Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Intervalo de año de publicación
1.
Eur Surg Res ; 58(3-4): 109-120, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28056458

RESUMEN

BACKGROUND: A prolonged inflammatory phase is seen in aberrant wound healing and in chronic wounds. Macrophages are central to wound healing. Distinct macrophage subtypes have differing roles both in initial inflammation and in later tissue repair. Broadly, these cells can be divided into M1 and M2 macrophages. M2 macrophage proliferation and differentiation is regulated by colony-stimulating factor 1 (CSF-1) signalling and can be blocked by GW2580, a competitive cFMS kinase inhibitor, thereby allowing for analysis of the effect of M2 blockade on progression of surgical wounds. MATERIALS AND METHODS: Macrophage Fas-induced apoptosis (MaFIA) transgenic mice with a macrophage-specific promoter used to express green fluorescent protein (GFP) were used to allow for cell tracking. The animals were treated by oral gavage with GW2580. Surgical wounds were created and harvested after 2 weeks for analysis. RESULTS: GW2580-treated mice had significantly more GFP+ cells in the surgical scar than vehicle-treated animals (GW2580, 68.0 ± 3.1%; vehicle, 42.8 ± 1.7%; p < 0.001), and GW2580 treatment depleted CD206+ M2 macrophages in the scar (GW2580, 1.4%; vehicle, 19.3%; p < 0.001). Treated animals showed significantly higher numbers of neutrophils (vehicle, 18.0%; GW2580, 51.3%; p < 0.01) and M1 macrophages (vehicle, 3.8%; GW2580, 12.8%; p < 0.01) in the scar compared to vehicle-treated animals. The total collagen content in the area of the scar was decreased in animals treated with GW2580 as compared to those treated with vehicle alone (GW2580, 67.1%; vehicle, 79.9%; p < 0.005). CONCLUSIONS: Depletion of M2 macrophages in surgical wounds via CSF-1 signalling blockade leads to persistent inflammation, with an increase in neutrophils and M1 macrophages and attenuated collagen deposition.


Asunto(s)
Macrófagos/fisiología , Herida Quirúrgica/inmunología , Cicatrización de Heridas/inmunología , Animales , Anisoles , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pirimidinas
2.
Stem Cells ; 34(5): 1354-68, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26840832

RESUMEN

Disorders affecting smooth muscle structure/function may require technologies that can generate large scale, differentiated and contractile smooth muscle cells (SMC) suitable for cell therapy. To date no clonal precursor population that provides large numbers of differentiated SMC in culture has been identified in a rodent. Identification of such cells may also enhance insight into progenitor cell fate decisions and the relationship between smooth muscle precursors and disease states that implicate differentiated SMC. In this study, we used classic clonal expansion techniques to identify novel self-renewing Islet 1 (Isl-1) positive primitive progenitor cells (PPC) within rat bone marrow that exhibited canonical stem cell markers and preferential differentiation towards a smooth muscle-like fate. We subsequently used molecular tagging to select Isl-1 positive clonal populations from expanded and de novo marrow cell populations. We refer to these previously undescribed cells as the PPC given its stem cell marker profile, and robust self-renewal capacity. PPC could be directly converted into induced smooth muscle cells (iSMC) using single transcription factor (Kruppel-like factor 4) knockdown or transactivator (myocardin) overexpression in contrast to three control cells (HEK 293, endothelial cells and mesenchymal stem cells) where such induction was not possible. iSMC exhibited immuno- and cytoskeletal-phenotype, calcium signaling profile and contractile responses similar to bona fide SMC. Passaged iSMC could be expanded to a scale sufficient for large scale tissue replacement. PPC and reprogramed iSMC so derived may offer future opportunities to investigate molecular, structure/function and cell-based replacement therapy approaches to diverse cardiovascular, respiratory, gastrointestinal, and genitourinary diseases that have as their basis smooth muscle cell functional aberrancy or numerical loss. Stem Cells 2016;34:1354-1368.


Asunto(s)
Reprogramación Celular , Proteínas con Homeodominio LIM/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Miocitos del Músculo Liso/citología , Factores de Transcripción/metabolismo , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Proliferación Celular , Autorrenovación de las Células , Separación Celular , Células Cultivadas , Células Clonales , Silenciador del Gen , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Fenotipo , Ratas Endogámicas F344 , Telomerasa/metabolismo , Transactivadores/metabolismo
3.
Stem Cells ; 33(9): 2726-37, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25969127

RESUMEN

Mesenchymal stem cells (MSCs) are currently under investigation as tools to preserve cardiac structure and function following acute myocardial infarction (AMI). However, concerns have emerged regarding safety of acute intracoronary (IC) MSC delivery. This study aimed to characterize innate prothrombotic activity of MSC and identify means of its mitigation toward safe and efficacious therapeutic IC MSC delivery post-AMI. Expression of the initiator of the coagulation cascade tissue factor (TF) on MSC was detected and quantified by immunofluorescence, FACS, and immunoblotting. MSC-derived TF antigen was catalytically active and capable of supporting thrombin generation in vitro. Addition of MSCs to whole citrated blood enhanced platelet thrombus deposition on collagen at arterial shear, an effect abolished by heparin coadministration. In a porcine AMI model, IC infusion of 25 × 10(6) MSC during reperfusion was associated with a decrease in coronary flow reserve but not when coadministered with an antithrombin agent (heparin). Heparin reduced MSC-associated thrombosis incorporating platelets and VWF within the microvasculature. Heparin-assisted therapeutic MSC delivery also reduced apoptosis in the infarct border zone at 24 hours, significantly improved infarct size, left ventricular (LV) ejection fraction, LV volumes, wall motion, and attenuated histologic evidence of scar formation at 6 weeks post-AMI. Heparin alone or heparin-assisted fibroblast control cell delivery had no such effect. Procoagulant TF activity of therapeutic MSCs is associated with reductions in myocardial perfusion when delivered IC may be successfully managed by heparin coadministration. This study highlights an important mechanistic insight into safety concerns associated with therapeutic IC MSC delivery for AMI.


Asunto(s)
Vasos Coronarios/metabolismo , Fibrinolíticos/uso terapéutico , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Microvasos/metabolismo , Tromboplastina/metabolismo , Animales , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/fisiología , Médula Ósea/metabolismo , Células Cultivadas , Vasos Coronarios/patología , Femenino , Fibrinolíticos/farmacología , Humanos , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Microvasos/efectos de los fármacos , Microvasos/patología , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Porcinos
5.
Burns ; 41(3): 548-57, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25234958

RESUMEN

The use of cell therapy to improve burn wound healing is limited as a validated cell source is not rapidly available after injury. Progenitor cells have shown potential to drive the intrinsic wound regeneration. Two sources of cells, allogeneic mesenchymal stem cells (MSC) and autologous culture modified monocytes (CMM), were assessed for their ability to influence burn wound healing. Both could be widely available shortly after injury. Cells were delivered in a fibrin matrix following contact burns in a porcine burns model. Application of MSC significantly decreased the area of unhealed burn compared to CMM or delivery matrix alone (6% MSC, 27% CMM, 24% Matrix, p<0.001). MSC treated wounds showed histological evidence of improved wound healing with increased collagen content (MSC 49%, CMM 42%, p<0.01), increased epidermal area (MSC 8.8%, CMM 6.1%, p<0.01) and dermal thickness (MSC 1108 µm, CMM 1007 µm, p<0.01) compared to CMM treated wounds. Labelled MSC and CMM were identified in the wounds after 2 weeks by immunohistochemistry and FACS. A single application of allogeneic MSC improves the rate of burn wound healing and improves the histological appearance of the burn wound. These cells show potential as a cell therapy that is rapidly available following burn.


Asunto(s)
Quemaduras/terapia , Trasplante de Células Madre Mesenquimatosas , Monocitos/trasplante , Piel/patología , Cicatrización de Heridas , Animales , Quemaduras/patología , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas/trasplante , Femenino , Sus scrofa , Porcinos , Trasplante Homólogo
6.
Lab Anim Res ; 31(4): 174-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26755920

RESUMEN

Procedures involving complex surgical techniques in rats, such as placement of abdominal aortic graft require extended duration of surgical anesthesia, which often can be achieved by repeated administrations of xylazine-ketamine combination. However such repeated anesthetic administration, in addition to being technically challenging, may be associated with potential adverse events due to cumulative effects of anesthesia. We report here the feasibility of using urethane at low dose (~1/10 the recommended anesthetic dose) in combination with a xylazine-ketamine mix to achieve an extended duration of surgical anesthesia in rats. The anesthesia induction phase was quick and smooth with an optimal phase of surgical anesthesia achieved for up to 90 minutes, which was significantly higher compared to that achieved with use of only xylazine-ketamine combination. The rectal temperature, heart rate and respiratory rate were within the physiological range with an uneventful recovery phase. Post surgery the rats were followed up to 3 months without any evidence of tumor or any other adverse effects related to the use of the urethane anesthetic combination. We conclude that low dose urethane can be effectively used in combination with xylazine and ketamine to achieve extended duration of surgical anesthesia up to 90 minutes in rats.

7.
Biomaterials ; 35(32): 9012-22, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25096850

RESUMEN

Site specific targeting remains elusive for gene and stem cell therapies in the cardiovascular field. One promising option involves use of devices that deliver larger and more sustained cell/gene payloads to specific disease sites using the versatility of percutaneous vascular access technology. Smooth muscle cells (SMCs) engineered to deliver high local concentrations of an angiogenic molecule (VEGF) were placed in an intravascular cell delivery device (ICDD) in a porcine model of chronic total occlusion (CTO) involving ameroid placement on the proximal left circumflex (LCx) artery. Implanted SMC were retained within the ICDD and were competent for VEGF production in vitro and in vivo. Following implantation, micro-CT analyses revealed that ICDD-VEGF significantly enhanced vasa vasora microvessel density with a concomitant increase in tissue VEGF protein levels and formation of endothelial cell colonies suggesting increased angiogenic potential. ICDD-VEGF markedly enhanced regional blood flow determined by microsphere and contrast CT analysis translating to a functional improvement in regional wall motion and global left ventricular (LV) systolic and diastolic function. Our data indicate robust, clinically relevant angiogenesis can be achieved in a human scale porcine chronic vascular occlusion model following ICDD-VEGF-based delivery of angiogenic cells. This may have implications for percutaneous delivery of numerous therapeutic factors promoting creation of microvascular bypass networks in chronic vaso-occlusive diseases.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Sistemas de Liberación de Medicamentos/métodos , Enfermedades Vasculares/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Humanos , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Porcinos , Ingeniería de Tejidos
8.
PLoS One ; 8(2): e57230, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23437346

RESUMEN

Monocyte/macrophages are implicated in initiation of angiogenesis, tissue/organ perfusion and atherosclerosis biology. We recently showed that chemokine receptor CX(3)CR1 is an essential regulator of monocyte/macrophage derived smooth muscle cell differentiation in the vessel wall after injury. Here we hypothesised the contribution of CX(3)CR1- CX(3)CL1 interaction to in vivo neovascularization and studied the functional consequences of genetic and pharmacologic targeting of CX(3)CR1 in formation, maturation and maintenance of microvascular integrity. Cells functionally deficient in CX(3)CR1 lacked matrix tunnelling and tubulation capacity in a 3D Matrigel assay. These morphogenic and cytokinetic responses were driven by CX(3)CL1-CX(3)CR1 interaction and totally abrogated by a Rho antagonist. To evaluate the role of CX(3)CR1 system in vivo, Matrigel plugs were implanted in competent CX(3)CR1(+/gfp) and functionally deficient CX(3)CR1(gfp/gfp) mice. Leaky microvessels (MV) were formed in the Matrigel implanted in CX(3)CR1(gfp/gfp) but not in CX(3)CR1(+/gfp) mice. In experimental plaque neovascularization immature MV phenotype was observed in CX(3)CR1(gfp/gfp) mice, lacking CX(3)CR1 positive smooth muscle-like cells, extracellular collagen and basement membrane (BM) laminin compared to competent CX(3)CR1(+/gfp) mice. This was associated with increased extravasation of platelets into the intima of CX(3)CR1(gfp/gfp) but not functionally competent CX(3)CR1 mice. Pharmacologic targeting using CX(3)CR1 receptor antagonist in wild type mice resulted in formation of plaque MV with poor BM coverage and a leaky phenotype. Our data indicate a hitherto unrecognised role for functional CX(3)CR1 in Matrigel and experimental plaque neovascularization in vivo, which may buttress MV collectively in favour of a more stable non-leaky phenotype.


Asunto(s)
Endotelio Vascular/metabolismo , Macrófagos/metabolismo , Microvasos/metabolismo , Monocitos/metabolismo , Receptores de Quimiocina/genética , Animales , Plaquetas/efectos de los fármacos , Plaquetas/patología , Receptor 1 de Quimiocinas CX3C , Permeabilidad Capilar , Movimiento Celular/efectos de los fármacos , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Colágeno/genética , Colágeno/metabolismo , Combinación de Medicamentos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Expresión Génica/efectos de los fármacos , Genes Reporteros , Proteínas Fluorescentes Verdes , Laminina/genética , Laminina/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microvasos/efectos de los fármacos , Microvasos/patología , Monocitos/efectos de los fármacos , Monocitos/patología , Neovascularización Patológica , Péptidos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteoglicanos , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/metabolismo
9.
Eur Heart J ; 34(10): 782-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22173909

RESUMEN

AIMS: We have previously reported the cardioprotective effects of endothelial progenitor cell (EPC)-conditioned media (CM) therapy post-myocardial infarction (MI). In the present study, we have determined the insulin-like growth factor-1 (IGF-1) contribution to EPC CM effects on cardiomyocyte survival, contractility, and angiogenesis in vivo. METHODS AND RESULTS: Conditioned media from porcine EPC were administered intracoronary in the presence and absence of specific neutralizing antibodies to IGF-1 or control IgG in a porcine model of MI. X-vivo (non-conditioned) medium was used as a control. Functional, histological, and biochemical parameters were evaluated at 24 h and 8-week post-therapy. Conditioned media therapy significantly abrogated infarct zone (IZ) apoptosis, hypocontractility, and impaired left ventricular (LV) relaxation observed in control infarcts acutely (24 h post-MI). At 8 weeks following treatment, CM therapy augmented LV contractility and relaxation, IZ angiogenesis and inhibited infarct size expansion, wall expansion, and wall thinning. All of these acute and chronic beneficial effects of CM therapy were vitiated by neutralizing antibodies to IGF-1 but not by control IgG. Moreover, the addition of neutralizing IGF-1 antibody to control medium had no effect on these structural or functional changes in the heart post-treatment. CONCLUSION: Insulin-like growth factor-1 within the EPC CM mediates potent acute myocardial repair and chronic remodelling effects post-MI. These findings may provide a rationale for comparative trials of specific growth factors vs. current progenitor cell strategies.


Asunto(s)
Cardiotónicos/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Infarto del Miocardio/terapia , Miocitos Cardíacos/fisiología , Trasplante de Células Madre/métodos , Animales , Anticuerpos Neutralizantes/fisiología , Apoptosis/fisiología , Biomarcadores/metabolismo , Supervivencia Celular , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/fisiología , Células Endoteliales/trasplante , Femenino , Ventrículos Cardíacos/patología , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/inmunología , Contracción Miocárdica/fisiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Neovascularización Fisiológica/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Madre/fisiología , Sus scrofa , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/terapia
10.
Wound Repair Regen ; 20(3): 311-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22564226

RESUMEN

The first trials using progenitor cells to improve burn wound healing are beginning. However, there remains a paucity of data on patients' opinions of the source of stem cells. In this study, 279 patients attending plastic surgery/burns outpatient and medical outpatient clinics were questioned to assess willingness to accept a tissue-engineered skin product derived from a variety of sources. Levels of acceptance for the use of progenitor cells derived from these sources for treatment across a range of disease states (burns, Parkinson's disease, diabetes, and for cosmetic use) were also assessed. Overall, 80% of those questioned would accept a tissue-engineered product. Autologous cells were the preferred choice of cells (acute burns 94%, diabetes 95%, Parkinson's 93.9%). Allogeneic cells were still widely accepted (acute burns 67%, diabetes 66.7%, Parkinson's 69.2%). There was no difference observed between plastic surgical patients and medical patients in acceptance of cell therapy for burns, Parkinson's disease, or diabetes. There is good potential acceptance for the use of both autologous and allogeneic cells for the treatment of acute burns and burns' scarring as well as in diabetes and Parkinson's disease. Disease state does not appear to influence overall acceptability and choice of cells.


Asunto(s)
Actitud Frente a la Salud , Quemaduras/cirugía , Enfermedad de Parkinson/cirugía , Aceptación de la Atención de Salud/estadística & datos numéricos , Células Madre , Cirugía Plástica/ética , Ingeniería de Tejidos/ética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Quemaduras/epidemiología , Quemaduras/psicología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad de Parkinson/epidemiología , Enfermedad de Parkinson/psicología , Aceptación de la Atención de Salud/psicología , Trasplante de Piel/métodos , Cirugía Plástica/psicología , Cirugía Plástica/estadística & datos numéricos , Encuestas y Cuestionarios , Ingeniería de Tejidos/estadística & datos numéricos , Trasplante Autólogo/ética , Trasplante Autólogo/psicología , Cicatrización de Heridas , Adulto Joven
11.
Burns ; 37(8): 1386-93, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21924836

RESUMEN

The development of a good blood supply is a key step in burn wound healing and appears to be regulated in part by myeloid cells. CX3CR1 positive cells have recently been identified as myeloid cells with a potential role in angiogenesis. The role of functional CX3CR1 system in burn wound healing is not previously investigated. A 2% contact burn was induced in CX3CR1(+/gfp) and CX3CR1(gfp/gfp) mice. These transgenic mice facilitate the tracking of CX3CR1 cells (CX3CR1(+/gfp)) and allow evaluation of the consequence of CX3CR1 functional knockout (CX3CR1(gfp/gfp)) on burn wound healing. The progression of wound healing was monitored before tissue was harvested and analyzed at day 6 and day 12 for migration of CX3CR1 cells into burn wound. Deficiency of a functional CX3CR1 system resulted in decreased recruitment of CX3CR1 positive cells into the burn wound associated with decreased myeloid cell recruitment (p<0.001) and reduced maintenance of new vessels (p<0.001). Burn wound healing was prolonged (p<0.05). Our study is the first to establish a role for CX3CR1 in burn wound healing which is associated with sub-dermal angiogenesis. This chemokine receptor pathway may be attractive for therapeutic manipulation as it could increase sub dermal angiogenesis and thereby improve time to healing.


Asunto(s)
Quemaduras/metabolismo , Quemaduras/patología , Células Mieloides/patología , Neovascularización Fisiológica/fisiología , Receptores de Quimiocina/deficiencia , Piel/irrigación sanguínea , Cicatrización de Heridas/fisiología , Animales , Quemaduras/fisiopatología , Receptor 1 de Quimiocinas CX3C , Quimiocina CX3CL1/metabolismo , Células Endoteliales/patología , Inmunohistoquímica , Macrófagos/patología , Ratones , Ratones Transgénicos , Modelos Animales , Piel/citología
12.
Atherosclerosis ; 216(1): 74-82, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21333993

RESUMEN

OBJECTIVES: We examined the role of C-fms+ cells in response to vascular injury with a focus on the temporal and spatial platelet interactions, monocyte survival and proliferation within the evolving neointimal lesion and monocyte proliferation within the circulation and specified monocyte reservoir sites. Finally, we investigated the therapeutic effect of C-fms kinase inhibition (CFKI) on neointimal hyperplasia post vessel injury. METHODS AND RESULTS: We utilized murine carotid-wire injury, a transgenic C-fms reporting mouse model, confocal microscopy, shear-flow studies, specific C-fms signalling inhibition to determine the activation, mobilization and recruitment of C-fms+ monocytes in the context of early and late vessel remodelling. C-fms+ cells were recruited as early as 4h and accumulated over time in the neointima following injury. Monocyte interaction with platelet thrombus under flow and in vivo, in addition to monocyte mobilisation into the circulation post-injury was impaired by CFKI administration. Sustained inhibition of C-fms over 1-2 weeks abrogated the neointimal response but preserved re-endothelialisation post-injury. CONCLUSION: These data establish C-fms as a key regulator of the vascular response to injury and a potentially attractive therapeutic target in disease states where neointimal hyperplasia, monocyte activation and pathologic remodelling are prominent and endothelial homeostasis is desirable.


Asunto(s)
Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Monocitos/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Túnica Íntima/metabolismo , Lesiones del Sistema Vascular/metabolismo , Animales , Plaquetas/metabolismo , Antígeno CD11b/metabolismo , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Hemorreología , Hiperplasia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Monocitos/efectos de los fármacos , Monocitos/patología , Adhesividad Plaquetaria , Inhibidores de Proteínas Quinasas/farmacología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Factores de Tiempo , Túnica Íntima/efectos de los fármacos , Túnica Íntima/lesiones , Túnica Íntima/patología , Lesiones del Sistema Vascular/tratamiento farmacológico , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/patología
13.
J Mol Histol ; 42(2): 187-93, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21305341

RESUMEN

Percutaneous coronary intervention has resulted in a paradigm shift in the treatment of coronary artery disease and myocardial infarction. However, neither bare-metal stents nor polymer-coated drug-eluting stents represent ideal therapies at this time due to the undesired in-stent stenosis or delayed thrombosis. Hence there is pressing clinical need for greater understanding of the cellular mechanisms involved. It is hoped that this in turn will provide insight into designing and developing the next generation of stents. Although immunohistochemistry and immunofluorescence are appropriate tools in understanding the molecular histology, performing these techniques on stented blood vessels is technically challenging because of poor permeability of antibodies into the stented blood vessels which are embedded in methacrylate-based resins and inadequate image resolution due to autofluorescence. Hence there is a need to develop techniques which can facilitate immunohistochemistry/immunofluorescence procedures on stented blood vessel cross-sections. In this study we describe an improved protocol for processing stented porcine coronary arteries for immunostaining with smooth muscle cell, endothelial cell, monocyte and macrophage markers. We first identified the optimal conditions for resin embedding of stented artery and cross sectioned the vessels using high speed precision wafering diamond blade. The sections were then ground using two levels of water sandpaper on a Metaserve 2000 grinder to achieve the desired thickness. For immunostaining, we developed a novel deplasticization protocol which favors optimal antibody permeabilization. Our protocol not only provides feasibility of improved immunostaining of stented artery sections but also results in high quality images.


Asunto(s)
Vasos Coronarios/patología , Técnicas de Preparación Histocitológica , Stents , Actinas/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Vasos Coronarios/enzimología , Vasos Coronarios/metabolismo , Endotelio Vascular/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Macrófagos/metabolismo , Metacrilatos/química , Microscopía Fluorescente , Células Mieloides/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Adhesión en Plástico , Receptores de Quimiocina/metabolismo , Porcinos
14.
Hypertension ; 56(3): 490-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20606112

RESUMEN

Arterial hypertension is associated with increased levels of reactive oxygen species, which may scavenge endothelium-derived NO and thereby diminish its vasorelaxant effects. However, the quantitatively relevant source of reactive oxygen species is unclear. Thus, this potential pathomechanism is not yet pharmacologically targetable. Several enzymatic sources of reactive oxygen species have been suggested: uncoupled endothelial NO synthase, xanthine oxidase, and NADPH oxidases. Here we show that increased reactive oxygen species formation in aortas of 12- to 14-month-old spontaneously hypertensive rats versus age-matched Wistar Kyoto rats is inhibited by the specific NADPH oxidase inhibitor VAS2870 but neither by the xanthine oxidase inhibitor oxypurinol nor the NO synthase inhibitor N(G)-nitro-l-arginine methyl ester. NADPH oxidase activity, as well as protein expression of its catalytic subunits, NOX1 and NOX2, was increased in the aortas of spontaneously hypertensive rats, whereas the expression of NOX4 protein, the most abundant NOX isoform, was not significantly changed. Impaired acetylcholine-induced relaxation of spontaneously hypertensive rat aortas was significantly improved by VAS2870. In conclusion, NOX1 and NOX2 but not NOX4 proteins are increased in aged spontaneously hypertensive rat aortas. Importantly, these NOX isoforms, in particular, ectopic expression of NOX1 in endothelial cells, appear to affect vascular function in an NADPH oxidase inhibitor-reversible manner. NADPH oxidases may, thus, be a novel target for the treatment of systemic hypertension.


Asunto(s)
Aorta/fisiopatología , Endotelio Vascular/fisiopatología , Hipertensión/fisiopatología , Glicoproteínas de Membrana/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasas/metabolismo , Estrés Oxidativo/fisiología , Acetilcolina/farmacología , Envejecimiento , Análisis de Varianza , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Western Blotting , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Técnica del Anticuerpo Fluorescente , Hipertensión/metabolismo , Masculino , NADPH Oxidasa 1 , NADPH Oxidasa 2 , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Especies Reactivas de Oxígeno/metabolismo , Xantina Oxidasa/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 30(6): 1080-7, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20453166

RESUMEN

Cell therapy to treat vascular and cardiovascular diseases has evolved over the past decade with improved understanding of progenitor cell mobilization, recruitment, and differentiation. The beneficial effects seen in several preclinical studies have prompted translation of adult vascular progenitor therapy to clinical trials. To date, progenitor cells isolated from bone marrow and peripheral blood have been tested in the context of acute myocardial infarction and chronic ischemic cardiomyopathy, with moderate benefit. This therapeutic effect occurs despite a relatively small number of injected progenitor cells and short-term residence in the target zone. Thus, indirect benefits, such as paracrine factors released from these cells, have been suggested as significant contributors to therapeutic efficacy. Several additional vascular progenitors of endothelial, smooth muscle, mesenchymal, and cardiac origin have been identified that may contribute to vasculogenesis. Indeed, a unifying paradigm for the most effective cell therapy strategies to date appears to be robust support of angiogenesis. Here we discuss a number of progenitor cells that currently show potential as cardiovascular therapeutics, either singly or in combination. We look at emerging cell types and disease targets that may be exploited for therapeutic benefit and future strategies that may maximize clinical efficacy.


Asunto(s)
Células Madre Adultas/trasplante , Vasos Sanguíneos/patología , Enfermedades Cardiovasculares/cirugía , Trasplante de Células Madre , Adolescente , Adulto , Células Madre Adultas/metabolismo , Anciano , Anciano de 80 o más Años , Proteínas Angiogénicas/metabolismo , Animales , Biomarcadores/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiopatología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/fisiopatología , Humanos , Persona de Mediana Edad , Neovascularización Fisiológica , Comunicación Paracrina , Recuperación de la Función , Regeneración , Resultado del Tratamiento , Función Ventricular Izquierda , Adulto Joven
16.
Arterioscler Thromb Vasc Biol ; 30(3): 533-5, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20007909

RESUMEN

OBJECTIVE: To determine whether CX(3)CR1(+) bone marrow cells have the capacity for smooth muscle cell (SMC) differentiation. METHODS AND RESULTS: CX(3)CR1(+) and CX(3)CR1(-) cells were isolated from marrow of CX(3)CR1 transgenic mice and cultured in SMC differentiation media. Phenotypic and functional analyses showed only CX(3)CR1(+) bone marrow cells exhibit colony cell outgrowth with SMC-specific protein expression, calcium signaling, and contraction responses similar to mature SMC. CONCLUSIONS: CX(3)CR1 marks a bone marrow cells population that enriches for progenitors with capacity to differentiate in vitro into SMC-like cells.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Miocitos del Músculo Liso/citología , Células Madre/citología , Animales , Células de la Médula Ósea/metabolismo , Receptor 1 de Quimiocinas CX3C , Señalización del Calcio , Células Cultivadas , Macrófagos/citología , Ratones , Ratones Transgénicos , Modelos Animales , Monocitos/citología , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo
17.
FASEB J ; 24(1): 81-92, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19745110

RESUMEN

Smooth muscle cells play a major role in numerous vascular diseases that contribute to remodeling, repair after injury, and arteriogenesis, and the source of these cells is thought to lie within the vessel wall and the circulating blood. Currently, the precise origin and mechanism of differentiation of extravascular smooth muscle progenitor cells (SPCs) is unclear. We show here that the CX(3)CR1 mononuclear cell population of murine bone marrow provides a source of SPCs that contributes to smooth muscle cells within the neointimal plaque after vascular injury. Moreover, CX(3)CR1-fractalkine (FKN) interaction in vivo is essential for smooth muscle cell differentiation of bone marrow-derived progenitor cells at the vessel wall level. Functional competence of bone marrow-derived CX(3)CR1 positive cells to interact with FKN is also crucial in part for neointima formation following vascular injury. Finally, in a pure preparation of bone marrow-derived CX(3)CR1 positive cells, we show that in vitro smooth muscle cell differentiation increases markedly in the presence of FKN. Our data highlight a novel functional relationship between the myeloid and vascular systems and in the context of vascular injury and repair underscores a key chemokine-receptor pathway that may regulate cell fate when smooth muscle cell differentiation is required.


Asunto(s)
Quimiocina CX3CL1/metabolismo , Mioblastos del Músculo Liso/citología , Mioblastos del Músculo Liso/metabolismo , Receptores de Quimiocina/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Receptor 1 de Quimiocinas CX3C , Traumatismos de las Arterias Carótidas/inmunología , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Diferenciación Celular , Ensayo de Unidades Formadoras de Colonias , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mioblastos del Músculo Liso/inmunología , Receptores de Quimiocina/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Túnica Íntima/inmunología , Túnica Íntima/metabolismo , Túnica Íntima/patología
18.
Differentiation ; 78(2-3): 169-76, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19715661

RESUMEN

Lentiviral vectors encoding for identifiable marker genes controlled by lineage-specific promoters can be used to track differentiation of bone marrow progenitors into endothelial cells and/or smooth muscle cells. Human VE-Cadherin and Smoothelin-B promoters were cloned into a self-inactivating lentiviral vector (HR-VECad and HR-SMTHB) and used to drive expression of green fluorescent protein (eGFP). These constructs demonstrated specific promoter activity in mature endothelial and smooth muscle cells respectively in vitro. Lin(-) bone marrow progenitor cells (Lin(-) BMCs) in culture were used to test vector ability to track vascular differentiation. HR-VECad transduced Lin(-) BMCs were plated on collagen-coated slides and grown in endothelial media, while HR-SMTHB transduced Lin(-) BMCs were cultured on fibronectin-coated slides and grown in smooth muscle media. For in vivo differentiation assessment, lentiviral transduced Lin(-) BMCs resuspended in Matrigel were injected subcutaneously into C57BL/6J mice. Explants were evaluated for eGFP expression. Lin(-) BMCs grown in endothelial differentiation media produced groups of polygonal endothelial-like cells by days 16-21. When transduced with HR-VECad vector, these expressed eGFP in distinct cells within the colony by days 18-21, and coexpressed VE-Cadherin and eNOS. Lin(-) BMCs grown in smooth muscle differentiation media produced spindle-shaped cells between days 10-14 in culture. When transduced with the HR-SMTHB vector, these cells showed eGFP expression at approximately 12 days, which increased over time and coexpressed alphaSMA, calponin and myosin heavy chain. Within Matrigel plugs containing HR-VECad transduced cells, eGFP(+) constituted 0.4+/-0.2% of total cells. In contrast, within Matrigel plugs containing HR-SMTHB transduced cells, eGFP(+) cells constituted 0.2+/-0.1% of total cells. These data demonstrate the feasibility of selectively marking BMC populations for cell fate determination.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Lentivirus/fisiología , Animales , Linaje de la Célula , Células Cultivadas , Colágeno/metabolismo , Combinación de Medicamentos , Endotelio Vascular/citología , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Células Madre Hematopoyéticas/virología , Humanos , Laminina/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Liso/citología , Neovascularización Fisiológica , Regiones Promotoras Genéticas , Proteoglicanos/metabolismo , Transducción Genética
19.
Stem Cells Dev ; 17(5): 941-51, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18564032

RESUMEN

Administration of endothelial progenitor cells (EPC) is a promising therapy for post-infarction cardiac repair. However, the mechanisms that underlie apparent beneficial effects on myocardial remodeling are unclear. In a porcine model of acute myocardial infarction, we investigated the therapeutic effects of a mixed population of culture modified peripheral blood mononuclear cells (termed hereafter porcine EPC). Porcine EPC were isolated using methods identical to those previously adopted for harvest of EPC in human cell therapy studies. In addition the therapeutic effects of paracrine factors secreted by these cells was evaluated in vitro and in vivo. Intracoronary injection of autologous porcine EPC was associated with increased infarct territory mass and improved regional ventricular systolic function at 2 months compared to control. Treatment with conditioned media derived from autologous EPC was associated with similar improved effects on infarct territory mass and function. Histologic analysis of the infarct territory revealed significantly increased cardiomyocyte size in EPC and conditioned media treated groups, when compared to controls. A paracrine EPC effect was also verified in a pure myocardial preparation in which cardiomyocytes devoid of fibroblast, neuronal and vascular elements directly responded by increasing cell mass when exposed to the same conditioned media. Analysis of conditioned media revealed elevated levels of TGFbeta1 (human 267.3+/-11.8 pg/ml, porcine 57.1+/-6.1 pg/ml), a recognized mediator of hypertrophic signaling in the heart. Neutralizing antibodies to TGFbeta1 attenuated the pro-hypertrophic effect of conditioned media, and use of recombinant TGFbeta1 added to fresh media replicated the pro-hypertrophic effects of conditioned media in vitro. These data demonstrate the potential of paracrine factors secreted from endothelial progenitor cells to induce cardiomyocyte hypertrophy contributing to increased infarct territory LV mass, with favorable medium term effects on regional function following myocardial infarction.


Asunto(s)
Cardiomegalia/etiología , Tratamiento Basado en Trasplante de Células y Tejidos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/terapia , Comunicación Paracrina , Células Madre/citología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Bioensayo , Cardiomegalia/fisiopatología , Tamaño de la Célula , Medios de Cultivo Condicionados , Modelos Animales de Enfermedad , Células Endoteliales/citología , Citometría de Flujo , Pruebas de Función Cardíaca , Ventrículos Cardíacos/anatomía & histología , Ventrículos Cardíacos/fisiopatología , Humanos , Imagen por Resonancia Magnética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Tamaño de los Órganos , Fenotipo , Ratas , Trasplante de Células Madre , Sus scrofa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA