Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Stem Cells ; 34(6): 1664-78, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26866290

RESUMO

Hematopoietic stem/progenitor cells (HSPCs) reside in specialized bone marrow microenvironmental niches, with vascular elements (endothelial/mesenchymal stromal cells) and CXCR4-CXCL12 interactions playing particularly important roles for HSPC entry, retention, and maintenance. The functional effects of CXCL12 are dependent on its local concentration and rely on complex HSPC-niche interactions. Two Junctional Adhesion Molecule family proteins, Junctional Adhesion Molecule-B (JAM)-B and JAM-C, are reported to mediate HSPC-stromal cell interactions, which in turn regulate CXCL12 production by mesenchymal stromal cells (MSCs). Here, we demonstrate that another JAM family member, JAM-A, is most highly expressed on human hematopoietic stem cells with in vivo repopulating activity (p < .01 for JAM-A(high) compared to JAM-A(Int or Low) cord blood CD34(+) cells). JAM-A blockade, silencing, and overexpression show that JAM-A contributes significantly (p < .05) to the adhesion of human HSPCs to IL-1ß activated human bone marrow sinusoidal endothelium. Further studies highlight a novel association of JAM-A with CXCR4, with these molecules moving to the leading edge of the cell upon presentation with CXCL12 (p < .05 compared to no CXCL12). Therefore, we hypothesize that JAM family members differentially regulate CXCR4 function and CXCL12 secretion in the bone marrow niche. Stem Cells 2016;34:1664-1678.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Molécula A de Adesão Juncional/metabolismo , Receptores CXCR4/metabolismo , Antígeno AC133/metabolismo , Antígenos CD34/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Adesão Celular/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Sangue Fetal/citologia , Técnicas de Silenciamento de Genes , Células HL-60 , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Células Jurkat , Ligação Proteica/efeitos dos fármacos , Nicho de Células-Tronco/efeitos dos fármacos
2.
Br Med Bull ; 108: 25-53, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24152971

RESUMO

BACKGROUND: Blood vessel formation is fundamental to development, while its dysregulation can contribute to serious disease. Expectations are that hundreds of millions of individuals will benefit from therapeutic developments in vascular biology. MSCs are central to the three main vascular repair mechanisms. SOURCES OF DATA: Key recent published literature and ClinicalTrials.gov. AREAS OF AGREEMENT: MSCs are heterogeneous, containing multi-lineage stem and partly differentiated progenitor cells, and are easily expandable ex vivo. There is no single marker defining native MSCs in vivo. Their phenotype is strongly determined by their specific microenvironment. Bone marrow MSCs have skeletal stem cell properties. Having a perivascular/vascular location, they contribute to vascular formation and function and might be harnessed to regenerate a blood supply to injured tissues. AREAS OF CONTROVERSY: These include MSC origin, phenotype and location in vivo and their ability to differentiate into functional cardiomyocytes and endothelial cells or act as vascular stem cells. In addition their efficacy, safety and potency in clinical trials in relation to cell source, dose, delivery route, passage and timing of administration, but probably even more on the local preconditioning and the mechanisms by which they exert their effects. GROWING POINTS: Understanding the origin and the regenerative environment of MSCs, and manipulating their homing properties, proliferative ability and functionality through drug discovery and reprogramming strategies are important for their efficacy in vascular repair for regenerative medicine therapies and tissue engineering approaches. AREAS TIMELY FOR DEVELOPING RESEARCH: Characterization of MSCs' in vivo origins and biological properties in relation to their localization within tissue niches, reprogramming strategies and newer imaging/bioengineering approaches.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Medicina Regenerativa/métodos , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea/métodos , Doenças Cardiovasculares/terapia , Quimiocinas/metabolismo , Humanos , Microvasos/crescimento & desenvolvimento , Dermatopatias/terapia , Engenharia Tecidual/métodos
3.
Mol Ther Methods Clin Dev ; 30: 350-366, 2023 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-37637380

RESUMO

Neovascular age-related macular degeneration (nAMD) is a leading cause of blindness in the aging population, with vascular endothelial growth factor (VEGF) playing a key role. Treatment with recombinant anti-VEGFs is the current standard of care; however, it is only effective for 1-2 months at a time and requires re-administration. Gene therapy could pave the way for stable, long-term expression of therapeutic anti-VEGF with a single dose, reducing the frequency of treatment and potentially improving clinical outcomes. As such, we have developed OXB-203, a lentiviral-based gene therapy encoding the anti-VEGF protein aflibercept. Aflibercept derived from OXB-203 exhibited comparable in vitro binding characteristics to VEGF as recombinant aflibercept. Furthermore, its biological potency was demonstrated by the equivalent inhibition of VEGF-induced human umbilical vein endothelial cell (HUVEC) proliferation and tubule formation as recombinant aflibercept. In a rat choroidal neovascularization (CNV) model of nAMD, a single subretinal administration of OXB-203 reduced laser-induced CNV lesion areas analogous to an intravitreal bolus of recombinant aflibercept. Finally, in a head-to-head comparative study, aflibercept derived from OXB-203 was shown to be expressed at significantly higher levels in ocular tissues than from an AAV8-aflibercept vector following a single subretinal delivery to rats. These findings support the therapeutic potential of OXB-203 for the management of nAMD.

4.
Br J Haematol ; 157(3): 299-311, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22324374

RESUMO

The bone marrow contains specific microenvironmental stem cell niches that maintain haemopoiesis. CXCL12-expressing mesenchymal stromal cells are closely associated with the bone marrow sinusoidal endothelia, forming key elements of the haemopoietic stem cell niche, yet their ability to regulate endothelial function is not clearly defined. Given that the murine nestin(+) cell line, MS-5, provides a clonal surrogate bone marrow stromal niche capable of regulating both murine and human primitive haemopoietic stem/progenitor cell (HSC/HPC) fate in vitro, we hypothesized that MS-5 cells might also support new blood vessel formation and function. Here, for the first time, we demonstrate that this is indeed the case. Using proteome arrays, we identified HSC/HPC active angiogenic factors that are preferentially secreted by haemopoietic supportive nestin(+) MS-5 cells, including CXCL12 (SDF-1), NOV (CCN3), HGF, Angiopoietin-1 and CCL2 (MCP-1). Concentrating on CXCL12, we confirmed its presence in MS-5 conditioned media and demonstrated that its antagonist in receptor binding, AMD-3100, which mobilizes HSC/HPCs and endothelial progenitors from bone marrow, could significantly reduce MS-5 mediated human vasculogenesis in vitro, principally by regulating human endothelial cell migration. Thus, the clonal nestin(+) MS-5 murine bone marrow stromal cell line not only promotes human haemopoiesis but also induces human vasculogenesis, with CXCL12 playing important roles in both processes.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica/fisiologia , Indutores da Angiogênese/metabolismo , Animais , Células da Medula Óssea/fisiologia , Comunicação Celular/fisiologia , Linhagem Celular , Movimento Celular/fisiologia , Proliferação de Células , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/fisiologia , Técnicas de Cocultura , Meios de Cultivo Condicionados , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Humanos , Camundongos , Proteômica/métodos
5.
Angiogenesis ; 14(3): 381-92, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21720855

RESUMO

As umbilical cord blood (UCB) is a rich source of endothelial colony-forming cells (ECFC), our aim was twofold: (1) to examine potential obstetric selection criteria for achieving the highest ECFC yields from UCB units, and (2) to determine whether transient storage temperatures of fresh UCB and cryopreservation of UCB units affected ECFC yield and function. ECFC quality was assessed before and after cryopreservation by their clonogenic proliferative potential. Of the 20 factors examined, placental weight was the only statistically significant obstetric factor that predicted ECFC frequency in UCB. Studies on the effects of storage revealed that transient storage of fresh UCB at 4°C reduced ECFC yield compared with storage at 22°C, while cryopreservation of UCB MNCs significantly reduced ECFC recoveries. To our knowledge, this is the first demonstration that placental weight and temperature of storage prior to processing or culture have significant effects on ECFC frequency in UCB. Our studies further support the evidence that cryopreservation of UCB MNCs compromises ECFC recovery.


Assuntos
Preservação de Sangue , Criopreservação , Parto Obstétrico , Células Endoteliais/citologia , Sangue Fetal/citologia , Células-Tronco/citologia , Células Endoteliais/metabolismo , Feminino , Sangue Fetal/metabolismo , Humanos , Recém-Nascido , Masculino , Tamanho do Órgão , Placenta/citologia , Placenta/metabolismo , Gravidez , Células-Tronco/metabolismo , Fatores de Tempo
6.
Sci Rep ; 7: 44137, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28276476

RESUMO

Circulating endothelial colony forming cells (ECFCs) contribute to vascular repair where they are a target for therapy. Since ECFC proliferative potential is increased in cord versus peripheral blood and to define regulatory factors controlling this proliferation, we compared the miRNA profiles of cord blood and peripheral blood ECFC-derived cells. Of the top 25 differentially regulated miRNAs selected, 22 were more highly expressed in peripheral blood ECFC-derived cells. After validating candidate miRNAs by q-RT-PCR, we selected miR-193a-3p for further investigation. The miR-193a-3p mimic reduced cord blood ECFC-derived cell proliferation, migration and vascular tubule formation, while the miR-193a-3p inhibitor significantly enhanced these parameters in peripheral blood ECFC-derived cells. Using in silico miRNA target database analyses combined with proteome arrays and luciferase reporter assays of miR-193a-3p mimic treated cord blood ECFC-derived cells, we identified 2 novel miR-193a-3p targets, the high mobility group box-1 (HMGB1) and the hypoxia upregulated-1 (HYOU1) gene products. HMGB1 silencing in cord blood ECFC-derived cells confirmed its role in regulating vascular function. Thus, we show, for the first time, that miR-193a-3p negatively regulates human ECFC vasculo/angiogenesis and propose that antagonising miR-193a-3p in less proliferative and less angiogenic ECFC-derived cells will enhance their vasculo/angiogenic function.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Regulação para Baixo/fisiologia , Células Endoteliais/metabolismo , Proteína HMGB1/biossíntese , MicroRNAs/metabolismo , Células Endoteliais/citologia , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/metabolismo , Proteína HMGB1/genética , Humanos , MicroRNAs/genética
7.
Hypertension ; 68(3): 749-59, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27456522

RESUMO

Offspring of hypertensive pregnancies are more likely to have microvascular rarefaction and increased blood pressure in later life. We tested the hypothesis that maternal angiogenic profile during a hypertensive pregnancy is associated with fetal vasculogenic capacity and abnormal postnatal microvascular remodeling. Infants (n=255) born after either hypertensive or normotensive pregnancies were recruited for quantification of postnatal dermal microvascular structure at birth and 3 months of age. Vasculogenic cell potential was assessed in umbilical vein endothelial cells from 55 offspring based on in vitro microvessel tube formation and proliferation assays. Maternal angiogenic profile (soluble fms-like tyrosine kinase-1, soluble endoglin, vascular endothelial growth factor, and placental growth factor) was measured from postpartum plasma samples to characterize severity of pregnancy disorder. At birth, offspring born after hypertensive pregnancy had similar microvessel density to those born after a normotensive pregnancy, but during the first 3 postnatal months, they had an almost 2-fold greater reduction in total vessel density (-17.7±16.4% versus -9.9±18.7%; P=0.002). This postnatal loss varied according to the vasculogenic capacity of the endothelial cells of the infant at birth (r=0.49; P=0.02). The degree of reduction in both in vitro and postnatal in vivo vascular development was proportional to levels of antiangiogenic factors in the maternal circulation. In conclusion, our data indicate that offspring born to hypertensive pregnancies have reduced vasculogenic capacity at birth that predicts microvessel density loss over the first 3 postnatal months. Degree of postnatal microvessel reduction is proportional to levels of antiangiogenic factors in the maternal circulation at birth.


Assuntos
Células Endoteliais/metabolismo , Hipertensão Induzida pela Gravidez/fisiopatologia , Microvasos/crescimento & desenvolvimento , Resultado da Gravidez , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto , Estudos de Coortes , Feminino , Desenvolvimento Fetal/fisiologia , Humanos , Lactente , Recém-Nascido , Fator de Crescimento Placentário/metabolismo , Pré-Eclâmpsia/fisiopatologia , Valor Preditivo dos Testes , Gravidez , Proteínas da Gravidez/sangue , Nascimento Prematuro/etiologia , Nascimento Prematuro/fisiopatologia , Estudos Retrospectivos , Medição de Risco , Veias Umbilicais/embriologia
8.
Stem Cells Dev ; 23(22): 2730-43, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24940843

RESUMO

Proangiogenic factors, vascular endothelial growth factor (VEGF), and fibroblast growth factor-2 (FGF-2) prime endothelial cells to respond to "hematopoietic" chemokines and cytokines by inducing/upregulating expression of the respective chemokine/cytokine receptors. Coculture of human endothelial colony forming cell (ECFC)-derived cells with human stromal cells in the presence of VEGF and FGF-2 for 14 days resulted in upregulation of the "hematopoietic" chemokine CXCL12 and its CXCR4 receptor by day 3 of coculture. Chronic exposure to the CXCR4 antagonist AMD3100 in this vasculo/angiogenesis assay significantly reduced vascular tubule formation, an observation recapitulated by delayed AMD3100 addition. While AMD3100 did not affect ECFC-derived cell proliferation, it did demonstrate a dual action. First, over the later stages of the 14-day cocultures, AMD3100 delayed tubule organization into maturing vessel networks, resulting in enhanced endothelial cell retraction and loss of complexity as defined by live cell imaging. Second, at earlier stages of cocultures, we observed that AMD3100 significantly inhibited the integration of exogenous ECFC-derived cells into established, but immature, vascular networks. Comparative proteome profiler array analyses of ECFC-derived cells treated with AMD3100 identified changes in expression of potential candidate molecules involved in adhesion and/or migration. Blocking antibodies to CD31, but not CD146 or CD166, reduced the ECFC-derived cell integration into these extant vascular networks. Thus, CXCL12 plays a key role not only in endothelial cell sensing and guidance, but also in promoting the integration of ECFC-derived cells into developing vascular networks.


Assuntos
Vasos Sanguíneos/fisiologia , Quimiocina CXCL12/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Sistema Hematopoético/metabolismo , Sistema Hematopoético/fisiologia , Benzilaminas , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Técnicas de Cocultura/métodos , Ciclamos , Células Endoteliais/efeitos dos fármacos , Sistema Hematopoético/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Humanos , Receptores CXCR4/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Regen Med ; 3(6): 863-76, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18947309

RESUMO

Endothelial progenitor cells (EPCs) are derived from the bone marrow (BM) and peripheral blood (PB), contributing to tissue repair in various pathological conditions via the formation of new blood vessels, that is, neovascularization. EPCs can be mobilized into the circulation in response to growth factors and cytokines released following stimuli such as vascular trauma, wounding and cancer. EPCs are involved in vasculogenesis during embryogenesis, but are now recognized to have a significant bearing upon disease outcome through their contribution to neovascularization in a variety of pathological states in adulthood. EPCs exist in very small numbers, especially in circulating blood in adults where they only account for 0.01% of all cells. We discuss the contribution and potential therapeutic applications of EPCs in disease, also noting the prognostic value of PB EPC numbers, especially in heart disease and cancer.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Células Endoteliais/citologia , Células-Tronco/citologia , Animais , Movimento Celular , Humanos , Neoplasias/terapia , Transplante de Células-Tronco
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa