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











Base de dados
Intervalo de ano de publicação
1.
Cell Stem Cell ; 29(4): 593-609.e7, 2022 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-35364013

RESUMO

The liver vascular network is patterned by sinusoidal and hepatocyte co-zonation. How intra-liver vessels acquire their hierarchical specialized functions is unknown. We study heterogeneity of hepatic vascular cells during mouse development through functional and single-cell RNA-sequencing. The acquisition of sinusoidal endothelial cell identity is initiated during early development and completed postnatally, originating from a pool of undifferentiated vascular progenitors at E12. The peri-natal induction of the transcription factor c-Maf is a critical switch for the sinusoidal identity determination. Endothelium-restricted deletion of c-Maf disrupts liver sinusoidal development, aberrantly expands postnatal liver hematopoiesis, promotes excessive postnatal sinusoidal proliferation, and aggravates liver pro-fibrotic sensitivity to chemical insult. Enforced c-Maf overexpression in generic human endothelial cells switches on a liver sinusoidal transcriptional program that maintains hepatocyte function. c-Maf represents an inducible intra-organotypic and niche-responsive molecular determinant of hepatic sinusoidal cell identity and lays the foundation for the strategies for vasculature-driven liver repair.


Assuntos
Capilares , Células Endoteliais , Animais , Endotélio , Fígado/patologia , Cirrose Hepática/patologia , Regeneração Hepática , Camundongos , Proteínas Proto-Oncogênicas c-maf
2.
Int J Comput Assist Radiol Surg ; 16(10): 1761-1773, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34424457

RESUMO

PURPOSE: Robotic systems have the potential to overcome inherent limitations of humans and offer substantial advantages to patients including reduction in surgery time. Our group has undertaken the challenge of developing autonomous wound closure system. One of the initial steps is to allow accurate assessment of wound skin topology and wound edge location. We present a vision-laser scanner to generate 3D point cloud for 3D reconstruction of wound's edge and topology. METHODS: When the laser range sensor measures Z coordinate, two encoders installed on the actuators of the gantry robot provide the precision values of X, Y coordinates simultaneously. The 3D point cloud of the wound skin is generated by recordings of X, Y and Z during scanning is performed over wound skin surface. To reduce the scanning time, we exploit a supplementary laser LED to project a regular laser spot on the wound skin surface, which can provide an additional measurement point by incorporating artificial neural network estimation approach. In the meantime, the point cloud of the wound edge can be extracted by detecting if the laser spot is located on the wound edge in the image from 2D camera. RESULTS: The mean absolute error (MAE) and standard deviation (σ) of wound edge are measured in MeshLab environment. The MAE (σ) in X (tangent), Y (tangent), and Z (normal) are 0.32 (0.22) mm, 0.37 (0.34) mm, and 0.61 (0.29) mm, respectively. The experimental results demonstrate that the vision-laser scanner attains high accuracy in determining wound edge location along the tangent of the wound skin. CONCLUSION: A vision-laser scanner is developed for 3D reconstruction of wound's edge and topology. The experimental tests on the different wound models revealed the effectiveness of the vision-laser scanner. The proposed scanner can generate 3D point cloud of the wound skin and its edge simultaneously, and thus significantly improve the accuracy of wound closure in clinical applications.


Assuntos
Imageamento Tridimensional , Lasers , Humanos , Luz , Redes Neurais de Computação
3.
Microvasc Res ; 138: 104205, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34146583

RESUMO

The rapid engraftment of vascular networks is critical for functional incorporation of tissue explants. However, existing methods for inducing angiogenesis utilize approaches that yield vasculature with poor temporal stability or inadequate mechanical integrity, which reduce their robustness in vivo. The transcription factor Ets variant 2 (Etv2) specifies embryonic hematopoietic and vascular endothelial cell (EC) development, and is transiently reactivated during postnatal vascular regeneration and tumor angiogenesis. This study investigates the role for Etv2 upregulation in forming stable vascular beds both in vitro and in vivo. Control and Etv2+ prototypical fetal-derived human umbilical vein ECs (HUVECs) and adult ECs were angiogenically grown into vascular beds. These vessel beds were characterized using fractal dimension and lacunarity, to quantify their branching complexity and space-filling homogeneity, respectively. Atomic force microscopy (AFM) was used to explore whether greater complexity and homogeneity lead to more mechanically stable vessels. Additionally, markers of EC integrity were used to probe for mechanistic clues. Etv2+ HUVECs exhibit greater branching, vessel density, and structural homogeneity, and decreased stiffness in vitro and in vivo, indicating a greater propensity for stable vessel formation. When co-cultured with colon tumor organoid tissue, Etv2+ HUVECs had decreased fractal dimension and lacunarity compared to Etv2+ HUVECs cultured alone, indicating that vessel density and homogeneity of vessel spacing increased due to the presence of Etv2. This study sets forth the novel concept that fractal dimension, lacunarity, and AFM are as informative as conventional angiogenic measurements, including vessel branching and density, to assess vascular perfusion and stability.


Assuntos
Forma Celular , Neoplasias do Colo/irrigação sanguínea , Fractais , Células Endoteliais da Veia Umbilical Humana/metabolismo , Processamento de Imagem Assistida por Computador , Microscopia de Força Atômica , Neovascularização Fisiológica , Fatores de Transcrição/metabolismo , Células Cultivadas , Técnicas de Cocultura , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Neovascularização Patológica , Protocaderinas/metabolismo , Técnicas de Cultura de Tecidos , Fatores de Transcrição/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
Nature ; 585(7825): 426-432, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32908310

RESUMO

Endothelial cells adopt tissue-specific characteristics to instruct organ development and regeneration1,2. This adaptability is lost in cultured adult endothelial cells, which do not vascularize tissues in an organotypic manner. Here, we show that transient reactivation of the embryonic-restricted ETS variant transcription factor 2 (ETV2)3 in mature human endothelial cells cultured in a serum-free three-dimensional matrix composed of a mixture of laminin, entactin and type-IV collagen (LEC matrix) 'resets' these endothelial cells to adaptable, vasculogenic cells, which form perfusable and plastic vascular plexi. Through chromatin remodelling, ETV2 induces tubulogenic pathways, including the activation of RAP1, which promotes the formation of durable lumens4,5. In three-dimensional matrices-which do not have the constraints of bioprinted scaffolds-the 'reset' vascular endothelial cells (R-VECs) self-assemble into stable, multilayered and branching vascular networks within scalable microfluidic chambers, which are capable of transporting human blood. In vivo, R-VECs implanted subcutaneously in mice self-organize into durable pericyte-coated vessels that functionally anastomose to the host circulation and exhibit long-lasting patterning, with no evidence of malformations or angiomas. R-VECs directly interact with cells within three-dimensional co-cultured organoids, removing the need for the restrictive synthetic semipermeable membranes that are required for organ-on-chip systems, therefore providing a physiological platform for vascularization, which we call 'Organ-On-VascularNet'. R-VECs enable perfusion of glucose-responsive insulin-secreting human pancreatic islets, vascularize decellularized rat intestines and arborize healthy or cancerous human colon organoids. Using single-cell RNA sequencing and epigenetic profiling, we demonstrate that R-VECs establish an adaptive vascular niche that differentially adjusts and conforms to organoids and tumoroids in a tissue-specific manner. Our Organ-On-VascularNet model will permit metabolic, immunological and physiochemical studies and screens to decipher the crosstalk between organotypic endothelial cells and parenchymal cells for identification of determinants of endothelial cell heterogeneity, and could lead to advances in therapeutic organ repair and tumour targeting.


Assuntos
Vasos Sanguíneos/citologia , Carcinogênese , Células Endoteliais/citologia , Hemodinâmica , Neoplasias/irrigação sanguínea , Organogênese , Organoides/irrigação sanguínea , Vasos Sanguíneos/crescimento & desenvolvimento , Técnicas de Cultura de Células/instrumentação , Técnicas de Cultura de Células/métodos , Cromatina/metabolismo , Epigênese Genética , Epigenômica , Células Endoteliais da Veia Umbilical Humana , Humanos , Técnicas In Vitro , Ilhotas Pancreáticas/irrigação sanguínea , Modelos Biológicos , Especificidade de Órgãos , RNA-Seq , Análise de Célula Única , Fatores de Transcrição , Transcriptoma
5.
Nat Commun ; 10(1): 5705, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31836710

RESUMO

Although kidney parenchymal tissue can be generated in vitro, reconstructing the complex vasculature of the kidney remains a daunting task. The molecular pathways that specify and sustain functional, phenotypic and structural heterogeneity of the kidney vasculature are unknown. Here, we employ high-throughput bulk and single-cell RNA sequencing of the non-lymphatic endothelial cells (ECs) of the kidney to identify the molecular pathways that dictate vascular zonation from embryos to adulthood. We show that the kidney manifests vascular-specific signatures expressing defined transcription factors, ion channels, solute transporters, and angiocrine factors choreographing kidney functions. Notably, the ontology of the glomerulus coincides with induction of unique transcription factors, including Tbx3, Gata5, Prdm1, and Pbx1. Deletion of Tbx3 in ECs results in glomerular hypoplasia, microaneurysms and regressed fenestrations leading to fibrosis in subsets of glomeruli. Deciphering the molecular determinants of kidney vascular signatures lays the foundation for rebuilding nephrons and uncovering the pathogenesis of kidney disorders.


Assuntos
Capilares/crescimento & desenvolvimento , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glomérulos Renais/irrigação sanguínea , Animais , Capilares/citologia , Capilares/metabolismo , Células Cultivadas , Embrião de Mamíferos , Endotélio Vascular/citologia , Endotélio Vascular/crescimento & desenvolvimento , Fator de Transcrição GATA5/genética , Fator de Transcrição GATA5/metabolismo , Perfilação da Expressão Gênica , Humanos , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B/metabolismo , Cultura Primária de Células , RNA-Seq , Análise de Célula Única , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo
6.
J Clin Invest ; 127(11): 4163-4178, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29035282

RESUMO

Age-related changes in the hematopoietic compartment are primarily attributed to cell-intrinsic alterations in hematopoietic stem cells (HSCs); however, the contribution of the aged microenvironment has not been adequately evaluated. Understanding the role of the bone marrow (BM) microenvironment in supporting HSC function may prove to be beneficial in treating age-related functional hematopoietic decline. Here, we determined that aging of endothelial cells (ECs), a critical component of the BM microenvironment, was sufficient to drive hematopoietic aging phenotypes in young HSCs. We used an ex vivo hematopoietic stem and progenitor cell/EC (HSPC/EC) coculture system as well as in vivo EC infusions following myelosuppressive injury in mice to demonstrate that aged ECs impair the repopulating activity of young HSCs and impart a myeloid bias. Conversely, young ECs restored the repopulating capacity of aged HSCs but were unable to reverse the intrinsic myeloid bias. Infusion of young, HSC-supportive BM ECs enhanced hematopoietic recovery following myelosuppressive injury and restored endogenous HSC function in aged mice. Coinfusion of young ECs augmented aged HSC engraftment and enhanced overall survival in lethally irradiated mice by mitigating damage to the BM vascular microenvironment. These data lay the groundwork for the exploration of EC therapies that can serve as adjuvant modalities to enhance HSC engraftment and accelerate hematopoietic recovery in the elderly population following myelosuppressive regimens.


Assuntos
Células Endoteliais/fisiologia , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Envelhecimento , Animais , Medula Óssea/irrigação sanguínea , Transplante de Medula Óssea , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/transplante , Camundongos Endogâmicos C57BL , Microvasos/patologia , Lesões Experimentais por Radiação/prevenção & controle , Tolerância a Radiação
7.
J Clin Invest ; 127(12): 4242-4256, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29058691

RESUMO

Angiocrine factors, such as Notch ligands, supplied by the specialized endothelial cells (ECs) within the bone marrow and splenic vascular niche play an essential role in modulating the physiology of adult hematopoietic stem and progenitor cells (HSPCs). However, the relative contribution of various Notch ligands, specifically jagged-2, to the homeostasis of HSPCs is unknown. Here, we show that under steady state, jagged-2 is differentially expressed in tissue-specific vascular beds, but its expression is induced in hematopoietic vascular niches after myelosuppressive injury. We used mice with EC-specific deletion of the gene encoding jagged-2 (Jag2) to demonstrate that while EC-derived jagged-2 was dispensable for maintaining the capacity of HSPCs to repopulate under steady-state conditions, by activating Notch2 it did contribute to the recovery of HSPCs in response to myelosuppressive conditions. Engraftment and/or expansion of HSPCs was dependent on the expression of endothelial-derived jagged-2 following myeloablation. Additionally, jagged-2 expressed in bone marrow ECs regulated HSPC cell cycle and quiescence during regeneration. Endothelial-deployed jagged-2 triggered Notch2/Hey1, while tempering Notch2/Hes1 signaling in HSPCs. Collectively, these data demonstrate that EC-derived jagged-2 activates Notch2 signaling in HSPCs to promote hematopoietic recovery and has potential as a therapeutic target to accelerate balanced hematopoietic reconstitution after myelosuppression.


Assuntos
Células-Tronco Adultas/metabolismo , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Proteína Jagged-2/biossíntese , Transdução de Sinais , Aloenxertos , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Deleção de Genes , Proteína Jagged-2/genética , Camundongos , Camundongos Transgênicos , Receptor Notch2/genética , Receptor Notch2/metabolismo , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo
8.
Nat Med ; 22(2): 154-62, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26779814

RESUMO

Although the lung can undergo self-repair after injury, fibrosis in chronically injured or diseased lungs can occur at the expense of regeneration. Here we study how a hematopoietic-vascular niche regulates alveolar repair and lung fibrosis. Using intratracheal injection of bleomycin or hydrochloric acid in mice, we show that repetitive lung injury activates pulmonary capillary endothelial cells (PCECs) and perivascular macrophages, impeding alveolar repair and promoting fibrosis. Whereas the chemokine receptor CXCR7, expressed on PCECs, acts to prevent epithelial damage and ameliorate fibrosis after a single round of treatment with bleomycin or hydrochloric acid, repeated injury leads to suppression of CXCR7 expression and recruitment of vascular endothelial growth factor receptor 1 (VEGFR1)-expressing perivascular macrophages. This recruitment stimulates Wnt/ß-catenin-dependent persistent upregulation of the Notch ligand Jagged1 (encoded by Jag1) in PCECs, which in turn stimulates exuberant Notch signaling in perivascular fibroblasts and enhances fibrosis. Administration of a CXCR7 agonist or PCEC-targeted Jag1 shRNA after lung injury promotes alveolar repair and reduces fibrosis. Thus, targeting of a maladapted hematopoietic-vascular niche, in which macrophages, PCECs and perivascular fibroblasts interact, may help to develop therapy to spur lung regeneration and alleviate fibrosis.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Capilares/metabolismo , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lesão Pulmonar/metabolismo , Pulmão/metabolismo , Proteínas de Membrana/metabolismo , Fibrose Pulmonar/metabolismo , Receptores CXCR/metabolismo , Regeneração/fisiologia , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Capilares/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Fibroblastos/efeitos dos fármacos , Fibrose , Imunofluorescência , Humanos , Ácido Clorídrico/toxicidade , Proteína Jagged-1 , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Oligopeptídeos/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Circulação Pulmonar/efeitos dos fármacos , Circulação Pulmonar/fisiologia , RNA Interferente Pequeno/farmacologia , Receptores CXCR/agonistas , Receptores Notch/metabolismo , Regeneração/efeitos dos fármacos , Proteínas Serrate-Jagged , Proteína Smad3/efeitos dos fármacos , Proteína Smad3/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Via de Sinalização Wnt
9.
Nat Cell Biol ; 17(2): 123-136, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25621952

RESUMO

The lung alveoli regenerate after surgical removal of the left lobe by pneumonectomy (PNX). How this alveolar regrowth/regeneration is initiated remains unknown. We found that platelets trigger lung regeneration by supplying stromal-cell-derived factor-1 (SDF-1, also known as CXCL12). After PNX, activated platelets stimulate SDF-1 receptors CXCR4 and CXCR7 on pulmonary capillary endothelial cells (PCECs) to deploy the angiocrine membrane-type metalloproteinase MMP14, stimulating alveolar epithelial cell (AEC) expansion and neo-alveolarization. In mice lacking platelets or platelet Sdf1, PNX-induced alveologenesis was diminished. Reciprocally, infusion of Sdf1(+/+) but not Sdf1-deficient platelets rescued lung regeneration in platelet-depleted mice. Endothelial-specific ablation of Cxcr4 and Cxcr7 in adult mice similarly impeded lung regeneration. Notably, mice with endothelial-specific Mmp14 deletion exhibited impaired expansion of AECs but not PCECs after PNX, which was not rescued by platelet infusion. Therefore, platelets prime PCECs to initiate lung regeneration, extending beyond their haemostatic contribution. Therapeutic targeting of this haemo-vascular niche could enable regenerative therapy for lung diseases.


Assuntos
Plaquetas/metabolismo , Capilares/metabolismo , Quimiocina CXCL12/metabolismo , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/fisiologia , Regeneração , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Células Endoteliais/enzimologia , Fator de Crescimento Epidérmico/metabolismo , Deleção de Genes , Ligantes , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Especificidade de Órgãos , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Pneumonectomia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4 , Transdução de Sinais , Trombopoetina/deficiência , Trombopoetina/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
Stem Cells ; 32(1): 177-90, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23963623

RESUMO

The ability to generate and maintain stable in vitro cultures of mouse endothelial cells (ECs) has great potential for genetic dissection of the numerous pathologies involving vascular dysfunction as well as therapeutic applications. However, previous efforts at achieving sustained cultures of primary stable murine vascular cells have fallen short, and the cellular requirements for EC maintenance in vitro remain undefined. In this study, we have generated vascular ECs from mouse embryonic stem (ES) cells and show that active Akt is essential to their survival and propagation as homogeneous monolayers in vitro. These cells harbor the phenotypical, biochemical, and functional characteristics of ECs and expand throughout long-term cultures, while maintaining their angiogenic capacity. Moreover, Akt-transduced embryonic ECs form functional perfused vessels in vivo that anastomose with host blood vessels. We provide evidence for a novel function of Akt in stabilizing EC identity, whereby the activated form of the protein protects mouse ES cell-derived ECs from TGFß-mediated transdifferentiation by downregulating SMAD3. These findings identify a role for Akt in regulating the developmental potential of ES cell-derived ECs and demonstrate that active Akt maintains endothelial identity in embryonic ECs by interfering with active TGFß-mediated processes that would ordinarily usher these cells to alternate fates.


Assuntos
Células-Tronco Embrionárias/metabolismo , Células Endoteliais/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Camundongos , Transdução de Sinais , Fator de Crescimento Transformador beta/antagonistas & inibidores
11.
Dev Cell ; 26(2): 204-19, 2013 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-23871589

RESUMO

Microvascular endothelial cells (ECs) within different tissues are endowed with distinct but as yet unrecognized structural, phenotypic, and functional attributes. We devised EC purification, cultivation, profiling, and transplantation models that establish tissue-specific molecular libraries of ECs devoid of lymphatic ECs or parenchymal cells. These libraries identify attributes that confer ECs with their organotypic features. We show that clusters of transcription factors, angiocrine growth factors, adhesion molecules, and chemokines are expressed in unique combinations by ECs of each organ. Furthermore, ECs respond distinctly in tissue regeneration models, hepatectomy, and myeloablation. To test the data set, we developed a transplantation model that employs generic ECs differentiated from embryonic stem cells. Transplanted generic ECs engraft into regenerating tissues and acquire features of organotypic ECs. Collectively, we demonstrate the utility of informational databases of ECs toward uncovering the extravascular and intrinsic signals that define EC heterogeneity. These factors could be exploited therapeutically to engineer tissue-specific ECs for regeneration.


Assuntos
Moléculas de Adesão Celular/metabolismo , Quimiocinas/metabolismo , Células Endoteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Regeneração , Fatores de Transcrição/metabolismo , Animais , Moléculas de Adesão Celular/biossíntese , Diferenciação Celular , Células Cultivadas , Quimiocinas/biossíntese , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/transplante , Células Endoteliais/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Camundongos , Microvasos/metabolismo , Fatores de Transcrição/biossíntese
12.
PLoS One ; 8(4): e62150, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23637986

RESUMO

In glioblastoma high expression of the CD133 gene, also called Prominin1, is associated with poor prognosis. The PDGF-driven proneural group represents a subset of glioblastoma in which CD133 is not overexpressed. Interestingly, this particular subset shows a relatively good prognosis. As with many other tumors, gliobastoma is believed to arise and be maintained by a restricted population of stem-like cancer cells that express the CD133 transmembrane protein. The significance of CD133(+) cells for gliomagenesis is controversial because of conflicting supporting evidence. Contributing to this inconsistency is the fact that the isolation of CD133(+) cells has largely relied on the use of antibodies against ill-defined glycosylated epitopes of CD133. To overcome this problem, we used a knock-in lacZ reporter mouse, Prom1(lacZ/+) , to track Prom1(+) cells in the brain. We found that Prom1 (prominin1, murine CD133 homologue) is expressed by cells that express markers characteristic of the neuronal, glial or vascular lineages. In proneural tumors derived from injection of RCAS-PDGF into the brains of tv-a;Ink4a-Arf(-/-) Prom1(lacZ/+) mice, Prom1(+) cells expressed markers for astrocytes or endothelial cells. Mice co-transplanted with proneural tumor sphere cells and Prom1(+) endothelium had a significantly increased tumor burden and more vascular proliferation (angiogenesis) than those co-transplanted with Prom1(-) endothelium. We also identified specific genes in Prom1(+) endothelium that code for endothelial signaling modulators that were not overexpressed in Prom1(-) endothelium. These factors may support proneural tumor progression and could be potential targets for anti-angiogenic therapy.


Assuntos
Antígenos CD/metabolismo , Encéfalo/patologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Glicoproteínas/metabolismo , Peptídeos/metabolismo , Antígeno AC133 , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/metabolismo , Proliferação de Células , Endotélio/metabolismo , Endotélio/patologia , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glicoproteínas/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neurogênese , Neurônios/metabolismo , Neurônios/patologia , Peptídeos/deficiência , Fenótipo
13.
Cell ; 151(3): 559-75, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-23084400

RESUMO

ETS transcription factors ETV2, FLI1, and ERG1 specify pluripotent stem cells into induced vascular endothelial cells (iVECs). However, iVECs are unstable and drift toward nonvascular cells. We show that human midgestation c-Kit(-) lineage-committed amniotic cells (ACs) can be reprogrammed into vascular endothelial cells (rAC-VECs) without transitioning through a pluripotent state. Transient ETV2 expression in ACs generates immature rAC-VECs, whereas coexpression with FLI1/ERG1 endows rAC-VECs with a vascular repertoire and morphology matching mature endothelial cells (ECs). Brief TGFß-inhibition functionalizes VEGFR2 signaling, augmenting specification of ACs into rAC-VECs. Genome-wide transcriptional analyses showed that rAC-VECs are similar to adult ECs in which vascular-specific genes are expressed and nonvascular genes are silenced. Functionally, rAC-VECs form stable vasculature in Matrigel plugs and regenerating livers. Therefore, short-term ETV2 expression and TGFß inhibition with constitutive ERG1/FLI1 coexpression reprogram mature ACs into durable rAC-VECs with clinical-scale expansion potential. Banking of HLA-typed rAC-VECs establishes a vascular inventory for treatment of diverse disorders.


Assuntos
Líquido Amniótico/citologia , Diferenciação Celular , Células Endoteliais/citologia , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Oncogênicas de Retroviridae/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos
14.
Cell ; 147(3): 539-53, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-22036563

RESUMO

To identify pathways involved in adult lung regeneration, we employ a unilateral pneumonectomy (PNX) model that promotes regenerative alveolarization in the remaining intact lung. We show that PNX stimulates pulmonary capillary endothelial cells (PCECs) to produce angiocrine growth factors that induce proliferation of epithelial progenitor cells supporting alveologenesis. Endothelial cells trigger expansion of cocultured epithelial cells, forming three-dimensional angiospheres reminiscent of alveolar-capillary sacs. After PNX, endothelial-specific inducible genetic ablation of Vegfr2 and Fgfr1 in mice inhibits production of MMP14, impairing alveolarization. MMP14 promotes expansion of epithelial progenitor cells by unmasking cryptic EGF-like ectodomains that activate the EGF receptor (EGFR). Consistent with this, neutralization of MMP14 impairs EGFR-mediated alveolar regeneration, whereas administration of EGF or intravascular transplantation of MMP14(+) PCECs into pneumonectomized Vegfr2/Fgfr1-deficient mice restores alveologenesis and lung inspiratory volume and compliance function. VEGFR2 and FGFR1 activation in PCECs therefore increases MMP14-dependent bioavailability of EGFR ligands to initiate and sustain alveologenesis.


Assuntos
Fatores de Crescimento Endotelial/metabolismo , Pulmão/citologia , Pulmão/fisiologia , Alvéolos Pulmonares/citologia , Animais , Células Endoteliais/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Neovascularização Fisiológica , Pneumonectomia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Regeneração , Células-Tronco/metabolismo , Técnicas de Cultura de Tecidos , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
Nature ; 468(7321): 310-5, 2010 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21068842

RESUMO

During embryogenesis, endothelial cells induce organogenesis before the development of circulation. These findings suggest that endothelial cells not only form passive conduits to deliver nutrients and oxygen, but also establish an instructive vascular niche, which through elaboration of paracrine trophogens stimulates organ regeneration, in a manner similar to endothelial-cell-derived angiocrine factors that support haematopoiesis. However, the precise mechanism by which tissue-specific subsets of endothelial cells promote organogenesis in adults is unknown. Here we demonstrate that liver sinusoidal endothelial cells (LSECs) constitute a unique population of phenotypically and functionally defined VEGFR3(+)CD34(-)VEGFR2(+)VE-cadherin(+)FactorVIII(+)CD45(-) endothelial cells, which through the release of angiocrine trophogens initiate and sustain liver regeneration induced by 70% partial hepatectomy. After partial hepatectomy, residual liver vasculature remains intact without experiencing hypoxia or structural damage, which allows study of physiological liver regeneration. Using this model, we show that inducible genetic ablation of vascular endothelial growth factor (VEGF)-A receptor-2 (VEGFR2) in the LSECs impairs the initial burst of hepatocyte proliferation (days 1-3 after partial hepatectomy) and subsequent reconstitution of the hepatovascular mass (days 4-8 after partial hepatectomy) by inhibiting upregulation of the endothelial-cell-specific transcription factor Id1. Accordingly, Id1-deficient mice also manifest defects throughout liver regeneration, owing to diminished expression of LSEC-derived angiocrine factors, including hepatocyte growth factor (HGF) and Wnt2. Notably, in in vitro co-cultures, VEGFR2-Id1 activation in LSECs stimulates hepatocyte proliferation. Indeed, intrasplenic transplantation of Id1(+/+) or Id1(-/-) LSECs transduced with Wnt2 and HGF (Id1(-/-)Wnt2(+)HGF(+) LSECs) re-establishes an inductive vascular niche in the liver sinusoids of the Id1(-/-) mice, initiating and restoring hepatovascular regeneration. Therefore, in the early phases of physiological liver regeneration, VEGFR2-Id1-mediated inductive angiogenesis in LSECs through release of angiocrine factors Wnt2 and HGF provokes hepatic proliferation. Subsequently, VEGFR2-Id1-dependent proliferative angiogenesis reconstitutes liver mass. Therapeutic co-transplantation of inductive VEGFR2(+)Id1(+)Wnt2(+)HGF(+) LSECs with hepatocytes provides an effective strategy to achieve durable liver regeneration.


Assuntos
Endotélio/metabolismo , Regeneração Hepática/fisiologia , Fígado/irrigação sanguínea , Fígado/citologia , Neovascularização Fisiológica/fisiologia , Transdução de Sinais , Animais , Proliferação de Células , Técnicas de Cocultura , Endotélio/citologia , Hepatectomia , Fator de Crescimento de Hepatócito/metabolismo , Hepatócitos/citologia , Proteína 1 Inibidora de Diferenciação/deficiência , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Camundongos , Fenótipo , Regulação para Cima , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteína Wnt2/metabolismo
16.
Circ Res ; 107(5): 667-76, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20634485

RESUMO

RATIONALE: Stromal cell-derived factor (SDF)-1/CXCR4 axis has an instrumental role during cardiac development and has been shown to be a potential therapeutic target for optimizing ventricular remodeling after acute myocardial infarction (AMI) and in ischemic cardiomyopathy. Although a therapeutic target, the specific role of cardiac myocyte CXCR4 (CM-CXCR4) expression following cardiogenesis and survival of cardiac myocyte and left ventricular remodeling after AMI is unknown. OBJECTIVE: We hypothesized that cardiac myocyte derived CXCR4 is critical for cardiac development, but it may have no role in adulthood secondary to the short transient expression of SDF-1 and the delayed expression of CM-CXCR4 following AMI. To address this issue, we developed congenital and conditional CM-CXCR4(-/-) mouse models. METHODS AND RESULTS: Two strains of CM-CXCR4(flox/flox) mice were generated by crossing CXCR4(flox/flox) mice with MCM-Cre(+/-) mouse and MLC2v-Cre(+/-) mouse on the C57BL/6J background, yielding CXCR4(flox/flox) MCM-Cre(+/-) and CXCR4(flox/flox)MLC2v-Cre(+/-) mice. Studies demonstrated recombination in both models congenitally in the MLC2v-Cre(+/-) mice and following tamoxifen administration in the MCM-Cre(+/-) mice. Surprisingly the CXCR4(flox/flox)MLC2v-Cre(+/-) are viable, had normal cardiac function, and had no evidence of ventricular septal defect. CXCR4(flox/flox)MCM(+/-) treated with tamoxifen 2 weeks before AMI demonstrated 90% decrease in cardiac CXCR4 expression 48 hours after AMI. Twenty-one days post AMI, echocardiography revealed no statistically significant difference in the wall thickness, left ventricular dimensions or ejection fraction (40.9+/-7.5 versus 34.4+/-2.6%) in CXCR4(flox/flox) mice versus CM-CXCR4(-/-) mice regardless of strategy of Cre expression. No differences in vascular density (2369+/-131 versus 2471+/-126 vessels/mm(2); CXCR4(flox/flox) versus CM-CXCR4(-/-) mouse), infarct size, collagen content, or noninfarct zone cardiac myocyte size were observed 21 days after AMI. CONCLUSIONS: We conclude that cardiac myocyte-derived CXCR4 is not essential for cardiac development and, potentially because of the mismatch in timings of peaks of SDF-1 and CXCR4, has no major role in ventricular remodeling after AMI.


Assuntos
Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Receptores CXCR4/metabolismo , Remodelação Ventricular , Animais , Miosinas Cardíacas/genética , Movimento Celular , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Modelos Animais de Doenças , Integrases/genética , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Cadeias Pesadas de Miosina/genética , Cadeias Leves de Miosina/genética , RNA Mensageiro/metabolismo , Receptores CXCR4/deficiência , Receptores CXCR4/genética , Fatores de Tempo , Transfecção , Função Ventricular Esquerda
17.
Tissue Eng Part A ; 16(7): 2157-9, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20367255

RESUMO

Angiogenesis is a fundamental prerequisite for tissue growth and thus an attractive target for cancer therapeutics. However, current efforts to halt tumor growth using antiangiogenic agents have been met with limited success. A reason for this may be that studies aimed at understanding tissue and organ formation have to this point utilized two-dimensional cell culture techniques, which fail to faithfully mimic the pathological architecture of disease in an in vivo context. In this issue of Tissue Engineering, the work of Fischbach-Teschl's group manipulate such variables as oxygen concentration, culture three-dimensionality, and cell-extracellular matrix interactions to more closely approximate the biophysical and biochemical microenvironment of tumor angiogenesis. In this article, we discuss how novel tissue engineering platforms provide a framework for the study of tumorigenesis under pathophysiologically relevant in vitro culture conditions.


Assuntos
Neoplasias/irrigação sanguínea , Neovascularização Patológica/patologia , Engenharia Tecidual/métodos , Animais , Técnicas de Cultura de Células , Transformação Celular Neoplásica/patologia , Humanos , Neoplasias/patologia , Neovascularização Fisiológica , Medicina Regenerativa , Alicerces Teciduais
18.
Ann Biomed Eng ; 33(10): 1405-10, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16240088

RESUMO

Motile cells capable of undergoing transendothelial migration, such as hematopoietic and leukemic cells, have been shown to sense and respond to a decrease in their surrounding gravity. In this study, we investigated the effects of microgravity on human leukemic cell proliferation and expression of receptors that control cell survival, such as the tyrosine kinase vascular endothelial growth factor receptor-2 (VEGFR-2). VEGFR-2 is shuttled between the nucleus and membrane, and through an autocrine activation of its ligand, VEGF-A, conveys signals that control cell survival. Autocrine or paracrine stimulation of VEGFR-2 facilitates localization of this receptor from the membrane to the nucleus--a process that results in increased survival of the leukemic cells. Here, we provide evidence that the mechanical forces altered by simulated microgravity localize and maintain VEGFR-2 in the membrane, and also block VEGF-A expression. This interferes with the shuttling of VEGFR-2 to the nucleus, resulting in a decrease in signaling and enhanced leukemic cell death. These data suggest that microgravity modulates cell survival through altering the cellular trafficking and activation state of tyrosine kinase receptors. This study has potential implications for understanding the regulation of receptor biology in pathophysiology, particularly VEGFR trafficking, thereby providing for the development of appropriate therapeutic strategies to abrogate intracrine stimulation triggered by VEGFR internalization.


Assuntos
Proliferação de Células , Sobrevivência Celular , Mecanotransdução Celular , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Simulação de Ausência de Peso , Células HL-60 , Humanos
19.
Circulation ; 111(9): 1175-83, 2005 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-15753226

RESUMO

BACKGROUND: CD133 (AC133) is a surface antigen that defines a broad population of stem cells, including myogenic and endothelial progenitors. CD133+ cells are rare in adult tissues, and the factors that support their differentiation into mature angiomyogenic cells are not known. These hurdles have hampered the use of CD133+ cells for therapeutic purposes. Because human fetal liver is a rich source of CD133+ cells, we sought to identify the growth factors that promote codifferentiation of these cells into angiogenic and myogenic cells. METHODS AND RESULTS: Human fetal liver CD133+ and CD133- cell subpopulations were cultured with 5'-azacytidine or vascular endothelial growth factor (VEGF165) and/or brain-derived nerve growth factor (BDNF). CD133+ but not CD133- cells from human fetal liver codifferentiated into spindle-shaped cells, as well as flat adherent multinucleated cells capable of spontaneous contractions in culture. The resulting spindle-shaped cells were confirmed to be endothelial cells by immunohistochemistry analysis for von Willebrand factor and by acetylated LDL uptake. Multinucleated cells were characterized as striated muscles by electron microscopy and immunohistochemistry analysis for myosin heavy chain. Presence of VEGF165 and BDNF significantly enhanced angiomyogenesis in vitro. Inoculation of cells derived from CD133+ cells, but not CD133- cells, into the ear pinna of NOD/SCID mice resulted in the formation of cardiomyocytes, as identified by immunostaining with cardiac troponin-T antibody. These cells generated electrical action potentials, detectable by ECG tracing. CONCLUSIONS: CD133 defines a population of human fetal liver cells capable of differentiating into both angiogenic and myogenic cells. Preconditioning of these CD133+ cells with VEGF165 and BDNF enhances the angiomyogenesis. CD133+ fetal liver cells ultimately may be used for therapeutic angiomyogenesis.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células Endoteliais/citologia , Endotélio Vascular/citologia , Fígado/citologia , Miócitos Cardíacos/citologia , Células-Tronco/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Antígeno AC133 , Potenciais de Ação , Animais , Antígenos CD/análise , Azacitidina/farmacologia , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Orelha Externa , Células Endoteliais/química , Perfilação da Expressão Gênica , Glicoproteínas/análise , Humanos , Lipoproteínas LDL/metabolismo , Fígado/embriologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Musculares/biossíntese , Proteínas Musculares/genética , Miócitos Cardíacos/fisiologia , Peptídeos/análise , Receptores Imunológicos/metabolismo , Receptores Depuradores , Transplante de Células-Tronco , Células-Tronco/citologia , Transplante Heterólogo , Fator de von Willebrand/análise
20.
Trends Mol Med ; 9(3): 109-17, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12657432

RESUMO

Adult bone marrow is a rich reservoir of hematopoietic and vascular stem and progenitor cells. Mobilization and recruitment of these cells are essential for tissue revascularization. Physiological stress, secondary to tissue injury or tumor growth, results in the release of angiogenic factors, including vascular endothelial growth factor (VEGF), which promotes mobilization of stem cells to the circulation, contributing to the formation of functional vasculature. VEGF interacts with its receptors, VEGFR2 and VEGFR1, expressed on endothelial and hematopoietic stem cells, and thereby promotes recruitment of these cells to neo-angiogenic sites, accelerating the revascularization process. The mobilization of stem cells from marrow is a dynamic process, regulated by shear stress imparted by blood flow, and the activation of metalloproteinases that induce the release of 'Kit ligand', facilitating egress from the marrow to the circulation. Identification of the molecular pathways that support the proliferation and differentiation of vascular stem and progenitor cells will open up new avenues for the design of clinical trials to accelerate tissue vascularization and organogenesis.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Neovascularização Fisiológica , Adenoviridae/genética , Adulto , Angiopoietina-1/sangue , Angiopoietina-1/genética , Animais , Linhagem da Célula , Movimento Celular , Quimiocinas/fisiologia , Endotélio Vascular/fisiologia , Humanos , Isquemia/terapia , Metaloproteases/fisiologia , Camundongos , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA