Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Glia ; 70(3): 430-450, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34626143

RESUMEN

Kallikrein related peptidase 6 (Klk6) is a secreted serine protease highly expressed in oligodendrocytes and implicated in demyelinating conditions. To gain insights into the significance of Klk6 to oligodendrocyte biology, we investigated the impact of global Klk6 gene knockout on CNS developmental myelination using the spinal cord of male and female mice as a model. Results demonstrate that constitutive loss of Klk6 expression accelerates oligodendrocyte differentiation developmentally, including increases in the expression of myelin proteins such as MBP, PLP and CNPase, in the number of CC-1+ mature oligodendrocytes, and myelin thickness by the end of the first postnatal week. Co-ordinate elevations in the pro-myelinating signaling pathways ERK and AKT, expression of fatty acid 2-hydroxylase, and myelin regulatory transcription factor were also observed in the spinal cord of 7d Klk6 knockouts. LC/MS/MS quantification of spinal cord lipids showed sphingosine and sphingomyelins to be elevated in Klk6 knockouts at the peak of myelination. Oligodendrocyte progenitor cells (OPCs)-derived from Klk6 knockouts, or wild type OPCs-treated with a Klk6 inhibitor (DFKZ-251), also showed increased MBP and PLP. Moreover, inhibition of Klk6 in OPC cultures enhanced brain derived neurotrophic factor-driven differentiation. Altogether, these findings suggest that oligodendrocyte-derived Klk6 may operate as an autocrine or paracrine rheostat, or brake, on pro-myelinating signaling serving to regulate myelin homeostasis developmentally and in the adult. These findings document for the first time that inhibition of Klk6 globally, or specifically in oligodendrocyte progenitors, is a strategy to increase early stages of oligodendrocyte differentiation and myelin production in the CNS.


Asunto(s)
Calicreínas/metabolismo , Oligodendroglía , Espectrometría de Masas en Tándem , Animales , Diferenciación Celular/fisiología , Femenino , Calicreínas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo
2.
Glia ; 69(9): 2111-2132, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33887067

RESUMEN

Excessive activation of the thrombin receptor, protease activated receptor 1 (PAR1) is implicated in diverse neuropathologies from neurodegenerative conditions to neurotrauma. PAR1 knockout mice show improved outcomes after experimental spinal cord injury (SCI), however information regarding the underpinning cellular and molecular mechanisms is lacking. Here we demonstrate that genetic blockade of PAR1 in female mice results in improvements in sensorimotor co-ordination after thoracic spinal cord lateral compression injury. We document improved neuron preservation with increases in Synapsin-1 presynaptic proteins and GAP43, a growth cone marker, after a 30 days recovery period. These improvements were coupled to signs of enhanced myelin resiliency and repair, including increases in the number of mature oligodendrocytes, their progenitors and the abundance of myelin basic protein. These significant increases in substrates for neural recovery were accompanied by reduced astrocyte (Serp1) and microglial/monocyte (CD68 and iNOS) pro-inflammatory markers, with coordinate increases in astrocyte (S100A10 and Emp1) and microglial (Arg1) markers reflective of pro-repair activities. Complementary astrocyte-neuron co-culture bioassays suggest astrocytes with PAR1 loss-of-function promote both neuron survival and neurite outgrowth. Additionally, the pro-neurite outgrowth effects of switching off astrocyte PAR1 were blocked by inhibiting TrkB, the high affinity receptor for brain derived neurotrophic factor. Altogether, these studies demonstrate unique modulatory roles for PAR1 in regulating glial-neuron interactions, including the capacity for neurotrophic factor signaling, and underscore its position at neurobiological intersections critical for the response of the CNS to injury and the capacity for regenerative repair and restoration of function.


Asunto(s)
Receptor PAR-1 , Traumatismos de la Médula Espinal , Animales , Astrocitos/metabolismo , Femenino , Ratones , Neuronas/metabolismo , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Médula Espinal/patología , Traumatismos de la Médula Espinal/metabolismo
3.
J Neurosci ; 40(7): 1483-1500, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31911460

RESUMEN

Myelin loss limits neurological recovery and myelin regeneration and is critical for restoration of function. We recently discovered that global knock-out of the thrombin receptor, also known as Protease Activated Receptor 1 (PAR1), accelerates myelin development. Here we demonstrate that knocking out PAR1 also promotes myelin regeneration. Outcomes in two unique models of myelin injury and repair, that is lysolecithin or cuprizone-mediated demyelination, showed that PAR1 knock-out in male mice improves replenishment of myelinating cells and remyelinated nerve fibers and slows early axon damage. Improvements in myelin regeneration in PAR1 knock-out mice occurred in tandem with a skewing of reactive astrocyte signatures toward a prorepair phenotype. In cell culture, the promyelinating effects of PAR1 loss of function are consistent with possible direct effects on the myelinating potential of oligodendrocyte progenitor cells (OPCs), in addition to OPC-indirect effects involving enhanced astrocyte expression of promyelinating factors, such as BDNF. These findings highlight previously unrecognized roles of PAR1 in myelin regeneration, including integrated actions across the oligodendrocyte and astroglial compartments that are at least partially mechanistically linked to the powerful BDNF-TrkB neurotrophic signaling system. Altogether, findings suggest PAR1 may be a therapeutically tractable target for demyelinating disorders of the CNS.SIGNIFICANCE STATEMENT Replacement of oligodendroglia and myelin regeneration holds tremendous potential to improve function across neurological conditions. Here we demonstrate Protease Activated Receptor 1 (PAR1) is an important regulator of the capacity for myelin regeneration across two experimental murine models of myelin injury. PAR1 is a G-protein-coupled receptor densely expressed in the CNS, however there is limited information regarding its physiological roles in health and disease. Using a combination of PAR1 knock-out mice, oligodendrocyte monocultures and oligodendrocyte-astrocyte cocultures, we demonstrate blocking PAR1 improves myelin production by a mechanism related to effects across glial compartments and linked in part to regulatory actions toward growth factors such as BDNF. These findings set the stage for development of new clinically relevant myelin regeneration strategies.


Asunto(s)
Enfermedades Desmielinizantes/fisiopatología , Regeneración Nerviosa/efectos de los fármacos , Receptor PAR-1/antagonistas & inhibidores , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Axones/patología , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/farmacología , Quelantes/toxicidad , Técnicas de Cocultivo , Cobre , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/patología , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Perfilación de la Expresión Génica , Lisofosfatidilcolinas/toxicidad , Masculino , Ratones , Ratones Noqueados , Vaina de Mielina/fisiología , Regeneración Nerviosa/fisiología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Receptor PAR-1/deficiencia , Receptor PAR-1/fisiología , Prueba de Desempeño de Rotación con Aceleración Constante , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/patología
4.
PLoS One ; 9(11): e112986, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25419565

RESUMEN

RATIONALE: The family of natriuretic peptides (NPs), including atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), exert important and diverse actions for cardiovascular and renal homeostasis. The autocrine and paracrine functions of the NPs are primarily mediated through the cellular membrane bound guanylyl cyclase-linked receptors GC-A (NPR-A) and GC-B (NPR-B). As the ligands and receptors each contain disulfide bonds, a regulatory role for the cell surface protein disulfide isomerase (PDI) was investigated. OBJECTIVE: We utilized complementary in vitro and in vivo models to determine the potential role of PDI in regulating the ability of the NPs to generate its second messenger, cyclic guanosine monophosphate. METHODS AND RESULTS: Inhibition of PDI attenuated the ability of ANP, BNP and CNP to generate cGMP in human mesangial cells (HMCs), human umbilical vein endothelial cells (HUVECs), and human aortic smooth muscle cells (HASMCs), each of which were shown to express PDI. In LLC-PK1 cells, where PDI expression was undetectable by immunoblotting, PDI inhibition had a minimal effect on cGMP generation. Addition of PDI to cultured LLC-PK1 cells increased intracellular cGMP generation mediated by ANP. Inhibition of PDI in vivo attenuated NP-mediated generation of cGMP by ANP. Surface Plasmon Resonance demonstrated modest and differential binding of the natriuretic peptides with immobilized PDI in a cell free system. However, PDI was shown to co-localize on the surface of cells with GC-A and GC-B by co-immunoprecpitation and immunohistochemistry. CONCLUSION: These data demonstrate for the first time that cell surface PDI expression and function regulate the capacity of natriuretic peptides to generate cGMP through interaction with their receptors.


Asunto(s)
Membrana Celular/metabolismo , GMP Cíclico/metabolismo , Péptidos Natriuréticos/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Animales , Bacitracina/farmacología , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Toxina Diftérica/farmacología , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Células LLC-PK1 , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Ratones Endogámicos C57BL , Microscopía Confocal , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Unión Proteica , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/genética , Interferencia de ARN , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Porcinos
5.
Circ Res ; 115(3): 364-75, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24906644

RESUMEN

RATIONALE: Macrophages regulate blood vessel structure and function in health and disease. The origins of tissue macrophages are diverse, with evidence for local production and circulatory renewal. OBJECTIVE: We identified a vascular adventitial population containing macrophage progenitor cells and investigated their origins and fate. METHODS AND RESULTS: Single-cell disaggregates from adult C57BL/6 mice were prepared from different tissues and tested for their capacity to form hematopoietic colony-forming units. Aorta showed a unique predilection for generating macrophage colony-forming units. Aortic macrophage colony-forming unit progenitors coexpressed stem cell antigen-1 and CD45 and were adventitially located, where they were the predominant source of proliferating cells in the aortic wall. Aortic Sca-1(+)CD45(+) cells were transcriptionally and phenotypically distinct from neighboring cells lacking stem cell antigen-1 or CD45 and contained a proliferative (Ki67(+)) Lin(-)c-Kit(+)CD135(-)CD115(+)CX3CR1(+)Ly6C(+)CD11b(-) subpopulation, consistent with the immunophenotypic profile of macrophage progenitors. Adoptive transfer studies revealed that Sca-1(+)CD45(+) adventitial macrophage progenitor cells were not replenished via the circulation from bone marrow or spleen, nor was their prevalence diminished by depletion of monocytes or macrophages by liposomal clodronate treatment or genetic deficiency of macrophage colony-stimulating factor. Rather adventitial macrophage progenitor cells were upregulated in hyperlipidemic ApoE(-/-) and LDL-R(-/-) mice, with adventitial transfer experiments demonstrating their durable contribution to macrophage progeny particularly in the adventitia, and to a lesser extent the atheroma, of atherosclerotic carotid arteries. CONCLUSIONS: The discovery and characterization of resident vascular adventitial macrophage progenitor cells provides new insight into adventitial biology and its participation in atherosclerosis and provokes consideration of the broader existence of local macrophage progenitors in other tissues.


Asunto(s)
Adventicia/citología , Aterosclerosis/patología , Línea Celular/inmunología , Macrófagos/citología , Células Madre/citología , Traslado Adoptivo , Adventicia/inmunología , Animales , Antígenos Ly/metabolismo , Aorta/citología , Aorta/inmunología , Apolipoproteínas E/genética , Aterosclerosis/inmunología , Femenino , Hiperlipidemias/inmunología , Hiperlipidemias/patología , Inmunofenotipificación , Antígenos Comunes de Leucocito/metabolismo , Macrófagos/metabolismo , Macrófagos/trasplante , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de LDL/genética , Bazo/citología , Células Madre/inmunología
6.
Arterioscler Thromb Vasc Biol ; 32(3): 704-11, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22223730

RESUMEN

OBJECTIVE: Tissue factor pathway inhibitor (TFPI) is the primary regulator of the tissue factor (TF) coagulation pathway. As such, TFPI may regulate the proangiogenic effects of TF. TFPI may also affect angiogenesis independently of TF, through sequences within its polybasic carboxyl terminus (TFPI C terminus [TFPIct]). We aimed to determine the effects of TFPI on angiogenesis and the role of TFPIct. METHODS AND RESULTS: Transgenic overexpression of TFPI attenuated angiogenesis in the murine hindlimb ischemia model and an aortic sprout assay. In vitro, TFPI inhibited endothelial cell migration. Peptides within the human TFPIct inhibited endothelial cell cord formation and migration in response to vascular endothelial growth factor (VEGF) 165 but not VEGF121. Furthermore, exposure to human TFPIct inhibited the phosphorylation of VEGF receptor 2 at residue Lys951, a residue known to be critical for endothelial cell migration. Finally, systemic delivery of a murine TFPIct peptide inhibited angiogenesis in the hindlimb model. CONCLUSION: These data demonstrate an inhibitory role for TFPI in angiogenesis that is, in part, mediated through peptides within its carboxyl terminus. In addition to its known role as a TF antagonist, TFPI, via its carboxyl terminus, may regulate angiogenesis by directly blocking VEGF receptor 2 activation and attenuating the migratory capacity of endothelial cells.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Isquemia/metabolismo , Lipoproteínas/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/deficiencia , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/farmacología , Animales , Sitios de Unión , Movimiento Celular , Modelos Animales de Enfermedad , Heparina/metabolismo , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Isquemia/genética , Isquemia/fisiopatología , Lipoproteínas/química , Lipoproteínas/deficiencia , Lipoproteínas/genética , Lipoproteínas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas Musculares/genética , Neovascularización Fisiológica/efectos de los fármacos , Péptidos/farmacología , Fosforilación , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Factores de Tiempo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
Circulation ; 125(4): 592-603, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22203692

RESUMEN

BACKGROUND: Hematopoiesis originates from the dorsal aorta during embryogenesis. Although adult blood vessels harbor progenitor populations for endothelial and smooth muscle cells, it is not known if they contain hematopoietic progenitor or stem cells. Here, we hypothesized that the arterial wall is a source of hematopoietic progenitor and stem cells in postnatal life. METHODS AND RESULTS: Single-cell aortic disaggregates were prepared from adult chow-fed C57BL/6 and apolipoprotein E-null (ApoE(-/-)) mice. In short- and long-term methylcellulose-based culture, aortic cells generated a broad spectrum of multipotent and lineage-specific hematopoietic colony-forming units, with a preponderance of macrophage colony-forming units. This clonogenicity was higher in lesion-free ApoE(-/-) mice and localized primarily to stem cell antigen-1-positive cells in the adventitia. Expression of stem cell antigen-1 in the aorta colocalized with canonical hematopoietic stem cell markers, as well as CD45 and mature leukocyte antigens. Adoptive transfer of labeled aortic cells from green fluorescent protein transgenic donors to irradiated C57BL/6 recipients confirmed the content of rare hematopoietic stem cells (1 per 4 000 000 cells) capable of self-renewal and durable, low-level reconstitution of leukocytes. Moreover, the predominance of long-term macrophage precursors was evident by late recovery of green fluorescent protein-positive colonies from recipient bone marrow and spleen that were exclusively macrophage colony-forming units. Although trafficking from bone marrow was shown to replenish some of the hematopoietic potential of the aorta after irradiation, the majority of macrophage precursors appeared to arise locally, suggesting long-term residence in the vessel wall. CONCLUSIONS: The postnatal murine aorta contains rare multipotent hematopoietic progenitor/stem cells and is selectively enriched with stem cell antigen-1-positive monocyte/macrophage precursors. These populations may represent novel, local vascular sources of inflammatory cells.


Asunto(s)
Aorta/citología , Aorta/crecimiento & desarrollo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Monocitos/citología , Traslado Adoptivo , Animales , Antígenos Ly/metabolismo , Apolipoproteínas E/genética , Biomarcadores/metabolismo , Trasplante de Médula Ósea , Linaje de la Célula/inmunología , Endotelio Vascular/citología , Endotelio Vascular/crecimiento & desarrollo , Proteínas Fluorescentes Verdes/genética , Inmunofenotipificación , Macrófagos/citología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Células Madre Multipotentes/citología , Células Madre Multipotentes/inmunología , Músculo Liso Vascular/citología , Músculo Liso Vascular/crecimiento & desarrollo , Quimera por Trasplante , Irradiación Corporal Total
8.
Am J Respir Cell Mol Biol ; 43(1): 35-45, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19648471

RESUMEN

Pulmonary hypertension (PH) is a commonly recognized complication of chronic respiratory disease. Enhanced vasoconstriction, pulmonary vascular remodeling, and in situ thrombosis contribute to the increased pulmonary vascular resistance observed in PH associated with hypoxic lung disease. The tissue factor pathway regulates fibrin deposition in response to acute and chronic vascular injury. We hypothesized that inhibition of the tissue factor pathway would result in attenuation of pathophysiologic parameters typically associated with hypoxia-induced PH. We tested this hypothesis using a chronic hypoxia-induced murine model of PH using mice that overexpress tissue factor pathway inhibitor (TFPI) via the smooth muscle-specific promoter SM22 (TFPI(SM22)). TFPI(SM22) mice have increased pulmonary TFPI expression compared with wild-type (WT) mice. In WT mice, exposure to chronic hypoxia (28 d at 10% O(2)) resulted in increased systolic right ventricular and mean pulmonary arterial pressures, changes that were significantly reduced in TFPI(SM22) mice. Chronic hypoxia also resulted in significant pulmonary vascular muscularization in WT mice, which was significantly reduced in TFPI(SM22) mice. Given the pleiotropic effects of TFPI, autocrine and paracrine mechanisms for these hemodynamic effects were considered. TFPI(SM22) mice had less pulmonary fibrin deposition than WT mice at 3 days after exposure to hypoxia, which is consistent with the antithrombotic effects of TFPI. Additionally, TFPI(SM22) mice had a significant reduction in the number of proliferating (proliferating cell nuclear antigen positive) pulmonary vascular smooth muscle cells compared with WT mice, which is consistent with in vitro findings. These findings demonstrate that overexpression of TFPI results in improved hemodynamic performance and reduced pulmonary vascular remodeling in a murine model of hypoxia-induced PH. This improvement is in part due to the autocrine and paracrine effects of TFPI overexpression.


Asunto(s)
Regulación de la Expresión Génica , Hipertensión Pulmonar/metabolismo , Hipoxia , Lipoproteínas/fisiología , Animales , Proliferación Celular , Hemodinámica , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Tromboplastina/metabolismo
9.
Proc Natl Acad Sci U S A ; 106(27): 11282-7, 2009 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-19541613

RESUMEN

Alternative RNA splicing may provide unique opportunities to identify drug targets and therapeutics. We identified an alternative spliced transcript for B-type natriuretic peptide (BNP) resulting from intronic retention. This transcript is present in failing human hearts and is reduced following mechanical unloading. The intron-retained transcript would generate a unique 34 amino acid (aa) carboxyl terminus while maintaining the remaining structure of native BNP. We generated antisera to this carboxyl terminus and identified immunoreactivity in failing human heart tissue. The alternatively spliced peptide (ASBNP) was synthesized and unlike BNP, failed to stimulate cGMP in vascular cells or vasorelax preconstricted arterial rings. This suggests that ASBNP may lack the dose-limiting effects of recombinant BNP. Given structural considerations, a carboxyl-terminal truncated form of ASBNP was generated (ASBNP.1) and was determined to retain the ability of BNP to stimulate cGMP in canine glomerular isolates and cultured human mesangial cells but lacked similar effects in vascular cells. In a canine-pacing model of heart failure, systemic infusion of ASBNP.1 did not alter mean arterial pressure but increased the glomerular filtration rate (GFR), suppressed plasma renin and angiotensin, while inducing natriuresis and diuresis. Consistent with its distinct in vivo effects, the activity of ASBNP.1 may not be explained through binding and activation of NPR-A or NPR-B. Thus, the biodesigner peptide ASBNP.1 enhances GFR associated with heart failure while lacking the vasoactive properties of BNP. These findings demonstrate that peptides with unique properties may be designed based on products of alternatively splicing.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Diseño de Fármacos , Riñón/efectos de los fármacos , Péptido Natriurético Encefálico/genética , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Bovinos , Perros , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Datos de Secuencia Molecular , Péptido Natriurético Encefálico/química , Péptido Natriurético Encefálico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo
10.
Endothelium ; 14(1): 1-5, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17364891

RESUMEN

Cells with an endothelial phenotype can be cultured from peripheral blood. These cells include cells of a monocytic origin with endothelial features (culture-modified mononuclear cells, CMMCs) and, at later time points, blood outgrowth endothelial cells (BOECs). Both are promising candidates for systemic cell-based cardiovascular therapies and each may have unique capabilities. Indeed, the combined use of both cell types has been shown to have synergistic therapeutic features requiring simultaneous delivery. However, the majority of preclinical studies of cell delivery have used splenectomized animals to increase systemic distribution. The goal of this study was to directly compare the distribution of these two cell types following systemic delivery in an intact animal model. A similar pattern of delivery was seen following delivery of both cell types with detection in the lung, liver, bone marrow, and spleen. Taken together, the data suggest that strategies using systemic delivery of circulation-derived cells must consider the distribution and efficiency of delivery in intact animals.


Asunto(s)
Células Endoteliales/fisiología , Células Endoteliales/trasplante , Endotelio Vascular/citología , Células Madre Hematopoyéticas/citología , Leucocitos Mononucleares/citología , Animales , Diferenciación Celular , Células Cultivadas , Células Endoteliales/citología , Masculino , Ratones , Ratones SCID , Porcinos , Distribución Tisular , Trasplante Heterólogo
11.
Thromb Haemost ; 92(3): 495-502, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15351845

RESUMEN

Tissue factor pathway inhibitor (TFPI) is a Kunitz-type protease inhibitor that regulates the extrinsic pathway of coagulation by inhibiting the factor VIIa/tissue factor (TF) catalytic complex. TFPI is expressed by both endothelial and smooth muscle cells in the vasculature and circulates at low levels. The role of local vascular TFPI in thrombosis and the development of vascular disease is unknown. To establish an experimental animal model to directly modulate smooth muscle cell-derived TFPI on the development of arterial thrombosis, transgenic mice in which a cDNA encoding murine TFPI is expressed from the murine SM22alpha promoter were generated. Expression of transgenic mRNA was 4-fold higher than the level of endogenous TFPI mRNA in arteries from transgenic mice. In situ hybridization confirmed that expression of the transgene was limited to medial vascular smooth muscle cells. Vascular TFPI activity was increased to 2 to 3-fold in carotid homogenates. There was no difference in plasma TFPI levels or hemostatic measures (PT, aPTT and tail vein bleeding times) between these mice and their wildtype littermates. In a ferric chloride-induced model of carotid thrombosis, homozygotic transgenic mice demonstrated resistance to thrombotic occlusion compared to wildtype littermates. In transgenic mice 22% occluded within 30 minutes of application while 84% of wild type mice occluded within the same time frame (p<0.01). Heterozygotic transgenic mice had an intermediate thrombotic phenotype. Taken together, these data indicated that local VSMC-specific TFPI overexpression attenuated ferric chloride-induced thrombosis without systemic or hemostatic effects. Furthermore, this transgenic mouse model should prove useful for studying the role of TFPI in the development and progression of vascular disease.


Asunto(s)
Arteriopatías Oclusivas/etiología , Modelos Animales de Enfermedad , Lipoproteínas/fisiología , Músculo Liso Vascular/citología , Trombosis/etiología , Animales , Arteriopatías Oclusivas/prevención & control , Enfermedades de las Arterias Carótidas/etiología , Terapia Genética , Genotipo , Lipoproteínas/deficiencia , Lipoproteínas/genética , Ratones , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , ARN Mensajero/análisis , Trombosis/prevención & control , Transgenes
12.
Am J Physiol Heart Circ Physiol ; 287(2): H512-7, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15072951

RESUMEN

Delivery of a heterogeneous population of cells with endothelial phenotype derived from peripheral blood has been shown to improve vascular responses after balloon arterial injury in an endothelium-dependent manner. Refinement of culture techniques has enabled the generation of outgrowth endothelial cells (OECs), a homogeneous population of distinctly endothelial cells expanded from circulating progenitor cells. The present study tested the hypothesis that OEC delivery would confer vascular protection after balloon arterial injury in a rabbit model. Rabbit peripheral blood mononuclear cells (PBMCs) were cultured in endothelial growth medium for 4-5 wk, yielding proliferative OECs with distinct endothelial phenotype (morphology, incorporation of acetylated LDL, and expression of endothelial nitric oxide synthase and caveolin-1 but not CD14). Animals underwent balloon carotid injury immediately followed by local delivery of autologous OECs for 20 min. Fluorescent-labeled OECs were detected in all layers at 4 wk, with immunostaining revealing maintenance of endothelial phenotype (von Willebrand factor-positive and RAM-11-negative) by luminal and nonluminal cells. To evaluate functional effects, additional animals received autologous OECs, saline, or freshly harvested PBMCs as noncultured cell controls by local dwell after balloon injury. Local OEC delivery improved endothelium-dependent vasoreactivity (P < 0.05 vs. saline and PBMC) and similarly reduced neointimal formation (P < 0.05 vs. saline and PBMC). These data suggest that OECs can be detected in injured arterial segments at 4 wk. Moreover, delivery of OECs confers greater vascular protection than PBMCs or saline controls and may thus offer a novel, autologous strategy to limit the response to mechanical injury.


Asunto(s)
Células Sanguíneas/citología , Traumatismos de las Arterias Carótidas/fisiopatología , Traumatismos de las Arterias Carótidas/cirugía , Trasplante de Células , Endotelio Vascular/citología , Células Madre/citología , Animales , Traumatismos de las Arterias Carótidas/etiología , Cateterismo/efectos adversos , Diferenciación Celular , Células Cultivadas , Endotelio Vascular/fisiopatología , Conejos , Trasplante Autólogo , Túnica Íntima/crecimiento & desarrollo , Vasodilatación
13.
Circulation ; 108(12): 1520-6, 2003 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-12952850

RESUMEN

BACKGROUND: Bone marrow-derived cells have been shown to contribute to endothelial replacement after vascular injury. In vitro culture of peripheral blood mononuclear cells produces cells with phenotypic characteristics of endothelium. To test the hypothesis that delivery of autologous culture-modified mononuclear cells (CMMCs) to injured arteries could attenuate the vascular response to injury, a rabbit model was studied. METHODS AND RESULTS: Rabbit peripheral blood mononuclear cells were cultured in endothelial growth media for 7 to 12 days, yielding highly proliferative cells with distinct endothelial phenotype (expressing CD31 and endothelial nitric oxide synthase and capable of acetylated LDL uptake). A rabbit model of balloon carotid injury was used to evaluate the effect of day 7 CMMC delivery on vascular responses. Animals underwent balloon injury and immediate delivery of autologous CMMCs or buffered saline by 20 minutes of local dwelling. Fluorescence-labeled CMMCs were detected in all vessel layers 4 weeks after delivery. Colonies of cells that localized to the lumen and stained for endothelial markers were also identified. Local CMMC administration at the time of balloon injury accelerated reendothelialization at 4 weeks compared with saline (P<0.05). Moreover, CMMC delivery markedly improved endothelium-dependent vasoreactivity at 4 weeks compared with saline (P<0.005). Finally, CMMC treatment reduced neointimal formation by 55% at 4 weeks (P<0.05). CONCLUSIONS: These data demonstrate that delivery of CMMCs to balloon-injured arteries is associated with accelerated reendothelialization, enhanced endothelium-dependent vasoreactivity, and reduced neointimal formation. Thus, delivery of autologous CMMCs represents a novel vasculoprotective approach to attenuate the response to acute vascular injury.


Asunto(s)
Enfermedades de las Arterias Carótidas/terapia , Traumatismos de las Arterias Carótidas/terapia , Leucocitos Mononucleares/fisiología , Leucocitos Mononucleares/trasplante , Trasplante Autólogo/métodos , Acetilcolina/farmacología , Angioplastia de Balón/efectos adversos , Animales , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/etiología , Traumatismos de las Arterias Carótidas/patología , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/citología , Endotelio Vascular/lesiones , Supervivencia de Injerto , Técnicas In Vitro , Leucocitos Mononucleares/citología , Conejos , Túnica Íntima/lesiones , Túnica Íntima/patología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Vasodilatadores/farmacología
14.
Arterioscler Thromb Vasc Biol ; 23(9): 1521-7, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12816877

RESUMEN

BACKGROUND: Caveolin-1 is a regulator of signaling events originating from plasma membrane microdomains termed caveolae. This study was performed to determine the regulatory role of caveolin-1 on the proliferative events induced by platelet-derived growth factor (PDGF) in vascular smooth muscle cells (VSMCs). METHODS AND RESULTS: Treatment of VSMCs with PDGF for 24 hours resulted in a loss of caveolin-1 protein expression and plasma membrane-associated caveolae, despite a 3-fold increase in caveolin-1 mRNA. Pretreatment of VSMCs with chloroquine, an inhibitor of lysosomal function, inhibited the PDGF-induced loss of caveolin-1. These studies demonstrated that caveolin-1 was a target of PDGF signaling events. Adenoviral overexpression of caveolin-1 was associated with a switch in PDGF-induced signaling events from a proliferative response to an apoptotic response. This overexpression inhibited PDGF-induced expression of cyclin D1 in the presence of unaffected mitogen-activated protein kinase activation. CONCLUSIONS: Taken together, these studies suggest that caveolin-1 is an inhibitor of PDGF proliferative responses and might be capable of transforming PDGF-induced proliferative signals into death signals.


Asunto(s)
Apoptosis/fisiología , Caveolinas/fisiología , Músculo Liso Vascular/patología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Transducción de Señal/fisiología , Animales , Caveolina 1 , Caveolinas/biosíntesis , Caveolinas/metabolismo , División Celular/efectos de los fármacos , División Celular/fisiología , Células Cultivadas , Vasos Coronarios/citología , Regulación hacia Abajo/efectos de los fármacos , Arteria Femoral/patología , Arteria Femoral/cirugía , Humanos , Arteria Ilíaca/química , Arteria Ilíaca/patología , Inmunohistoquímica/métodos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Conejos
15.
Thromb Haemost ; 89(4): 747-51, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12669131

RESUMEN

Tissue factor (TF) is a small-molecular-weight glycoprotein that initiates the extrinsic coagulation pathway but may have important noncoagulation vascular functions as well. Tissue factor pathway inhibitor (TFPI) is a major physiological inhibitor of TF-initiated coagulation. Enhancement of vascular TFPI either by overexpression using gene transfer or delivery of protein to the vessel has been shown to reduce neointimal formation. However, the inherent role of TFPI in this process has not been defined. To do so, we utilized a murine model of vascular remodeling using flow cessation in mice, which are heterozygous for a genetic deletion of the first Kunitz domain of TFPI or wild type littermates. The heterozygotic mice had 50% of wild type TFPI activity in plasma as well as vascular homogenates. To study the effect of TFPI deficiency on neointimal formation, age matched TFPI(K1)+/- and wildtype littermates underwent unilateral common carotid artery ligation. Mice were sacrificed at 4 weeks and the ligated carotid arteries were analyzed. There was a significantly greater neointima to media ratio and less luminal area in the TFPI(K1)+/- mice compared to their TFPI(K1)+/+ littermates. The proliferative index of intimal cells in TFPI(K1)+/- mice at 1 week was significantly higher compared to TFPI(K1)+/+ mice. We conclude that TFPI deficiency enhances neointimal formation and proliferation associated with flow cessation. This suggests that TFPI may regulate vascular remodeling primarily through modulation of neointimal formation.


Asunto(s)
Lipoproteínas/deficiencia , Animales , Vasos Sanguíneos/patología , Arterias Carótidas/patología , División Celular , Coagulantes/metabolismo , Modelos Animales de Enfermedad , Eliminación de Gen , Técnicas de Transferencia de Gen , Heterocigoto , Homocigoto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estructura Terciaria de Proteína , Tromboplastina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...