Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Dev Cell ; 56(11): 1677-1693.e10, 2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34038707

RESUMO

Single-cell transcriptomics (scRNA-seq) has revolutionized the understanding of the spatial architecture of tissue structure and function. Advancing the "transcript-centric" view of scRNA-seq analyses is presently restricted by the limited resolution of proteomics and genome-wide techniques to analyze post-translational modifications. Here, by combining spatial cell sorting with transcriptomics and quantitative proteomics/phosphoproteomics, we established the spatially resolved proteome landscape of the liver endothelium, yielding deep mechanistic insight into zonated vascular signaling mechanisms. Phosphorylation of receptor tyrosine kinases was detected preferentially in the central vein area, resulting in an atypical enrichment of tyrosine phosphorylation. Prototypic biological validation identified Tie receptor signaling as a selective and specific regulator of vascular Wnt activity orchestrating angiocrine signaling, thereby controlling hepatocyte function during liver regeneration. Taken together, the study has yielded fundamental insight into the spatial organization of liver endothelial cell signaling. Spatial sorting may be employed as a universally adaptable strategy for multiomic analyses of scRNA-seq-defined cellular (sub)-populations.


Assuntos
Regeneração Hepática/genética , Fígado/crescimento & desenvolvimento , Fosfoproteínas/genética , Transcriptoma/genética , Células Endoteliais/metabolismo , Endotélio/crescimento & desenvolvimento , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/genética , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Fosforilação/genética , Proteômica/métodos , RNA-Seq , Regeneração/genética , Análise de Célula Única , Via de Sinalização Wnt/genética
2.
Clin Sci (Lond) ; 135(6): 829-846, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33720353

RESUMO

The endothelial-to-mesenchymal transition (EndMT) is a critical process that occurs during the development of the outflow tract (OFT). Malformations of the OFT can lead to the occurrence of conotruncal defect (CTD). SOX7 duplication has been reported in patients with congenital CTD, but its specific role in OFT development remains poorly understood. To decipher this, histological analysis showed that SRY-related HMG-box 7 (SOX7) was regionally expressed in the endocardial endothelial cells and in the mesenchymal cells of the OFT, where EndMT occurs. Experiments, using in vitro collagen gel culture system, revealed that SOX7 was a negative regulator of EndMT that inhibited endocardial cell (EC) migration and resulted in decreased number of mesenchymal cells. Forced expression of SOX7 in endothelial cells blocked further migration and improved the expression of the adhesion protein vascular endothelial (VE)-cadherin (VE-cadherin). Moreover, a VE-cadherin knockdown could partly reverse the SOX7-mediated repression of cell migration. Luciferase and electrophoretic mobility shift assay (EMSA) demonstrated that SOX7 up-regulated VE-cadherin by directly binding to the gene's promoter in endothelial cells. The coding exons and splicing regions of the SOX7 gene were also scanned in the 536 sporadic CTD patients and in 300 unaffected controls, which revealed four heterozygous SOX7 mutations. Luciferase assays revealed that two SOX7 variants weakened the transactivation of the VE-cadherin promoter. In conclusion, SOX7 inhibited EndMT during OFT development by directly up-regulating the endothelial-specific adhesion molecule VE-cadherin. SOX7 mutations can lead to impaired EndMT by regulating VE-cadherin, which may give rise to the molecular mechanisms associated with SOX7 in CTD pathogenesis.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Endocárdio/embriologia , Cardiopatias Congênitas/embriologia , Fatores de Transcrição SOXF/metabolismo , Animais , Antígenos CD/genética , Caderinas/genética , Movimento Celular , Embrião de Mamíferos , Endocárdio/citologia , Endotélio/crescimento & desenvolvimento , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Ratos , Fatores de Transcrição SOXF/genética
3.
Sci Rep ; 10(1): 11568, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32665552

RESUMO

Angiogenesis assays based on in vitro capillary-like growth of endothelial cells (EC) are widely used, either to evaluate the effect of anti- and pro-angiogenesis drugs of interest, or to test and compare the functional capacities of various types of EC and progenitor cells. Among the different methods applied to study angiogenesis, the most commonly used is the "Endothelial Tube Formation Assay" (ETFA). In suitable culture conditions, EC form two-dimensional (2D) branched structures that can lead to a meshed pseudo-capillary network. An alternative approach to ETFA is the "Fibrin Bead Assay" (FBA), based on the use of Cytodex 3 microspheres, which promote the growth of 3D capillary-like patterns from coated EC, suitable for high throughput in vitro angiogenesis studies. The analytical evaluation of these two widely used assays still remains challenging in terms of observation method and image analysis. We previously developed the "Angiogenesis Analyzer" for ImageJ (AA), a tool allowing analysis of ETFA-derived images, according to characteristics of the pseudo-capillary networks. In this work, we developed and implemented a new algorithm for AA able to recognize microspheres and to analyze the attached capillary-like structures from the FBA model. Such a method is presented for the first time in fully automated mode and using non-destructive image acquisition. We detailed these two algorithms and used the new AA version to compare both methods (i.e. ETFA and FBA) in their efficiency, accuracy and statistical relevance to model angiogenesis patterns of Human Umbilical Vein EC (HUVEC). Although the two methods do not assess the same biological step, our data suggest that they display specific and complementary information on the angiogenesis processes analysis.


Assuntos
Morfogênese/genética , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Fisiológica/genética , Fator A de Crescimento do Endotélio Vascular/genética , Endotélio/crescimento & desenvolvimento , Endotélio/metabolismo , Endotélio/patologia , Fibrina/química , Células Endoteliais da Veia Umbilical Humana , Humanos , Neovascularização Patológica/genética , Neovascularização Patológica/patologia
4.
Sci Rep ; 9(1): 15698, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31666588

RESUMO

Endothelial cells contain several nanoscale domains such as caveolae, fenestrations and transendothelial channels, which regulate signaling and transendothelial permeability. These structures can be covered by filter-like diaphragms. A transmembrane PLVAP (plasmalemma vesicle associated protein) protein has been shown to be necessary for the formation of diaphragms. The expression, subcellular localization and fenestra-forming role of PLVAP in liver sinusoidal endothelial cells (LSEC) have remained controversial. Here we show that fenestrations in LSEC contain PLVAP-diaphragms during the fetal angiogenesis, but they lose the diaphragms at birth. Although it is thought that PLVAP only localizes to diaphragms, we found luminal localization of PLVAP in adult LSEC using several imaging techniques. Plvap-deficient mice revealed that the absence of PLVAP and diaphragms did not affect the morphology, the number of fenestrations or the overall vascular architecture in the liver sinusoids. Nevertheless, PLVAP in fetal LSEC (fenestrations with diaphragms) associated with LYVE-1 (lymphatic vessel endothelial hyaluronan receptor 1), neuropilin-1 and VEGFR2 (vascular endothelial growth factor receptor 2), whereas in the adult LSEC (fenestrations without diaphragms) these complexes disappeared. Collectively, our data show that PLVAP can be expressed on endothelial cells without diaphragms, contradict the prevailing concept that biogenesis of fenestrae would be PLVAP-dependent, and reveal previously unknown PLVAP-dependent molecular complexes in LSEC during angiogenesis.


Assuntos
Diafragma/metabolismo , Endotélio/metabolismo , Fígado/metabolismo , Proteínas de Membrana/genética , Animais , Capilares/crescimento & desenvolvimento , Capilares/metabolismo , Capilares/ultraestrutura , Cavéolas/metabolismo , Cavéolas/ultraestrutura , Diafragma/crescimento & desenvolvimento , Diafragma/ultraestrutura , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Endotélio/crescimento & desenvolvimento , Endotélio/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Fígado/ultraestrutura , Proteínas de Membrana/metabolismo , Camundongos , Transdução de Sinais/genética
5.
Sci Rep ; 9(1): 8001, 2019 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-31142801

RESUMO

Generating new kidneys using tissue engineering technologies is an innovative strategy for overcoming the shortage of donor organs for transplantation. Here we report how to efficiently engineer the kidney vasculature of decellularized rat kidney scaffolds by using human induced pluripotent stem cell (hiPSCs)-derived endothelial cells (hiPSC-ECs). In vitro, hiPSC-ECs responded to flow stress by acquiring an alignment orientation, and attached to and proliferated on the acellular kidney sections, maintaining their phenotype. The hiPSC-ECs were able to self-organize into chimeric kidney organoids to form vessel-like structures. Ex vivo infusion of hiPSC-ECs through the renal artery and vein of acellular kidneys resulted in the uniform distribution of the cells in all the vasculature compartments, from glomerular capillaries to peritubular capillaries and small vessels. Ultrastructural analysis of repopulated scaffolds through transmission and scanning electron microscopy demonstrated the presence of continuously distributed cells along the vessel wall, which was also confirmed by 3D reconstruction of z-stack images showing the continuity of endothelial cell coverage inside the vessels. Notably, the detection of fenestrae in the endothelium of glomerular capillaries but not in the vascular capillaries was clear evidence of site-specific endothelial cell specialisation.


Assuntos
Rim/química , Neovascularização Fisiológica/genética , Organoides/crescimento & desenvolvimento , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Vasos Sanguíneos/química , Vasos Sanguíneos/crescimento & desenvolvimento , Diferenciação Celular/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio/química , Endotélio/crescimento & desenvolvimento , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Rim/crescimento & desenvolvimento , Organoides/química , Ratos
6.
ACS Appl Mater Interfaces ; 11(10): 10337-10350, 2019 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-30753784

RESUMO

Tunable/sustained drug loading/releasing are of significance in addressing low cytotoxicity, long-term performance, and localized mild healing response in biomedical applications. With an ingenious design, a self-healing sandwiched layer-by-layer (LBL) coating was constructed by using chitosan/heparin as adopted polyelectrolytes with embedding of micelles, in which the chitosan backbone was grafted with catechol and the micelle was modified with exposed phenylboronic acid, endowing the coating with enhanced stability by abundant interactions among coating components (e.g., boric acid ester bond formation, weak intermolecular cross-linking, π-π interactions, and H-bonding). Moreover, rapamycin and atorvastatin calcium were selected as drug candidates and loaded into micelles, followed by drug-releasing behavior study. It was found that the LBL coating maintained a linear growth mode up to 30 cycles, giving a favorable tunability of coating construction and drug loading. The coating could also support sustained release of payloads and provide wild tissue response. With the systematic in vitro and in vivo study, such catechol-phenylboronic acid-enhanced LBL coating with drug loading would also address enhanced antiplatelet adhesion/activation and direct cell fate of endothelial cells and smooth muscle cells via tuning of coating cycles and loaded drugs. With modular assembly, such coating indicated potential for achieving enhanced re-endothelialization for vascular implants.


Assuntos
Antibacterianos/química , Quitosana/química , Materiais Revestidos Biocompatíveis/química , Preparações de Ação Retardada/química , Endotélio/efeitos dos fármacos , Antibacterianos/farmacologia , Ácidos Borônicos/química , Catecóis/química , Quitosana/farmacologia , Materiais Revestidos Biocompatíveis/farmacologia , Preparações de Ação Retardada/farmacologia , Liberação Controlada de Fármacos , Endotélio/crescimento & desenvolvimento , Endotélio/microbiologia , Humanos , Micelas , Próteses e Implantes/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/patogenicidade
7.
J Mol Cell Cardiol ; 121: 145-154, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30003882

RESUMO

BACKGROUND: Antiproliferative drugs in drug eluting stents (DES) are associated with complications due to impaired re-endothelialization. Additionally, adventitial neovascularization has been suggested to contribute to in-stent restenosis (ISR). Since Vascular Endothelial Growth Factors (VEGFs) are the key mediators of angiogenesis, we investigated feasibility and efficacy of local gene therapy for ISR utilizing soluble decoy VEGF receptors to reduce biological activity of adventitial VEGFs. METHOD: Sixty-nine adult WHHL rabbit aortas were subjected to endothelial denudation. Six weeks later catheter-mediated local intramural infusion of 1.5x10e10 pfu adenoviruses encoding soluble VEGF Receptor-1 (sVEGFR1), sVEGFR2, sVEGFR3 or control LacZ and bare metal stent implantation were performed in the same aortic segment. Marker protein expression was assessed at 6d in LacZ cohort. Immunohistochemistry, morphometrical analyses and angiography were performed at d14, d42 and d90. RESULTS: Transgene expression was localized to adventitia. All decoy receptors reduced the size of vasa-vasorum at 14d, AdsVEGFR2 animals also had reduced density of adventitial vasa-vasorum, whereas AdsVEGFR3 increased the density of vasa-vasorum. At d42, AdsVEGFR1 and AdsVEGFR2 reduced ISR (15.7 ±â€¯6.9% stenosis, P < 0.01 and 16.5 ±â€¯2.7%, P < 0.05, respectively) vs. controls (28.3 ±â€¯7.6%). Moreover, AdsVEGFR-3 treatment led to a non-significant trend in the reduction of adventitial lymphatics at all time points and these animals had significantly more advanced neointimal atherosclerosis at 14d and 42d vs. control animals. CONCLUSIONS: Targeting adventitial neovascularization using sVEGFR1 and sVEGFR2 is a novel strategy to reduce ISR. The therapeutic effects dissipate at late follow up following short expression profile of adenoviral vectors. However, inhibition of VEGFR3 signaling accelerates neoatherosclerosis.


Assuntos
Constrição Patológica/terapia , Reestenose Coronária/terapia , Terapia Genética , Neointima/terapia , Neovascularização Patológica/tratamento farmacológico , Túnica Adventícia/fisiopatologia , Animais , Aorta/fisiopatologia , Constrição Patológica/genética , Constrição Patológica/fisiopatologia , Reestenose Coronária/genética , Reestenose Coronária/fisiopatologia , Stents Farmacológicos , Endotélio/citologia , Endotélio/efeitos dos fármacos , Endotélio/crescimento & desenvolvimento , Endotélio Vascular/fisiopatologia , Humanos , Neointima/genética , Neointima/fisiopatologia , Neovascularização Patológica/genética , Neovascularização Patológica/fisiopatologia , Coelhos , Vasa Vasorum/fisiopatologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Fatores de Crescimento do Endotélio Vascular/genética , Fatores de Crescimento do Endotélio Vascular/uso terapêutico
8.
Oncogene ; 36(19): 2680-2692, 2017 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-27893712

RESUMO

During metastasis to distant sites, tumor cells migrate to blood vessels. In vivo, breast tumor cells utilize a specialized mode of migration known as streaming, where a linear assembly of tumor cells migrate directionally towards blood vessels on fibronectin-collagen I-containing extracellular matrix (ECM) fibers in response to chemotactic signals. We have successfully reconstructed tumor cell streaming in vitro by co-plating tumors cells, macrophages and endothelial cells on 2.5 µm thick ECM-coated micro-patterned substrates. We found that tumor cells and macrophages, when plated together on the micro-patterned substrates, do not demonstrate sustained directional migration in only one direction (sustained directionality) but show random bi-directional walking. Sustained directionality of tumor cells as seen in vivo was established in vitro when beads coated with human umbilical vein endothelial cells were placed at one end of the micro-patterned 'ECM fibers' within the assay. We demonstrated that these endothelial cells supply the hepatocyte growth factor (HGF) required for the chemotactic gradient responsible for sustained directionality. Using this in vitro reconstituted streaming system, we found that directional streaming is dependent on, and most effectively blocked, by inhibiting the HGF/C-Met signaling pathway between endothelial cells and tumor cells. Key observations made with the in vitro reconstituted system implicating C-Met signaling were confirmed in vivo in mammary tumors using the in vivo invasion assay and intravital multiphoton imaging of tumor cell streaming. These results establish HGF/C-Met as a central organizing signal in blood vessel-directed tumor cell migration in vivo and highlight a promising role for C-Met inhibitors in blocking tumor cell streaming and metastasis in vivo, and for use in human trials.


Assuntos
Neoplasias da Mama/genética , Proliferação de Células/genética , Fator de Crescimento de Hepatócito/genética , Proteínas Proto-Oncogênicas c-met/genética , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/patologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Células Endoteliais/patologia , Endotélio/crescimento & desenvolvimento , Endotélio/patologia , Matriz Extracelular/genética , Matriz Extracelular/patologia , Feminino , Humanos , Macrófagos/patologia , Transdução de Sinais
9.
Gene Expr Patterns ; 21(2): 103-10, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27221232

RESUMO

Extravillous cytotrophoblasts isolated from first trimester placenta, and immortalised cell lines derived from them, have the intrinsic ability to form endothelial-like tubes when cultured on Matrigel™ extracellular matrix. This in vitro tube formation may model placental angiogenesis and/or endovascular differentiation by trophoblasts. To interpret the relevance of this phenomenon to placental development, we used a gene expression microarray approach to identify which genes and pathways are associated with the tube-forming phenotype of HTR8/SVneo first trimester trophoblasts (HTR8-M), compared with HTR8/SVneo not forming tubes on plastic culture surface (HTR8-P). Furthermore, we used weighted gene co-expression network analysis (WGCNA) of microarray data to identify modules of co-expressed genes underlying the biological processes. There were 481 genes differentially expressed between HTR8-M and HTR8-P and these were significantly enriched for blood vessel development and related gene ontologies. WGCNA clustered the genes into 9 co-expression modules. One module was significantly associated with HTR8-M (p = 1.15E-05) and contained genes involved in actin cytoskeleton organization, cell migration and blood vessel development, consistent with tube formation on Matrigel. Another module was significantly associated with HTR8-P (p = 1.94E-05) and was enriched for genes involved in mitosis, consistent with proliferation by cells on plastic which do not differentiate. Up-regulation of angiogenesis and vascular development pathways in endovascular trophoblasts in vivo could underpin spiral artery remodelling processes, which are defective in preeclamptic pregnancies.


Assuntos
Vasos Sanguíneos/metabolismo , Biossíntese de Proteínas/genética , Transcriptoma/genética , Trofoblastos/metabolismo , Vasos Sanguíneos/crescimento & desenvolvimento , Diferenciação Celular/genética , Movimento Celular/genética , Endotélio/crescimento & desenvolvimento , Endotélio/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Placenta/metabolismo , Gravidez , Primeiro Trimestre da Gravidez
10.
Stem Cell Reports ; 6(5): 692-703, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27117782

RESUMO

Hematopoietic cells emerge from hemogenic endothelium in the developing embryo. Mechanisms behind human hematopoietic stem and progenitor cell development remain unclear. Using a human pluripotent stem cell differentiation model, we report that cyclic AMP (cAMP) induction dramatically increases HSC-like cell frequencies. We show that hematopoietic cell generation requires cAMP signaling through the Exchange proteins activated by cAMP (cAMP-Epac) axis; Epac signaling inhibition decreased both hemogenic and non-hemogenic endothelium, and abrogated hematopoietic cell generation. Furthermore, in hematopoietic progenitor and stem-like cells, cAMP induction mitigated oxidative stress, created a redox-state balance, and enhanced C-X-C chemokine receptor type 4 (CXCR4) expression, benefiting the maintenance of these primitive cells. Collectively, our study provides insights and mechanistic details on the previously unrecognized role of cAMP signaling in regulating human hematopoietic development. These findings advance the mechanistic understanding of hematopoietic development toward the development of transplantable human hematopoietic cells for therapeutic needs.


Assuntos
Diferenciação Celular/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Receptores CXCR4/genética , AMP Cíclico/genética , Endotélio/crescimento & desenvolvimento , Endotélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Estresse Oxidativo/genética , Transdução de Sinais
11.
Curr Stem Cell Res Ther ; 11(3): 265-73, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26951130

RESUMO

Angiogenesis is a complex process in which capillaries are produced from blood vessels that already exists. Endothelial cells (ECs) and endothelial progenitor cells (EPCs) are pivotal for this process and for the maintenance/restorage of the endothelium. Decreased numbers and dysfunction of these cells have been related to growing cardiovascular risks. Peroxisome-proliferator-activated receptor (PPAR) is a large family of nuclear receptors, characterized by three isotypes: α, ß and γ. Numerous studies have shown that PPAR activation is involved in the pathology of a wide range of cardiovascular diseases and has a role in endothelial function, thrombosis and inflammation, etc., suggesting that PPAR agonists may be good candidates to treat the cardiovascular disease. However, controversial results exist on whether this nuclear receptor is inductive or depressive in the process of angiogenesis. Herein, this review will provide a detailed discussion of the up-to-date investigation of the role of PPARs in angiogenesis, with particular reference to their effects on angiogenesis-related cells--i.e., ECs, EPCs, vascular smooth-muscle cells (VSMCs), macrophages and endometrial cells--and will discuss the current and potential future applications of PPAR activators.


Assuntos
Células Endoteliais/citologia , Endotélio/citologia , Músculo Liso Vascular/citologia , Neovascularização Fisiológica/fisiologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Doenças Cardiovasculares/patologia , Endométrio/citologia , Endotélio/crescimento & desenvolvimento , Feminino , Humanos , Macrófagos/citologia , Engenharia Tecidual/métodos
12.
Nat Commun ; 6: 6429, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25752958

RESUMO

Angiogenic blood vessel growth requires several distinct but integrated cellular activities. Endothelial cell sprouting and proliferation lead to the expansion of the vasculature and give rise to a highly branched, immature plexus, which is subsequently reorganized into a mature and stable network. Although it is known that integrin-mediated cell-matrix interactions are indispensable for embryonic angiogenesis, little is known about the function of integrins in different steps of vascular morphogenesis. Here, by investigating the integrin ß1-subunit with inducible and endothelial-specific gene targeting in the postnatal mouse retina, we show that ß1 integrin promotes endothelial sprouting but is a negative regulator of proliferation. In maturing vessels, integrin ß1 is indispensable for proper localization of VE-cadherin and thereby cell-cell junction integrity. The sum of our findings establishes that integrin ß1 has critical functions in the growing and maturing vasculature, and is required for the formation of stable, non-leaky blood vessels.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Endotélio/crescimento & desenvolvimento , Integrina beta1/metabolismo , Junções Intercelulares/fisiologia , Morfogênese/fisiologia , Neovascularização Fisiológica/fisiologia , Vasos Retinianos/crescimento & desenvolvimento , Animais , Western Blotting , Encéfalo/anatomia & histologia , Proliferação de Células , Endotélio/metabolismo , Marcação de Genes , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Imunoprecipitação , Junções Intercelulares/metabolismo , Camundongos , Microscopia Eletrônica , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Vasos Retinianos/ultraestrutura
13.
Tissue Eng Part A ; 20(1-2): 147-59, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23895198

RESUMO

Osteochondral tissue repair requires formation of vascularized bone and avascular cartilage. Mesenchymal stem cells stimulate angiogenesis both in vitro and in vivo but it is not known if these proangiogenic properties change as a result of chondrogenic or osteogenic differentiation. We investigated the angiogenic/antiangiogenic properties of equine bone marrow-derived mesenchymal stem cells (eBMSCs) before and after differentiation in vitro. Conditioned media from chondrogenic and osteogenic cell pellets and undifferentiated cells was applied to endothelial tube formation assays using Matrigel™. Additionally, the cell secretome was analysed using LC-MS/MS mass spectrometry and screened for angiogenesis and neurogenesis-related factors using protein arrays. Endothelial tube-like formation was supported by conditioned media from undifferentiated eBMSCs. Conversely, chondrogenic and osteogenic conditioned media was antiangiogenic as shown by significantly decreased length of endothelial tube-like structures and degree of branching compared to controls. Undifferentiated cells produced higher levels of angiogenesis-related proteins compared to chondrogenic and osteogenic pellets. In summary, eBMSCs produce an array of angiogenesis-related proteins and support angiogenesis in vitro via a paracrine mechanism. However, when these cells are differentiated chondrogenically or osteogenically, they produce a soluble factor(s) that inhibits angiogenesis. With respect to osteochondral tissue engineering, this may be beneficial for avascular articular cartilage formation but unfavourable for bone formation where a vascularized tissue is desired.


Assuntos
Osso e Ossos/fisiologia , Diferenciação Celular , Condrogênese , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica , Osteogênese , Engenharia Tecidual/métodos , Animais , Células da Medula Óssea/citologia , Osso e Ossos/efeitos dos fármacos , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Cromatografia Líquida , Colágeno/farmacologia , Meios de Cultivo Condicionados/farmacologia , Combinação de Medicamentos , Endotélio/crescimento & desenvolvimento , Cavalos , Humanos , Cinética , Laminina/farmacologia , Espectrometria de Massas , Neovascularização Fisiológica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fenótipo , Proteoglicanas/farmacologia , Proteômica
14.
Tissue Eng Part A ; 20(5-6): 954-65, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24138406

RESUMO

Pluripotent embryonic stem cells (ESCs) are capable of differentiating into all mesoderm-derived cell lineages, including endothelial, hematopoietic, and cardiac cell types. Common strategies to direct mesoderm differentiation of ESCs rely on exposing the cells to a series of biochemical and biophysical cues at different stages of differentiation to promote maturation toward specific cell phenotypes. Shear forces that mimic cardiovascular physiological forces can evoke a myriad of responses in somatic and stem cell populations, and have, thus, been studied as a means to direct stem cell differentiation. However, elucidating the effects of shear pre-conditioning on the subsequent vascular differentiation and morphogenesis of ESCs has yet to be examined. In this study, ESC monolayers were subjected to physiological shear (5 dyn/cm(2)) or static conditions for 2 days on collagen IV-coated substrates before initiating embryoid body (EB) differentiation. Immediately after the pre-conditioning period, shear pre-conditioned and statically cultured ESCs exhibited similar morphologies and largely retained a pluripotent phenotype; however, ESCs exposed to fluid shear expressed increased levels of endothelial marker genes Flk-1 (∼3-fold), VE-cadherin (∼3-fold), and PECAM (∼2-fold), compared with statically cultured ESCs. After 7 days of EB culture, ∼70% of EBs formed from shear pre-conditioned ESCs expressed significantly higher levels of endothelial marker genes compared with EBs formed from statically cultured ESCs. Interestingly, unlike EBs formed from statically cultured ESCs, EBs formed from fluid shear stress pre-conditioned ESCs exhibited a centrally localized region of VE-cadherin(+) cells that persisted for at least 10 days of differentiation. These results demonstrate that fluid shear stress pre-conditioning not only promotes ESC endothelial gene expression but also subsequently impacts the organization of endothelial cells within EBs. Together, these studies highlight a novel approach to promote in vitro morphogenesis of developmental vasculogenic models and potentially promote pre-vascularization of tissue-engineered constructs derived from pluripotent stem cells.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Corpos Embrioides/citologia , Endotélio/efeitos dos fármacos , Endotélio/crescimento & desenvolvimento , Morfogênese/efeitos dos fármacos , Resistência ao Cisalhamento , Estresse Mecânico , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Camundongos , Fatores de Tempo , Fator de von Willebrand/metabolismo
15.
J Bone Miner Res ; 28(9): 1870-1884, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23649506

RESUMO

Activating transcription factor 4 (ATF4) is a critical transcription factor for bone remodeling; however, its role in bone angiogenesis has not been established. Here we show that ablation of the Atf4 gene expression in mice severely impaired skeletal vasculature and reduced microvascular density of the bone associated with dramatically decreased expression of hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF) in osteoblasts located on bone surfaces. Results from in vivo studies revealed that hypoxia/reoxygenation induction of HIF-1α and VEGF expression leading to bone angiogenesis, a key adaptive response to hypoxic conditions, was severely compromised in mice lacking the Atf4 gene. Loss of ATF4 completely prevented endothelial sprouting from embryonic metatarsals, which was restored by addition of recombinant human VEGF protein. In vitro studies revealed that ATF4 promotion of HIF-1α and VEGF expression in osteoblasts was highly dependent upon the presence of hypoxia. ATF4 interacted with HIF-1α in hypoxic osteoblasts, and loss of ATF4 increased HIF-1α ubiquitination and reduced its protein stability without affecting HIF-1α mRNA stability and protein translation. Loss of ATF4 increased the binding of HIF-1α to prolyl hydroxylases, the enzymes that hydroxylate HIF-1a protein and promote its proteasomal degradation via the pVHL pathway. Furthermore, parathyroid hormone-related protein (PTHrP) and receptor activator of NF-κB ligand (RANKL), both well-known activators of osteoclasts, increased release of VEGF from the bone matrix and promoted angiogenesis through the protein kinase C- and ATF4-dependent activation of osteoclast differentiation and bone resorption. Thus, ATF4 is a new key regulator of the HIF/VEGF axis in osteoblasts in response to hypoxia and of VEGF release from bone matrix, two critical steps for bone angiogenesis.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Osso e Ossos/irrigação sanguínea , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator 4 Ativador da Transcrição/deficiência , Animais , Matriz Óssea/efeitos dos fármacos , Matriz Óssea/metabolismo , Reabsorção Óssea/patologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Endotélio/efeitos dos fármacos , Endotélio/crescimento & desenvolvimento , Endotélio/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Ossos do Metatarso/irrigação sanguínea , Ossos do Metatarso/metabolismo , Camundongos , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Oxigênio/farmacologia , Prolil Hidroxilases/metabolismo , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/genética , Proteína Quinase C/metabolismo , Estabilidade Proteica/efeitos dos fármacos , Estabilidade de RNA/efeitos dos fármacos , Estabilidade de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ubiquitinação/efeitos dos fármacos , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
16.
Nat Commun ; 4: 1831, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23652019

RESUMO

Caveolae are abundant in endothelial cells and are thought to have important roles in endothelial cell biology. The cavin proteins are key components of caveolae, and are expressed at varied amounts in different tissues. Here we use knockout mice to determine the roles of cavins 2 and 3 in caveolar morphogenesis in vivo. Deletion of cavin 2 causes loss of endothelial caveolae in lung and adipose tissue, but has no effect on the abundance of endothelial caveolae in heart and other tissues. Changes in the morphology of endothelium in cavin 2 null mice correlate with changes in caveolar abundance. Cavin 3 is not required for making caveolae in the tissues examined. Cavin 2 determines the size of cavin complexes, and acts to shape caveolae. Cavin 1, however, is essential for normal oligomerization of caveolin 1. Our data reveal that endothelial caveolae are heterogeneous, and identify cavin 2 as a determinant of this heterogeneity.


Assuntos
Cavéolas/metabolismo , Endotélio/crescimento & desenvolvimento , Endotélio/metabolismo , Deleção de Genes , Proteínas de Membrana/genética , Morfogênese , Especificidade de Órgãos , Animais , Caveolina 1/química , Caveolina 1/metabolismo , Forma Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Endotélio/ultraestrutura , Pulmão/citologia , Pulmão/metabolismo , Pulmão/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peso Molecular , Complexos Multiproteicos/metabolismo , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Estrutura Quaternária de Proteína , Proteínas de Ligação a RNA
17.
Free Radic Biol Med ; 60: 272-81, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23416364

RESUMO

High-density lipoprotein (HDL) plays a key role in protecting against atherosclerosis. In cardiovascular disease, HDL can be nitrated and chlorinated by myeloperoxidase (MPO). In this study, we discovered that MPO-oxidized HDL is dysfunctional in promoting endothelial repair compared to normal HDL. Proliferation assay, wound healing, and transwell migration experiments showed that MPO-oxidized HDL was associated with a reduced stimulation of endothelial cell (EC) proliferation and migration. In addition, we found that Akt and ERK1/2 phosphorylation in ECs was significantly lower when ECs were incubated with oxidized HDL compared with normal HDL. To further determine whether oxidized HDL diminished EC migration through the PI3K/Akt and MEK/ERK pathways, we performed experiments with inhibitors of both these pathways. The transwell experiments performed in the presence of these inhibitors showed that the migration capacity was reduced and the differences observed between normal HDL and oxidized HDL were diminished. Furthermore, to study the effects of oxidized HDL on endothelial cells in vivo, we performed a carotid artery electric injury model on nude mice injected with either normal or oxidized HDL. Oxidized HDL inhibited reendothelialization compared to normal HDL in vivo. These findings implicate a key role for MPO-oxidized HDL in the pathogenesis of cardiovascular disease.


Assuntos
Doenças Cardiovasculares/metabolismo , Endotélio/crescimento & desenvolvimento , Lipoproteínas HDL/administração & dosagem , Peroxidase/metabolismo , Animais , Doenças Cardiovasculares/fisiopatologia , Catálise , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio/metabolismo , Halogenação , Humanos , Lipoproteínas HDL/química , Lipoproteínas HDL/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Nitratos/administração & dosagem , Nitratos/química , Nitratos/metabolismo , Oxirredução , Proteínas Proto-Oncogênicas c-akt/metabolismo
18.
Bioarchitecture ; 2(6): 220-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23267416

RESUMO

Because little is known how microtubules contribute to cell migration in a physiological three-dimensional environment, we analyzed microtubule function and dynamics during in vitro angiogenesis in which endothelial cells form networks on a reconstituted basement membrane. Endothelial network formation resulted from distinct cell behaviors: matrix reorganization by myosin-mediated contractile forces, and active cell migration along reorganized, bundled matrix fibers. Inhibition of microtubule dynamics inhibited persistent cell migration, but not matrix reorganization. In addition, microtubule polymerization dynamics and CLASP2-binding to microtubules were spatially regulated to promote microtubule growth into endothelial cell protrusions along matrix tension tracks. We propose that microtubules counter-act contractile forces of the cortical actin cytoskeleton and are required to stabilize endothelial cell protrusions in a soft three-dimensional environment.


Assuntos
Endotélio/crescimento & desenvolvimento , Microtúbulos/metabolismo , Morfogênese , Fenômenos Biomecânicos , Movimento Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio/metabolismo , Matriz Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Miosinas/metabolismo
19.
Differentiation ; 83(5): 260-70, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22466671

RESUMO

Chorion, amnion and villi are reservoirs of mesenchymal stromal cells (StC) and the hypothesis that StC from fetal tissues retain higher plasticity compared to adult StC has been suggested. Aimed at investigating this aspect, a series of in vitro experiments were performed with StC isolated from first trimester human chorionic villi (CVStC). CVStC were cultured in: (i) standard mesenchymal medium (MM) and (ii) AmniomaxII® (AM), specifically designed to grow amnion-derived cells in prenatal diagnostic procedures. Cells were then exposed to distinct differentiation treatments and distinguished according to morphology, immunophenotype and molecular markers. Human StC obtained from adult bone marrow (BMStC) were used as control. CVStC cultured either in MM or AM presented stromal morphology and immunophenotype, were negative for pluripotency factors (Nanog, Oct-4 and Sox-2), lacked detectable telomerase activity and retained high genomic stability. In AM, however, CVStC exhibited a faster proliferation rate compared to BMStC or CVStC kept in MM. During differentiation, CVStC were less efficient than BMStC in acquiring adipocytes and osteocytes features; the cardiomyogenic conversion occurred at low efficiency in both cell types. Remarkably, in the presence of pro-angiogenic factors, CVStC reprogrammed toward an endothelial-like phenotype at significantly higher efficiency than BMStC. This effect was particularly evident in CVStC expanded in AM. Mechanistically, the reduced CVStC expression of anti-angiogenic microRNA could support this process. The present study demonstrates that, despite of fetal origin, CVStC exhibit restricted plasticity, distinct from that of BMStC and predominantly directed toward the endothelial lineage.


Assuntos
Diferenciação Celular , Linhagem da Célula , Meios de Cultura , Endotélio/crescimento & desenvolvimento , Células-Tronco Mesenquimais/citologia , Medula Óssea/crescimento & desenvolvimento , Proliferação de Células , Vilosidades Coriônicas/crescimento & desenvolvimento , Endotélio/citologia , Instabilidade Genômica , Proteínas de Homeodomínio/metabolismo , Humanos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Telomerase/metabolismo
20.
J Steroid Biochem Mol Biol ; 129(3-5): 145-52, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22212769

RESUMO

Angiogenesis plays a pivotal role in cardiovascular diseases such as ischemic heart disease, limb ischemia and heart failure, and has recently been shown to mediate various biological activities related to the pathogenesis of these diseases. In the present study, we evaluated the role of aldosterone in angiogenesis. Tube formation assay on Matrigel using human umbilical vein endothelial cells (HUVEC) revealed that aldosterone inhibited endothelial morphogenesis in a manner sensitive to eplerenone, a selective mineralocorticoid receptor antagonist. The anti-angiogenic effect of aldosterone was further confirmed by an in vivo angiogenesis assay using a Matrigel plug model in mice. Reverse transcription-mediated polymerase chain reaction and immunoblotting demonstrated that aldosterone downregulated the expression levels of vascular endothelial growth factor receptor-2 (VEGFR-2) and peroxisome proliferators-activated receptor gamma (PPAR gamma). VEGFR-2 expression was found to be enhanced in response to PPAR gamma activation by troglitazone, and attenuated by GW9662, a specific antagonist of PPAR gamma. In the tube formation assay, endothelial morphogenesis was stimulated by troglitazone, and inhibited by GW9662, indicating that PPAR gamma activation mediates positive regulation of angiogenesis through enhancement of VEGFR-2 expression. These data suggest that aldosterone inhibits angiogenesis through VEGFR-2 downregulation, subsequent to, at least in part, attenuation of PPAR gamma expression. The present findings provide a new insight into the possible therapeutic application of mineralocorticoid receptor blockade to various cardiovascular diseases.


Assuntos
Aldosterona/farmacologia , Inibidores da Angiogênese/farmacologia , Regulação para Baixo/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , PPAR gama/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Colágeno/farmacologia , Combinação de Medicamentos , Endotélio/crescimento & desenvolvimento , Eplerenona , Células Endoteliais da Veia Umbilical Humana , Humanos , Laminina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Morfogênese/efeitos dos fármacos , PPAR gama/metabolismo , Proteoglicanas/farmacologia , Espironolactona/análogos & derivados , Espironolactona/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...