, we propose an organoid-based biological model for in vitro blood vessel generation, achieved through co-culturing endothelial and vascular smooth muscle cells (VSMCs). Initially, the organoids underwent comprehensive characterization, revealing VSMCs (α-SMA + cells) at the periphery and endothelial cells (CD31+ cells) at the core. Additionally, ephrin B2 and ephrin B4, genes implicated in arterial and venous formation respectively, were used to validate the obtained organoid. Moreover, the data indicates exclusive HIF-1α expression in VSMCs, identified through various methodologies. Subsequently, we tested the hypothesis that the generated blood vessels have the capacity to modulate the osteogenic phenotype, demonstrating the ability of HIF-1α to promote osteogenic signals, primarily by influencing Runx2 expression. Overall, this study underscores that the methodology employed to create blood vessel organoids establishes an experimental framework capable of producing a 3D culture model of both venous and arterial endothelial tissues. This model effectively guides morphogenesis from mesenchymal stem cells through paracrine signaling, ultimately leading to an osteogenic acquisition phenotype, with the dynamic involvement of HIF-1α.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia , Músculo Liso Vascular , Miócitos de Músculo Liso , Organoides , Osteogênese , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Osteogênese/genética , Organoides/metabolismo , Organoides/citologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/citologia , Células Cultivadas , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/citologia , Vasos Sanguíneos/crescimento & desenvolvimento , Técnicas de Cocultura/métodos , Diferenciação Celular , Células Endoteliais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia
8.
Talanta ; 277: 126415, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38878513

RESUMO

Endothelial cells (ECs) migration is a crucial early step in vascular repair and tissue neovascularization. While extensive research has elucidated the biochemical drivers of endothelial motility, the impact of biophysical cues, including vessel geometry and topography, remains unclear. Herein, we present a novel approach to reconstruct 3D self-assembly blood vessels-on-a-chip that accurately replicates real vessel geometry and topography, surpassing conventional 2D flat tube formation models. This vessels-on-a-chip system enables real-time monitoring of vasculogenesis and ECs migration at high spatiotemporal resolution. Our findings reveal that ECs exhibit increased migration speed and directionality in response to narrower vessel geometries, transitioning from a rounded to a polarized morphology. These observations underscore the critical influence of vessel size in regulating ECs migration and morphology. Overall, our study highlights the importance of biophysical factors in shaping ECs behavior, emphasizing the need to consider such factors in future studies of endothelial function and vessel biology.


Assuntos
Vasos Sanguíneos , Movimento Celular , Células Endoteliais da Veia Umbilical Humana , Humanos , Vasos Sanguíneos/citologia , Vasos Sanguíneos/fisiologia , Células Endoteliais/citologia , Dispositivos Lab-On-A-Chip , Neovascularização Fisiológica
10.
Circulation ; 150(6): 451-465, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38682338

RESUMO

BACKGROUND: Most organs are maintained lifelong by resident stem/progenitor cells. During development and regeneration, lineage-specific stem/progenitor cells can contribute to the growth or maintenance of different organs, whereas fully differentiated mature cells have less regenerative potential. However, it is unclear whether vascular endothelial cells (ECs) are also replenished by stem/progenitor cells with EC-repopulating potential residing in blood vessels. It has been reported recently that some EC populations possess higher clonal proliferative potential and vessel-forming capacity compared with mature ECs. Nevertheless, a marker to identify vascular clonal repopulating ECs (CRECs) in murine and human individuals is lacking, and, hence, the mechanism for the proliferative, self-renewal, and vessel-forming potential of CRECs is elusive. METHODS: We analyzed colony-forming, self-renewal, and vessel-forming potential of ABCG2 (ATP binding cassette subfamily G member 2)-expressing ECs in human umbilical vessels. To study the contribution of Abcg2-expressing ECs to vessel development and regeneration, we developed Abcg2CreErt2;ROSA TdTomato mice and performed lineage tracing during mouse development and during tissue regeneration after myocardial infarction injury. RNA sequencing and chromatin methylation chromatin immunoprecipitation followed by sequencing were conducted to study the gene regulation in Abcg2-expressing ECs. RESULTS: In human and mouse vessels, ECs with higher ABCG2 expression (ABCECs) possess higher clonal proliferative potential and in vivo vessel-forming potential compared with mature ECs. These cells could clonally contribute to vessel formation in primary and secondary recipients after transplantation. These features of ABCECs meet the criteria of CRECs. Results from lineage tracing experiments confirm that Abcg2-expressing CRECs (AbcCRECs) contribute to arteries, veins, and capillaries in cardiac tissue development and vascular tissue regeneration after myocardial infarction. Transcriptome and epigenetic analyses reveal that a gene expression signature involved in angiogenesis and vessel development is enriched in AbcCRECs. In addition, various angiogenic genes, such as Notch2 and Hey2, are bivalently modified by trimethylation at the 4th and 27th lysine residue of histone H3 (H3K4me3 and H3K27me3) in AbcCRECs. CONCLUSIONS: These results are the first to establish that a single prospective marker identifies CRECs in mice and human individuals, which holds promise to provide new cell therapies for repair of damaged vessels in patients with endothelial dysfunction.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Humanos , Camundongos , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/citologia , Neovascularização Fisiológica , Proliferação de Células , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/terapia , Regeneração , Células Endoteliais da Veia Umbilical Humana/metabolismo , Camundongos Transgênicos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/citologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Linhagem da Célula
11.
Nature ; 627(8005): 839-846, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38509363

RESUMO

The bone marrow adjusts blood cell production to meet physiological demands in response to insults. The spatial organization of normal and stress responses are unknown owing to the lack of methods to visualize most steps of blood production. Here we develop strategies to image multipotent haematopoiesis, erythropoiesis and lymphopoiesis in mice. We combine these with imaging of myelopoiesis1 to define the anatomy of normal and stress haematopoiesis. In the steady state, across the skeleton, single stem cells and multipotent progenitors distribute through the marrow enriched near megakaryocytes. Lineage-committed progenitors are recruited to blood vessels, where they contribute to lineage-specific microanatomical structures composed of progenitors and immature cells, which function as the production sites for each major blood lineage. This overall anatomy is resilient to insults, as it was maintained after haemorrhage, systemic bacterial infection and granulocyte colony-stimulating factor (G-CSF) treatment, and during ageing. Production sites enable haematopoietic plasticity as they differentially and selectively modulate their numbers and output in response to insults. We found that stress responses are variable across the skeleton: the tibia and the sternum respond in opposite ways to G-CSF, and the skull does not increase erythropoiesis after haemorrhage. Our studies enable in situ analyses of haematopoiesis, define the anatomy of normal and stress responses, identify discrete microanatomical production sites that confer plasticity to haematopoiesis, and uncover unprecedented heterogeneity of stress responses across the skeleton.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas , Estresse Fisiológico , Animais , Feminino , Masculino , Camundongos , Envelhecimento/fisiologia , Infecções Bacterianas/patologia , Infecções Bacterianas/fisiopatologia , Vasos Sanguíneos/citologia , Linhagem da Célula , Eritropoese , Fator Estimulador de Colônias de Granulócitos/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Hemorragia/patologia , Hemorragia/fisiopatologia , Linfopoese , Megacariócitos/citologia , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Mielopoese , Crânio/irrigação sanguínea , Crânio/patologia , Crânio/fisiopatologia , Esterno/irrigação sanguínea , Esterno/citologia , Esterno/metabolismo , Estresse Fisiológico/fisiologia , Tíbia/irrigação sanguínea , Tíbia/citologia , Tíbia/metabolismo
12.
Dev Dyn ; 253(5): 519-541, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38112237

RESUMO

BACKGROUND: Mural cells are an essential perivascular cell population that associate with blood vessels and contribute to vascular stabilization and tone. In the embryonic zebrafish vasculature, pdgfrb and tagln are commonly used as markers for identifying pericytes and vascular smooth muscle cells. However, the overlapping and distinct expression patterns of these markers in tandem have not been fully described. RESULTS: Here, we used the Tg(pdgfrb:Gal4FF; UAS:RFP) and Tg(tagln:NLS-EGFP) transgenic lines to identify single- and double-positive perivascular cell populations on the cranial, axial, and intersegmental vessels between 1 and 5 days postfertilization. From this comparative analysis, we discovered two novel regions of tagln-positive cell populations that have the potential to function as mural cell precursors. Specifically, we found that the hypochord-a reportedly transient structure-contributes to tagln-positive cells along the dorsal aorta. We also identified a unique mural cell progenitor population that resides along the midline between the neural tube and notochord and contributes to intersegmental vessel mural cell coverage. CONCLUSION: Together, our findings highlight the variability and versatility of tracking both pdgfrb and tagln expression in mural cells of the developing zebrafish embryo and reveal unexpected embryonic cell populations that express pdgfrb and tagln.


Assuntos
Animais Geneticamente Modificados , Pericitos , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/fisiologia , Pericitos/citologia , Pericitos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
13.
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
14.
Am J Physiol Renal Physiol ; 325(1): F22-F37, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37167273

RESUMO

Increased mechanical endothelial cell stretch contributes to the development of numerous cardiovascular and renal pathologies. Recent studies have shone a light on the importance of sex-dependent inflammation in the pathogenesis of renal disease states. The endothelium plays an intimate and critical role in the orchestration of immune cell activation through upregulation of adhesion molecules and secretion of cytokines and chemokines. While endothelial cells are not recognized as professional antigen-presenting cells, in response to cytokine stimulation, endothelial cells can express both major histocompatibility complex (MHC) I and MHC II. MHCs are essential to forming a part of the immunological synapse interface during antigen presentation to adaptive immune cells. Whether MHC I and II are increased under increased mechanical stretch is unknown. Due to hypertension being multifactorial, we hypothesized that increased mechanical endothelial stretch promotes the regulation of MHCs and key costimulatory proteins on mouse renal endothelial cells (MRECs) in a stretch-dependent manner. MRECs derived from both sexes underwent 5%, 10%, or 15% uniaxial cyclical stretch, and immunological synapse interface proteins were determined by immunofluorescence microscopy, immunoblot analysis, and RNA sequencing. We found that increased endothelial mechanical stretch conditions promoted downregulation of MHC I in male MRECs but upregulation in female MRECs. Moreover, MHC II was upregulated by mechanical stretch in both male and female MRECs, whereas CD86 and CD70 were regulated in a sex-dependent manner. By bulk RNA sequencing, we found that increased mechanical endothelial cell stretch promoted differential gene expression of key antigen processing and presentation genes in female MRECs, demonstrating that females have upregulation of key antigen presentation pathways. Taken together, our data demonstrate that mechanical endothelial stretch regulates endothelial activation and immunological synapse interface formation in renal endothelial cells in a sex-dependent manner.NEW & NOTEWORTHY Endothelial cells contribute to the development of renal inflammation and have the unique ability to express antigen presentation proteins. Whether increased endothelial mechanical stretch regulates immunological synapse interface proteins remains unknown. We found that antigen presentation proteins and costimulatory proteins on renal endothelial cells are modulated by mechanical stretch in a sex-dependent manner. Our data provide novel insights into the sex-dependent ability of renal endothelial cells to present antigens in response to endothelial mechanical stimuli.


Assuntos
Vasos Sanguíneos , Células Endoteliais , Sinapses Imunológicas , Rim , Células Endoteliais/fisiologia , Células Cultivadas , Masculino , Feminino , Animais , Camundongos , Rim/irrigação sanguínea , Camundongos Endogâmicos C57BL , Vasos Sanguíneos/citologia , Fenômenos Biomecânicos , Inflamação/metabolismo , Secretoma/metabolismo , Caracteres Sexuais , Complexo Principal de Histocompatibilidade , Antígeno B7-2/metabolismo , Apresentação de Antígeno
15.
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
16.
Nat Metab ; 4(1): 123-140, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35102339

RESUMO

Vascular mural cells (vMCs) play an essential role in the development and maturation of the vasculature by promoting vessel stabilization through their interactions with endothelial cells. Whether endothelial metabolism influences mural cell recruitment and differentiation is unknown. Here, we show that the oxidative pentose phosphate pathway (oxPPP) in endothelial cells is required for establishing vMC coverage of the dorsal aorta during early vertebrate development in zebrafish and mice. We demonstrate that laminar shear stress and blood flow maintain oxPPP activity, which in turn, promotes elastin expression in blood vessels through production of ribose-5-phosphate. Elastin is both necessary and sufficient to drive vMC recruitment and maintenance when the oxPPP is active. In summary, our work demonstrates that endothelial cell metabolism regulates blood vessel maturation by controlling vascular matrix composition and vMC recruitment.


Assuntos
Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Matriz Extracelular/metabolismo , Fosforilação Oxidativa , Via de Pentose Fosfato , Animais , Biomarcadores , Elastina/biossíntese , Elastina/genética , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Expressão Gênica , Genes Reporter , Glucose/metabolismo , Hemodinâmica , Camundongos , Camundongos Knockout , Modelos Biológicos , Estresse Oxidativo , Pentosefosfatos/metabolismo , Peixe-Zebra
17.
Science ; 375(6584): eabi7377, 2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35084939

RESUMO

Cerebrovascular diseases are a leading cause of death and neurologic disability. Further understanding of disease mechanisms and therapeutic strategies requires a deeper knowledge of cerebrovascular cells in humans. We profiled transcriptomes of 181,388 cells to define a cell atlas of the adult human cerebrovasculature, including endothelial cell molecular signatures with arteriovenous segmentation and expanded perivascular cell diversity. By leveraging this reference, we investigated cellular and molecular perturbations in brain arteriovenous malformations, which are a leading cause of stroke in young people, and identified pathologic endothelial transformations with abnormal vascular patterning and the ontology of vascularly derived inflammation. We illustrate the interplay between vascular and immune cells that contributes to brain hemorrhage and catalog opportunities for targeting angiogenic and inflammatory programs in vascular malformations.


Assuntos
Vasos Sanguíneos/citologia , Encéfalo/irrigação sanguínea , Malformações Arteriovenosas Intracranianas/patologia , Transcriptoma , Adulto , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiologia , Vasos Sanguíneos/fisiopatologia , Células Cultivadas , Córtex Cerebral/irrigação sanguínea , Hemorragia Cerebral/patologia , Hemorragia Cerebral/fisiopatologia , Circulação Cerebrovascular , Células Endoteliais/citologia , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos , Inflamação , Malformações Arteriovenosas Intracranianas/metabolismo , Monócitos/citologia , Monócitos/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiologia , Pericitos/citologia , Pericitos/fisiologia , RNA-Seq , Análise de Célula Única
18.
Biotechnol Bioeng ; 119(1): 118-133, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34617587

RESUMO

Three dimensional printable formulation of self-standing and vascular-supportive structures using multi-materials suitable for organ engineering is of great importance and highly challengeable, but, it could advance the 3D printing scenario from printable shape to functional unit of human body. In this study, the authors report a 3D printable formulation of such self-standing and vascular-supportive structures using an in-house formulated multi-material combination of albumen/alginate/gelatin-based hydrogel. The rheological properties and relaxation behavior of hydrogels were analyzed before the printing process. The suitability of the hydrogel in 3D printing of various customizable and self-standing structures, including a human ear model, was examined by extrusion-based 3D printing. The structural, mechanical, and physicochemical properties of the printed scaffolds were studied systematically. Results supported the 3D printability of the formulated hydrogel with self-standing structures, which are customizable to a specific need. In vitro cell experiment showed that the formulated hydrogel has excellent biocompatibility and vascular supportive behavior with the extent of endothelial sprout formation when tested with human umbilical vein endothelial cells. In conclusion, the present study demonstrated the suitability of the extrusion-based 3D printing technique for manufacturing complex shapes and structures using multi-materials with high fidelity, which have great potential in organ engineering.


Assuntos
Endotélio Vascular , Hidrogéis/química , Neovascularização Fisiológica , Impressão Tridimensional , Engenharia Tecidual/métodos , Animais , Vasos Sanguíneos/citologia , Vasos Sanguíneos/efeitos dos fármacos , Células Cultivadas , Orelha/irrigação sanguínea , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Alicerces Teciduais/química
19.
Diabetes ; 71(2): 321-328, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34753798

RESUMO

Adipose-derived stem cells (ADSCs) can differentiate into vascular lineages and participate in vascular remodeling. Perivascular ADSCs (PV-ADSCs) draw attention because of their unique location. The heterogeneity of subcutaneous (SUB) and abdominal ADSCs were well addressed, but PV-ADSCs' heterogeneity has not been investigated. In this study, we applied single-cell analysis to compare SUB-ADSCs and PV-ADSCs regarding their subpopulations, functions, and cell fates. We uncovered four subpopulations of PV-ADSCs (Dpp4+, Col4a2+/Icam1+, Clec11a+/Cpe+, and Sult1e1+ cells), among which the Clec11a+ subpopulation potentially participated in and regulated PV-ADSC differentiation toward a smooth muscle cell (SMC) phenotype. Distinct characteristics between PV-ADSCs and SUB-ADSCs were revealed.


Assuntos
Vasos Sanguíneos/citologia , Células-Tronco/fisiologia , Gordura Subcutânea/citologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/fisiologia , Análise de Célula Única , Células-Tronco/citologia
20.
Dev Cell ; 56(23): 3276-3287.e8, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34741805

RESUMO

The process of implantation and the cellular interactions at the embryo-maternal interface are intrinsically difficult to analyze, as the implanting embryo is concealed by the uterine tissues. Therefore, the mechanisms mediating the interconnection of the embryo and the mother are poorly understood. Here, we established a 3D biomimetic culture environment that harbors the key features of the murine implantation niche. This culture system enabled direct analysis of trophoblast invasion and revealed the first embryonic interactions with the maternal vasculature. We found that implantation is mediated by the collective migration of penetrating strands of trophoblast giant cells, which acquire the expression of vascular receptors, ligands, and adhesion molecules, assembling a network for communication with the maternal blood vessels. In particular, Pdgf signaling cues promote the establishment of the heterologous contacts. Together, the biomimetic platform and our findings thereof elucidate the hidden dynamics of the early interactions at the implantation site.


Assuntos
Blastocisto/metabolismo , Vasos Sanguíneos/metabolismo , Comunicação Celular , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Troca Materno-Fetal , Trofoblastos/metabolismo , Animais , Biomimética , Blastocisto/citologia , Vasos Sanguíneos/citologia , Técnicas de Cultura de Células , Movimento Celular , Implantação do Embrião , Embrião de Mamíferos/citologia , Feminino , Células Gigantes/citologia , Células Gigantes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Gravidez , Trofoblastos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA