Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
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
2.
Nature ; 505(7481): 97-102, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24256728

RESUMO

Chemical or traumatic damage to the liver is frequently associated with aberrant healing (fibrosis) that overrides liver regeneration. The mechanism by which hepatic niche cells differentially modulate regeneration and fibrosis during liver repair remains to be defined. Hepatic vascular niche predominantly represented by liver sinusoidal endothelial cells deploys paracrine trophogens, known as angiocrine factors, to stimulate regeneration. Nevertheless, it is not known how pro-regenerative angiocrine signals from liver sinusoidal endothelial cells is subverted to promote fibrosis. Here, by combining an inducible endothelial-cell-specific mouse gene deletion strategy and complementary models of acute and chronic liver injury, we show that divergent angiocrine signals from liver sinusoidal endothelial cells stimulate regeneration after immediate injury and provoke fibrosis after chronic insult. The pro-fibrotic transition of vascular niche results from differential expression of stromal-derived factor-1 receptors, CXCR7 and CXCR4 (refs 18, 19, 20, 21), in liver sinusoidal endothelial cells. After acute injury, CXCR7 upregulation in liver sinusoidal endothelial cells acts with CXCR4 to induce transcription factor Id1, deploying pro-regenerative angiocrine factors and triggering regeneration. Inducible deletion of Cxcr7 in sinusoidal endothelial cells (Cxcr7(iΔEC/iΔEC)) from the adult mouse liver impaired liver regeneration by diminishing Id1-mediated production of angiocrine factors. By contrast, after chronic injury inflicted by iterative hepatotoxin (carbon tetrachloride) injection and bile duct ligation, constitutive FGFR1 signalling in liver sinusoidal endothelial cells counterbalanced CXCR7-dependent pro-regenerative response and augmented CXCR4 expression. This predominance of CXCR4 over CXCR7 expression shifted angiocrine response of liver sinusoidal endothelial cells, stimulating proliferation of desmin(+) hepatic stellate-like cells and enforcing a pro-fibrotic vascular niche. Endothelial-cell-specific ablation of either Fgfr1 (Fgfr1(iΔEC/iΔEC)) or Cxcr4 (Cxcr4(iΔEC/iΔEC)) in mice restored the pro-regenerative pathway and prevented FGFR1-mediated maladaptive subversion of angiocrine factors. Similarly, selective CXCR7 activation in liver sinusoidal endothelial cells abrogated fibrogenesis. Thus, we demonstrate that in response to liver injury, differential recruitment of pro-regenerative CXCR7-Id1 versus pro-fibrotic FGFR1-CXCR4 angiocrine pathways in vascular niche balances regeneration and fibrosis. These results provide a therapeutic roadmap to achieve hepatic regeneration without provoking fibrosis.


Assuntos
Cirrose Hepática/patologia , Regeneração Hepática/fisiologia , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Transdução de Sinais , Doença Aguda , Animais , Ductos Biliares/cirurgia , Tetracloreto de Carbono , Doença Hepática Crônica Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Crônica Induzida por Substâncias e Drogas/patologia , Quimiocina CXCL12/metabolismo , Doença Crônica , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Ligadura , Camundongos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo
3.
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
4.
Blood ; 120(6): 1344-7, 2012 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-22709690

RESUMO

Transplantation of ex vivo expanded human umbilical cord blood cells (hCB) only partially enhances the hematopoietic recovery after myelosuppressive therapy. Incubation of hCB with optimal combinations of cytokines and niche cells, such as endothelial cells (ECs), could augment the efficiency of hCB expansion. We have devised an approach to cultivate primary human ECs (hECs) in serum-free culture conditions. We demonstrate that coculture of CD34(+) hCB in direct cellular contact with hECs and minimal concentrations of thrombopoietin/Kit-ligand/Flt3-ligand resulted in a 400-fold expansion of total hematopoietic cells, 150-fold expansion of CD45(+)CD34(+) progenitor cells, and 23-fold expansion of CD45(+) Lin(-)CD34(hi+)CD45RA(-)CD49f(+) stem and progenitor cells over a 12-day period. Compared with cytokines alone, coculture of hCB with hECs permitted greater expansion of cells capable of multilineage engraftment and serial transplantation, hallmarks of long-term repopulating hematopoietic stem cells. Therefore, hECs establish a cellular platform for expansion of hematopoietic stem and progenitor cells and treatment of hematologic disorders.


Assuntos
Vasos Sanguíneos/citologia , Proliferação de Células , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/fisiologia , Nicho de Células-Tronco/fisiologia , Alicerces Teciduais , Animais , Técnicas de Cultura de Células/métodos , Células Cultivadas , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sangue Fetal/fisiologia , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Engenharia Tecidual/métodos
5.
Cells ; 11(19)2022 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-36230937

RESUMO

Alpha-2-macroglobulin (A2M) is a protease inhibitor that regulates extracellular matrix (ECM) stability and turnover. Here, we show that A2M is expressed by endothelial cells (ECs) from human eye choroid. We demonstrate that retinal pigment epithelium (RPE)-conditioned medium induces A2M expression specifically in ECs. Experiments using chemical inhibitors, blocking antibodies, and recombinant proteins revealed a key role of VEGF-A in RPE-mediated A2M induction in ECs. Furthermore, incubation of ECs with RPE-conditioned medium reduces matrix metalloproteinase-2 gelatinase activity of culture supernatants, which is partially restored after A2M knockdown in ECs. We propose that dysfunctional RPE or choroidal blood vessels, as observed in retinal diseases such as age-related macular degeneration, may disrupt the crosstalk mechanism we describe here leading to alterations in the homeostasis of choroidal ECM, Bruch's membrane and visual function.


Assuntos
alfa 2-Macroglobulinas Associadas à Gravidez , Epitélio Pigmentado da Retina , Anticorpos Bloqueadores , Meios de Cultivo Condicionados , Células Endoteliais , Feminino , Gelatinases , Humanos , Metaloproteinase 2 da Matriz , Gravidez , Inibidores de Proteases , Proteínas Recombinantes , Fatores de Transcrição , Fator A de Crescimento do Endotélio Vascular
6.
Int J Cancer ; 128(3): 715-25, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20725999

RESUMO

Hyperthermic intraperitoneal chemotherapy (HIPEC) has shown promise in treatment of ovarian carcinosis. Despite its efficiency for the treatment of peritoneal carcinosis from digestive tract neoplasia, it has failed to demonstrate significant benefit in ovarian cancers. It is therefore essential to understand the mechanism underlying resistance to HIPEC in ovarian cancers. Mesenchymal stem cells (MSC) play an important role in the development of ovarian cancer metastasis and resistance to treatments. A recent study suggests that MSCs may be cytotoxic for cancer cells upon heat shock. In contrast, we describe the protective role of MSC against hyperthermia. Using cytokine arrays we determined that the tumor associated MSC (TAMC) secrete pro-tumoral cytokines. We studied the effect of hyperthermia in co-culture setting of TAMC or BM-MCS associated with ovarian cancer cell lines (SKOV3 and CaOV3) with polyvariate flow cytometry. We demonstrate that hyperthermia does not challenge survival of TAMC or bone marrow derived MSC (BM-MSC). Both TAMC and BM-MSC displayed strong protective effect inducing thermotolerance in ovarian cancer cells (OCC). Transwell experiments demonstrated the role of secreted factors. We showed that CXCL12 was inducing thermotolerance and that inhibition of CXCL12/CXCR4 interaction restored cytotoxicity of hyperthermia in co-culture experiments. Contrary to the previous published study we demonstrated that TAMC and BM-MSC co-cultured with OCC induced thermotolerance in a CXCL12 dependant manner. Targeting the interaction between stromal and cancer cells through CXCL12 inhibition might restore hyperthermia sensitivity in ovarian cancers, and thus improve HIPEC efficiency.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Neoplasias Ovarianas/patologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular , Quimiocina CXCL12/antagonistas & inibidores , Técnicas de Cocultura , Feminino , Citometria de Fluxo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Temperatura Alta , Humanos , Hipertermia Induzida , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/mortalidade , Receptores CXCR4/antagonistas & inibidores , Taxa de Sobrevida
7.
J Exp Med ; 218(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34287647

RESUMO

Chronic obstructive pulmonary disease (COPD) is marked by airway inflammation and airspace enlargement (emphysema) leading to airflow obstruction and eventual respiratory failure. Microvasculature dysfunction is associated with COPD/emphysema. However, it is not known if abnormal endothelium drives COPD/emphysema pathology and/or if correcting endothelial dysfunction has therapeutic potential. Here, we show the centrality of endothelial cells to the pathogenesis of COPD/emphysema in human tissue and using an elastase-induced murine model of emphysema. Airspace disease showed significant endothelial cell loss, and transcriptional profiling suggested an apoptotic, angiogenic, and inflammatory state. This alveolar destruction was rescued by intravenous delivery of healthy lung endothelial cells. Leucine-rich α-2-glycoprotein-1 (LRG1) was a driver of emphysema, and deletion of Lrg1 from endothelial cells rescued vascular rarefaction and alveolar regression. Hence, targeting endothelial cell biology through regenerative methods and/or inhibition of the LRG1 pathway may represent strategies of immense potential for the treatment of COPD/emphysema.


Assuntos
Células Endoteliais/patologia , Pulmão/patologia , Enfisema Pulmonar/patologia , Administração Intravenosa , Animais , Biomarcadores/metabolismo , Modelos Animais de Doenças , Células Endoteliais/transplante , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Glicoproteínas/metabolismo , Humanos , Pulmão/irrigação sanguínea , Pulmão/fisiopatologia , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Elastase Pancreática/metabolismo , Fenótipo , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Enfisema Pulmonar/genética , Enfisema Pulmonar/fisiopatologia , Índice de Gravidade de Doença , Fumar , Transcriptoma/genética
8.
Mol Ther Methods Clin Dev ; 20: 703-715, 2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33738325

RESUMO

Ex vivo hematopoietic stem and progenitor cell (HSPC) expansion platforms are under active development, designed to increase HSPC numbers and thus engraftment ability of allogeneic cord blood grafts or autologous HSPCs for gene therapies. Murine and in vitro models have not correlated well with clinical outcomes of HSPC expansion, emphasizing the need for relevant pre-clinical models. Our rhesus macaque HSPC competitive autologous transplantation model utilizing genetically barcoded HSPC allows direct analysis of the relative short and long-term engraftment ability of lentivirally transduced HSPCs, along with additional critical characteristics such as HSPC clonal diversity and lineage bias. We investigated the impact of ex vivo expansion of macaque HSPCs on the engineered endothelial cell line (E-HUVECs) platform regarding safety, engraftment of transduced and E-HUVEC-expanded HSPC over time compared to non-expanded HSPC for up to 51 months post-transplantation, and both clonal diversity and lineage distribution of output from each engrafted cell source. Short and long-term engraftment were comparable for E-HUVEC expanded and the non-expanded HSPCs in both animals, despite extensive proliferation of CD34+ cells during 8 days of ex vivo culture for the E-HUVEC HSPCs, and optimization of harvesting and infusion of HSPCs co-cultured on E-HUVEC in the second animal. Long-term hematopoietic output from both E-HUVEC expanded and unexpanded HSPCs was highly polyclonal and multilineage. Overall, the comparable HSPC kinetics of macaques to humans, the ability to study post-transplant clonal patterns, and simultaneous multi-arm comparisons of grafts without the complication of interpreting allogeneic effects makes our model ideal to test ex vivo HSPC expansion platforms, particularly for gene therapy applications.

9.
J Exp Med ; 217(6)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32196081

RESUMO

The activity and survival of retinal photoreceptors depend on support functions performed by the retinal pigment epithelium (RPE) and on oxygen and nutrients delivered by blood vessels in the underlying choroid. By combining single-cell and bulk RNA sequencing, we categorized mouse RPE/choroid cell types and characterized the tissue-specific transcriptomic features of choroidal endothelial cells. We found that choroidal endothelium adjacent to the RPE expresses high levels of Indian Hedgehog and identified its downstream target as stromal GLI1+ mesenchymal stem cell-like cells. In vivo genetic impairment of Hedgehog signaling induced significant loss of choroidal mast cells, as well as an altered inflammatory response and exacerbated visual function defects after retinal damage. Our studies reveal the cellular and molecular landscape of adult RPE/choroid and uncover a Hedgehog-regulated choroidal immunomodulatory signaling circuit. These results open new avenues for the study and treatment of retinal vascular diseases and choroid-related inflammatory blinding disorders.


Assuntos
Corioide/imunologia , Corioide/patologia , Endotélio/imunologia , Imunomodulação , Análise de Célula Única , Animais , Proliferação de Células , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Proteínas Hedgehog/metabolismo , Inflamação/genética , Mastócitos/metabolismo , Melanócitos/metabolismo , Melanócitos/patologia , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Epitélio Pigmentado da Retina/metabolismo , Transdução de Sinais , Transcrição Gênica , Proteína GLI1 em Dedos de Zinco/metabolismo
10.
Sci Immunol ; 3(19)2018 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-29330161

RESUMO

The thymus is not only extremely sensitive to damage but also has a remarkable ability to repair itself. However, the mechanisms underlying this endogenous regeneration remain poorly understood, and this capacity diminishes considerably with age. We show that thymic endothelial cells (ECs) comprise a critical pathway of regeneration via their production of bone morphogenetic protein 4 (BMP4) ECs increased their production of BMP4 after thymic damage, and abrogating BMP4 signaling or production by either pharmacologic or genetic inhibition impaired thymic repair. EC-derived BMP4 acted on thymic epithelial cells (TECs) to increase their expression of Foxn1, a key transcription factor involved in TEC development, maintenance, and regeneration, and its downstream targets such as Dll4, a key mediator of thymocyte development and regeneration. These studies demonstrate the importance of the BMP4 pathway in endogenous tissue regeneration and offer a potential clinical approach to enhance T cell immunity.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Células Endoteliais/metabolismo , Regeneração/fisiologia , Timo/metabolismo , Timo/fisiologia , Animais , Proliferação de Células/fisiologia , Células Endoteliais/fisiologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Feminino , Fatores de Transcrição Forkhead/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Linfócitos T/metabolismo , Linfócitos T/fisiologia
11.
Stem Cells Transl Med ; 6(3): 864-876, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28297579

RESUMO

Successful expansion of bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs) would benefit many HSPC transplantation and gene therapy/editing applications. However, current expansion technologies have been limited by a loss of multipotency and self-renewal properties ex vivo. We hypothesized that an ex vivo vascular niche would provide prohematopoietic signals to expand HSPCs while maintaining multipotency and self-renewal. To test this hypothesis, BM autologous CD34+ cells were expanded in endothelial cell (EC) coculture and transplanted in nonhuman primates. CD34+ C38- HSPCs cocultured with ECs expanded up to 17-fold, with a significant increase in hematopoietic colony-forming activity compared with cells cultured with cytokines alone (colony-forming unit-granulocyte-erythroid-macrophage-monocyte; p < .005). BM CD34+ cells that were transduced with green fluorescent protein lentivirus vector and expanded on ECs engrafted long term with multilineage polyclonal reconstitution. Gene marking was observed in granulocytes, lymphocytes, platelets, and erythrocytes. Whole transcriptome analysis indicated that EC coculture altered the expression profile of 75 genes in the BM CD34+ cells without impeding the long-term engraftment potential. These findings show that an ex vivo vascular niche is an effective platform for expansion of adult BM HSPCs. Stem Cells Translational Medicine 2017;6:864-876.


Assuntos
Células da Medula Óssea/citologia , Células Endoteliais/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Animais , Antígenos CD34/metabolismo , Linhagem da Célula , Proliferação de Células , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Humanos , Primatas , Fatores de Tempo
12.
Nat Commun ; 8: 15374, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28524846

RESUMO

The outer blood-retina barrier is established through the coordinated terminal maturation of the retinal pigment epithelium (RPE), fenestrated choroid endothelial cells (ECs) and Bruch's membrane, a highly organized basement membrane that lies between both cell types. Here we study the contribution of choroid ECs to this process by comparing their gene expression profile before (P5) and after (P30) the critical postnatal period when mice acquire mature visual function. Transcriptome analyses show that expression of extracellular matrix-related genes changes dramatically over this period. Co-culture experiments support the existence of a novel regulatory pathway: ECs secrete factors that remodel RPE basement membrane, and integrin receptors sense these changes triggering Rho GTPase signals that modulate RPE tight junctions and enhance RPE barrier function. We anticipate our results will spawn a search for additional roles of choroid ECs in RPE physiology and disease.


Assuntos
Membrana Basal/metabolismo , Lâmina Basilar da Corioide/metabolismo , Matriz Extracelular/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Junções Íntimas/metabolismo , Animais , Biotinilação , Barreira Hematorretiniana/metabolismo , Adesão Celular , Sobrevivência Celular , Células Cultivadas , Corioide/metabolismo , Técnicas de Cocultura , Eletrorretinografia , Feminino , Integrinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Permeabilidade , Proteína-Lisina 6-Oxidase/metabolismo , RNA Mensageiro/metabolismo , Análise de Sequência de RNA
14.
J Clin Invest ; 125(3): 1243-54, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25664855

RESUMO

Pluripotent stem cells (PSCs) represent an alternative hematopoietic stem cell (HSC) source for treating hematopoietic disease. The limited engraftment of human PSC-derived (hPSC-derived) multipotent progenitor cells (MPP) has hampered the clinical application of these cells and suggests that MPP require additional cues for definitive hematopoiesis. We hypothesized that the presence of a vascular niche that produces Notch ligands jagged-1 (JAG1) and delta-like ligand-4 (DLL4) drives definitive hematopoiesis. We differentiated hes2 human embryonic stem cells (hESC) and Macaca nemestrina-induced PSC (iPSC) line-7 with cytokines in the presence or absence of endothelial cells (ECs) that express JAG1 and DLL4. Cells cocultured with ECs generated substantially more CD34+CD45+ hematopoietic progenitors compared with cells cocultured without ECs or with ECs lacking JAG1 or DLL4. EC-induced cells exhibited Notch activation and expressed HSC-specific Notch targets RUNX1 and GATA2. EC-induced PSC-MPP engrafted at a markedly higher level in NOD/SCID/IL-2 receptor γ chain-null (NSG) mice compared with cytokine-induced cells, and low-dose chemotherapy-based selection further increased engraftment. Long-term engraftment and the myeloid-to-lymphoid ratio achieved with vascular niche induction were similar to levels achieved for cord blood-derived MPP and up to 20-fold higher than those achieved with hPSC-derived MPP engraftment. Our findings indicate that endothelial Notch ligands promote PSC-definitive hematopoiesis and production of long-term engrafting CD34+ cells, suggesting these ligands are critical for HSC emergence.


Assuntos
Endotélio Vascular/citologia , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Multipotentes/fisiologia , Animais , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/fisiologia , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Humanos , Macaca nemestrina , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Nicho de Células-Tronco
15.
J Am Coll Surg ; 195(5): 627-9, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12437248

RESUMO

BACKGROUND: Surgical skills training is an integral component of the Advanced Trauma Life Support (ATLS) Course. Teaching techniques are continuously reevaluated and updated. Recognition of recurring technical errors in the performance of cricothyroidotomy in canine models prompted this comparison to the performance of the procedure in human cadavers. STUDY DESIGN: Thirty-three ATLS physician students performed cricothyroidotomy in canine models. Ten flight nurses performed a bimonthly surgical skills practicum on similarly prepared animals. Neck specimens were excised, fixed, and later mapped by the investigators. Subsequent courses used human cadavers obtained through the Wright State University School of Medicine Anatomical Gift Program. Cricothyroidotomy sites were mapped in situ. RESULTS: In the canine models, 47 necks with 52 attempted cricothyroidotomies were inspected and mapped by the investigators. Four specimens had multiple tracheotomy sites: three had two and one had three. If these multiple attempts are excluded from analysis, 13 of the 43 cricothyroidotomies in the canine models were misplaced (30.2%). Cricothyroidotomy placement in human cadavers was correct in 27 of 28 attempts (96.4%). CONCLUSIONS: It is imperative that cricothyroidotomy, a high-risk procedure, be taught in an appropriate model to best prepare students to perform it in a life-saving situation. Placement accuracy in canine models is low. Alternative models for teaching this procedure should be considered.


Assuntos
Competência Clínica , Procedimentos Cirúrgicos Otorrinolaringológicos/educação , Traumatologia/educação , Animais , Cadáver , Cães , Humanos , Modelos Anatômicos , Modelos Animais , Procedimentos Cirúrgicos Otorrinolaringológicos/normas , Traumatologia/normas
16.
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
17.
PLoS One ; 6(10): e26918, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22046410

RESUMO

The mechanism by which hematopoietic stem and progenitor cells (HSPCs) through interaction with their niches maintain and reconstitute adult hematopoietic cells is unknown. To functionally and genetically track localization of HSPCs with their niches, we employed novel mutant loxPs, lox66 and lox71 and Cre-recombinase technology to conditionally delete c-Kit in adult mice, while simultaneously enabling GFP expression in the c-Kit-deficient cells. Conditional deletion of c-Kit resulted in hematopoietic failure and splenic atrophy both at steady state and after marrow ablation leading to the demise of the treated adult mice. Within the marrow, the c-Kit-expressing GFP(+) cells were positioned to Kit ligand (KL)-expressing niche cells. This c-Kit-mediated cellular adhesion was essential for long-term maintenance and expansion of HSPCs. These results lay the foundation for delivering KL within specific niches to maintain and restore hematopoiesis.


Assuntos
Hematopoese , Proteínas Proto-Oncogênicas c-kit/fisiologia , Nicho de Células-Tronco , Células-Tronco/fisiologia , Animais , Medula Óssea , Adesão Celular , Movimento Celular , Proliferação de Células , Células-Tronco Hematopoéticas , Camundongos , Fator de Células-Tronco , Células-Tronco/citologia
18.
Cancer Res ; 70(18): 7273-82, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20807818

RESUMO

Tumor angiogenesis is essential for malignant growth and metastasis. Bone marrow (BM)-derived endothelial progenitor cells (EPC) contribute to angiogenesis-mediated tumor growth. EPC ablation can reduce tumor growth; however, the lack of a marker that can track EPCs from the BM to tumor neovasculature has impeded progress in understanding the molecular mechanisms underlying EPC biology. Here, we report the use of transgenic mouse and lentiviral models to monitor the BM-derived compartment of the tumor stroma; this approach exploits the selectivity of the transcription factor inhibitor of DNA binding 1 (Id1) for EPCs to track EPCs in the BM, blood, and tumor stroma, as well as mature EPCs. Acute ablation of BM-derived EPCs using Id1-directed delivery of a suicide gene reduced circulating EPCs and yielded significant defects in angiogenesis-mediated tumor growth. Additionally, use of the Id1 proximal promoter to express microRNA-30-based short hairpin RNA inhibited the expression of critical EPC-intrinsic factors, confirming that signaling through vascular endothelial growth factor receptor 2 is required for EPC-mediated tumor biology. By exploiting the selectivity of Id1 gene expression in EPCs, our results establish a strategy to track and target EPCs in vivo, clarifying the significant role that EPCs play in BM-mediated tumor angiogenesis.


Assuntos
Carcinoma Pulmonar de Lewis/genética , Células Endoteliais/fisiologia , Proteína 1 Inibidora de Diferenciação/genética , Células-Tronco/fisiologia , Animais , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Galinhas , Células Endoteliais/patologia , Perfilação da Expressão Gênica , Proteína 1 Inibidora de Diferenciação/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Células-Tronco/patologia , Células Estromais/patologia , Regulação para Cima
19.
Nat Cell Biol ; 12(11): 1046-56, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20972423

RESUMO

Endothelial cells establish an instructive vascular niche that reconstitutes haematopoietic stem and progenitor cells (HSPCs) through release of specific paracrine growth factors, known as angiocrine factors. However, the mechanism by which endothelial cells balance the rate of proliferation and lineage-specific differentiation of HSPCs is unknown. Here, we demonstrate that Akt activation in endothelial cells, through recruitment of mTOR, but not the FoxO pathway, upregulates specific angiocrine factors that support expansion of CD34(-)Flt3(-) KLS HSPCs with long-term haematopoietic stem cell (LT-HSC) repopulation capacity. Conversely, co-activation of Akt-stimulated endothelial cells with p42/44 MAPK shifts the balance towards maintenance and differentiation of the HSPCs. Selective activation of Akt1 in the endothelial cells of adult mice increased the number of colony forming units in the spleen and CD34(-)Flt3(-) KLS HSPCs with LT-HSC activity in the bone marrow, accelerating haematopoietic recovery. Therefore, the activation state of endothelial cells modulates reconstitution of HSPCs through the modulation of angiocrine factors, with Akt-mTOR-activated endothelial cells supporting the self-renewal of LT-HSCs and expansion of HSPCs, whereas MAPK co-activation favours maintenance and lineage-specific differentiation of HSPCs.


Assuntos
Diferenciação Celular , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Comunicação Parácrina , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Crescimento Transformadores/metabolismo , Animais , Comunicação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Ativação Enzimática , Hematopoese , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Serina-Treonina Quinases TOR/metabolismo
20.
Cell Stem Cell ; 6(3): 251-64, 2010 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-20207228

RESUMO

Bone marrow endothelial cells (ECs) are essential for reconstitution of hematopoiesis, but their role in self-renewal of long-term hematopoietic stem cells (LT-HSCs) is unknown. We have developed angiogenic models to demonstrate that EC-derived angiocrine growth factors support in vitro self-renewal and in vivo repopulation of authentic LT-HSCs. In serum/cytokine-free cocultures, ECs, through direct cellular contact, stimulated incremental expansion of repopulating CD34(-)Flt3(-)cKit(+)Lineage(-)Sca1(+) LT-HSCs, which retained their self-renewal ability, as determined by single-cell and serial transplantation assays. Angiocrine expression of Notch ligands by ECs promoted proliferation and prevented exhaustion of LT-HSCs derived from wild-type, but not Notch1/Notch2-deficient, mice. In transgenic notch-reporter (TNR.Gfp) mice, regenerating TNR.Gfp(+) LT-HSCs were detected in cellular contact with sinusoidal ECs. Interference with angiocrine, but not perfusion, function of SECs impaired repopulation of TNR.Gfp(+) LT-HSCs. ECs establish an instructive vascular niche for clinical-scale expansion of LT-HSCs and a cellular platform to identify stem cell-active trophogens.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Transdução de Sinais , Animais , Comunicação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados , Ligantes , Camundongos , Camundongos Knockout , Receptor Notch1/deficiência , Receptor Notch1/metabolismo , Receptor Notch2/deficiência , Receptor Notch2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA