Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Vasc Surg ; 65(5): 1483-1492, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27514438

RESUMEN

OBJECTIVE: Adipose-derived stem cells (ASCs) are a potential adult mesenchymal stem cell source for restoring endothelial function in patients with critical limb ischemia. Fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor (VEGF) play a major role in angiogenesis and wound healing. This study evaluated the effects of FGF and VEGF on the proliferation, migration, and potential endothelial differentiation of human ASCs with regards to their use as endothelial cell substitutes. METHODS: ASCs were isolated from clinical lipoaspirates and cultured in M199 medium with fetal bovine serum (10%), FGF2 (10 ng/mL), VEGF (50 ng/mL), or combinations of FGF2 and VEGF. Cell proliferation rates, viability, and migration were measured by growth curves, MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide), and scratch assays. For cell attachment determinations, ASCs were seeded onto a scaffold of small intestinal submucosa for 5 days. Endothelial differentiation capabilities of ASCs were confirmed by expression of endothelial cell-specific markers using quantitative polymerase chain reaction, immunofluorescence staining, and cord formation on Matrigel (BD Biosciences, San Jose, Calif). PD173074, a selective inhibitor of FGF receptor, was used to confirm the importance of FGF signaling. RESULTS: ASCs treated with FGF or combinations of FGF and VEGF showed increased proliferation rates and consistent differentiation toward an endothelial cell lineage increase in platelet endothelial cell adhesion molecule (CD31), von Willebrand factor, endothelial nitric oxide synthase, and vascular endothelial cadherin message, and in protein and cord formation on Matrigel. FGF and VEGF stimulated ASC migration and increased the attachment and retention after seeding onto a matrix graft of small intestinal submucosa. Blockade of FGF signaling with PD173074 abrogated ASC endothelial cell differentiation potential. CONCLUSIONS: These results indicate that FGF and VEGF are ASC promoters for proliferation, migration, attachment, and endothelial differentiation. FGF and VEGF have a costimulatory effect on ASC endotheliogenesis. These results further suggest that ASCs with enhanced FGF signaling may potentially be used for tissue engineering and cell-based therapies in patients with critical limb ischemia.


Asunto(s)
Tejido Adiposo/citología , Inductores de la Angiogénesis/farmacología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Progenitoras Endoteliales/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Biomarcadores/metabolismo , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Progenitoras Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Humanos , Intestino Delgado/metabolismo , Células Madre Mesenquimatosas/metabolismo , Fenotipo , Factores de Tiempo , Andamios del Tejido
2.
J Vasc Surg ; 63(3): 805-14, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25499702

RESUMEN

OBJECTIVE: Previous work demonstrated the effectiveness of autologous adipose-derived stem cells (ASCs) as endothelial cell (EC) substitutes in vascular tissue engineering. We further this work toward clinical translation by evaluating ASC function after (1) replacement of fetal bovine serum (FBS) with autologous human plasma (HP) in culture and (2) cryopreservation. METHODS: Human ASCs and plasma, isolated from periumbilical fat and peripheral blood, respectively, were collected from the same donors. ASCs were differentiated in endothelial growth medium supplemented with FBS (2%) vs HP (2%). Proliferation was measured by growth curves and MTT assay. Endothelial differentiation was measured by quantitative polymerase chain reaction, assessment of acetylated low-density lipoprotein uptake, and cord formation after plating on Matrigel (BD Biosciences, San Jose, Calif). Similar studies were conducted before and after cryopreservation of ASCs and included assessment of cell retention on the luminal surface of a vascular graft. RESULTS: ASCs expanded in HP-supplemented medium showed (1) similar proliferation to FBS-cultured ASCs, (2) consistent differentiation toward an EC lineage (increases in CD31, von Willebrand factor, and CD144 message; acetylated low-density lipoprotein uptake; and cord formation on Matrigel), and (3) retention on the luminal surface after seeding and subsequent flow conditioning. Cryopreservation did not significantly alter ASC viability, proliferation, acquisition of endothelial characteristics, or retention after seeding onto a vascular graft. CONCLUSIONS: This study suggests that (1) replacement of FBS with autologous HP--a step necessary for the translation of this technology into human use--does not significantly impair proliferation or endothelial differentiation of ASCs used as EC substitutes and (2) ASCs are tolerant to cryopreservation in terms of maintaining EC characteristics and retention on a vascular graft.


Asunto(s)
Tejido Adiposo/citología , Bioprótesis , Prótesis Vascular , Técnicas de Cultivo de Célula , Criopreservación , Células Progenitoras Endoteliales/metabolismo , Plasma/metabolismo , Ingeniería de Tejidos/métodos , Biomarcadores/metabolismo , Antígeno CD146/genética , Antígeno CD146/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Humanos , Lipoproteínas LDL/metabolismo , Neovascularización Fisiológica , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factores de Tiempo , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
3.
J Surg Res ; 196(2): 404-15, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25891676

RESUMEN

BACKGROUND: Cancer patients with chemotherapy-induced immunosuppression have poor surgical site wound healing. Prior literature supports the use of human adipose-derived stem cell (hASC) lipoinjection to improve wound healing. It has been established that multipotent hASCs facilitate neovascularization, accelerate epithelialization, and quicken wound closure in animal models. Although hASC wound therapy may benefit surgical cancer patients, the chemotherapeutic effects on hASCs are unknown. We hypothesized that paclitaxel, a chemotherapeutic agent, impairs hASC growth, multipotency, and induces apoptosis. METHODS: hASCs were isolated and harvested from consented, chemotherapy and radiation naive patients. Growth curves, MTT (3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide), and EdU (5-ethynyl-2-deoxyguridine) assays measured cytotoxicity and proliferation. Oil Red O stain, Alizarin Red stain, matrigel tube formation assay, and quantitative polymerase chain reaction analyzed hASC differentiation. Annexin V assay measured apoptosis. Immunostaining and Western blot determined tumor necrosis factor α (TNF-α) expression. RESULTS: hASCs were selectively more sensitive to paclitaxel (0.01-30 µM) than fibroblasts (P < 0.05). After 12 d, paclitaxel caused hASC growth arrest, whereas control hASCs proliferated (P = 0.006). Paclitaxel caused an 80.6% reduction in new DNA synthesis (P < 0.001). Paclitaxel severely inhibited endothelial differentiation and capillary-like tube formation. Differentiation markers, lipoprotein lipase (adipogenic), alkaline phosphatase (osteogenic), CD31, and van Willebrand factor (endothelial), were significantly decreased (all P < 0.05) confirming paclitaxel impaired differentiation. Paclitaxel was also found to induce apoptosis and TNF-α was upregulated in paclitaxel-treated hASCs (P < 0.001). CONCLUSIONS: Paclitaxel is more cytotoxic to hASCs than fibroblasts. Paclitaxel inhibits hASC proliferation, differentiation, and induces apoptosis, possibly through the TNF-α pathway. Paclitaxel's severe inhibition of endothelial differentiation indicates neovascularization disruption, possibly causing poor wound healing in cancer patients receiving chemotherapy.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Antineoplásicos Fitogénicos/efectos adversos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Paclitaxel/efectos adversos , Tejido Adiposo/citología , Células Madre Adultas/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Células Endoteliales/citología , Fibroblastos/efectos de los fármacos , Humanos , Osteogénesis/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
4.
Wound Repair Regen ; 23(5): 728-36, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26043659

RESUMEN

In breast reconstructive procedures, adipose-derived stem cells (ASCs) that are present in clinical fat grafting isolates are considered to play the main role in improving wound healing. In patients following chemotherapy for breast cancer, poor soft tissue wound healing is a major problem. However, it is unclear if tamoxifen (TAM) as the most widely used hormonal therapeutic agent for breast cancer treatment, affects the ASCs and ultimately wound healing. This study evaluated whether TAM exposure to in vitro human ASCs modulate cellular functions. Human ASCs were isolated and treated with TAM at various concentrations. The effects of TAM on cell cycle, cell viability and proliferation rates of ASCs were examined by growth curves, MTT assay and BrdU incorporation, respectively. Annexin V and JC-1 Mitochondrial Membrane Potential assays were used to analyze ASC apoptosis rates. ASCs were cultured in derivative-specific differentiation media with or without TAM (5 uM) for 3 weeks. Adipogenic and osteogenic differentiation levels were measured by quantitative RT-PCR and histological staining. TAM has cytotoxic effects on human ASCs through apoptosis and inhibition of proliferation in dose- and time-dependent manners. TAM treatment significantly down-regulates the capacity of ASCs for adipogenic and osteogenic differentiation (p<0.05 vs. control), and inhibit the ability of the ASCs to subsequently formed cords in Matrigel. This study is the first findings to our knowledge that demonstrated that TAM inhibited ASC proliferation and multi-lineage ASC differentiation rates. These results may provide insight into the role of TAM with associated poor soft tissue wound healing and decreased fat graft survival in cancer patients receiving TAM.


Asunto(s)
Tejido Adiposo/trasplante , Células Madre/citología , Tamoxifeno/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos , Heridas y Lesiones/terapia , Tejido Adiposo/citología , Adulto , Anciano , Apoptosis , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Antagonistas de Estrógenos/uso terapéutico , Femenino , Humanos , Persona de Mediana Edad , Trasplante de Células Madre , Células Madre/efectos de los fármacos , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología
5.
J Biomed Sci ; 21: 55, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24898615

RESUMEN

BACKGROUND: Adipose tissue provides a readily available source of autologous stem cells. Adipose-derived stem cells (ASCs) have been proposed as a source for endothelial cell substitutes for lining the luminal surface of tissue engineered bypass grafts. Endothelial nitric oxide synthase (eNOS) is a key protein in endothelial cell function. Currently, endothelial differentiation from ASCs is limited by poor eNOS expression. The goal of this study was to investigate the role of three molecules, sphingosine-1-phosphate (S1P), bradykinin, and prostaglandin-E1 (PGE1) in ASC endothelial differentiation. Endothelial differentiation markers (CD31, vWF and eNOS) were used to evaluate the level of ASCs differentiation capability. RESULTS: ASCs demonstrated differentiation capability toward to adipose, osteocyte and endothelial like cell phenotypes. Bradykinin, S1P and PGE were used to promote differentiation of ASCs to an endothelial phenotype. Real-time PCR showed that all three molecules induced significantly greater expression of endothelial differentiation markers CD31, vWF and eNOS than untreated cells. Among the three molecules, S1P showed the highest up-regulation on endothelial differentiation markers. Immunostaining confirmed presence of more eNOS in cells treated with S1P than the other groups. Cell growth measurements by MTT assay, cell counting and EdU DNA incorporation suggest that S1P promotes cell growth during ASCs endothelial differentiation. The S1P1 receptor was expressed in ASC-differentiated endothelial cells and S1P induced up-regulation of PI3K. CONCLUSIONS: S1P up-regulates endothelial cell markers including eNOS in ASCs differentiated to endothelial like cells. This up-regulation appears to be mediated by the up-regulation of PI3K via S1P1 receptor. ASCs treated with S1P offer promising use as endothelial cell substitutes for tissue engineered vascular grafts and vascular networks.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Endoteliales/citología , Lisofosfolípidos/administración & dosificación , Óxido Nítrico Sintasa/metabolismo , Esfingosina/análogos & derivados , Tejido Adiposo , Alprostadil/administración & dosificación , Bradiquinina/administración & dosificación , Células Endoteliales/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Lisoesfingolípidos/biosíntesis , Esfingosina/administración & dosificación , Receptores de Esfingosina-1-Fosfato , Células Madre/citología , Células Madre/efectos de los fármacos
6.
J Surg Res ; 192(2): 656-63, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25091340

RESUMEN

BACKGROUND: Diabetic (DM) patients frequently lack autologous vascular tissue required for revascularization procedures and dialysis access creation. We have developed a tissue-engineered graft that uses adipose-derived stem cells (ASC) as endothelial cell substitutes. Here, we compare DM versus nondiabetic (NDM) ASC in terms of isolation efficiency, proliferation, commitment toward endothelial lineage, and seeding onto the luminal surface of a graft. METHODS: ASC were isolated from liposuction specimens of vascular surgery patients. Proliferation was assessed by constructing growth curves over 14 d. ASC were differentiated in endothelial growth medium (EGM2). Endothelial commitment was assessed by measuring endothelial cell-specific gene expression (CD31, von Willebrand factor) and by cord formation on Matrigel. Finally, ASC were seeded onto a vascular scaffold, flow conditioned, and imaged with confocal microscopy. RESULTS: Diabetes did not alter ASC isolation efficiency (224,028 ± 20,231 cells/g adipose for DM (n = 53) versus 259,345 ± 15,441 cells/g adipose for NDM (n = 145; P = 0.21). Growth curves for DM (n = 6) and NDM (n = 6) also appeared similar. After culture in EGM2, upregulation of CD31 and von Willebrand factor message was observed in NDM; these markers were found within the primary cultures of DM but no upregulation was observed after culture in EGM2. Both groups exhibited similar cord formation on Matrigel and retention to vascular scaffolds. CONCLUSIONS: Isolation and proliferation studies suggest that adipose is a promising source of stem cells for tissue engineering in the DM population. The angiogenic potential of DM ASC appears intact; however, differences in acquisition of endothelial cell markers suggest that differentiation may be inhibited or delayed by diabetes.


Asunto(s)
Tejido Adiposo/citología , Células Madre Adultas/citología , Prótesis Vascular , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/patología , Células Endoteliales/citología , Adulto , Biomarcadores , Cadáver , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Femenino , Humanos , Lipectomía , Masculino , Neovascularización Fisiológica
7.
Mol Genet Metab ; 104(3): 346-55, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21724437

RESUMEN

The Smith-Lemli-Opitz syndrome (SLOS) is an inherited disorder of cholesterol synthesis caused by mutations in DHCR7 which encodes the final enzyme in the cholesterol synthesis pathway. The immediate precursor to cholesterol synthesis, 7-dehydrocholesterol (7-DHC) accumulates in the plasma and cells of SLOS patients which has led to the idea that the accumulation of abnormal sterols and/or reduction in cholesterol underlies the phenotypic abnormalities of SLOS. We tested the hypothesis that 7-DHC accumulates in membrane caveolae where it disturbs caveolar bilayer structure-function. Membrane caveolae from skin fibroblasts obtained from SLOS patients were isolated and found to accumulate 7-DHC. In caveolar-like model membranes containing 7-DHC, subtle, but complex alterations in intermolecular packing, lipid order and membrane width were observed. In addition, the BK(Ca) K(+) channel, which co-migrates with caveolin-1 in a membrane fraction enriched with cholesterol, was impaired in SLOS cells as reflected by reduced single channel conductance and a 50 mV rightward shift in the channel activation voltage. In addition, a marked decrease in BK(Ca) protein but not mRNA expression levels was seen suggesting post-translational alterations. Accompanying these changes was a reduction in caveolin-1 protein and mRNA levels, but membrane caveolar structure was not altered. These results are consistent with the hypothesis that 7-DHC accumulation in the caveolar membrane results in defective caveolar signaling. However, additional cellular alterations beyond mere changes associated with abnormal sterols in the membrane likely contribute to the pathogenesis of SLOS.


Asunto(s)
Caveolas/metabolismo , Deshidrocolesteroles/metabolismo , Fibroblastos/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Síndrome de Smith-Lemli-Opitz/metabolismo , Caveolina 1/metabolismo , Células Cultivadas , Deshidrocolesteroles/química , Genotipo , Humanos , Immunoblotting , Membranas Artificiales , Microscopía Electrónica , Estructura Molecular , Piel/citología , Esteroles/metabolismo , Difracción de Rayos X
8.
J Surg Res ; 168(2): 306-14, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19959190

RESUMEN

BACKGROUND: Herein we evaluate the potential of adipose-derived stem cells (ASC) to differentiate into smooth muscle cells (SMC) and their potential for use in a tissue-engineered vascular graft. MATERIALS AND METHODS: We isolated ASC (CD13+29+90+) from the peri-umbilical adipose tissue of patients undergoing vascular surgery, and cultured them in media containing angiotensin II (AngII), sphingosylphosphorylcholine (SPC), or transforming growth factor-beta 1 (TGFß1) for up to 3 weeks. SMC differentiation was assessed by (1) expression of early (calponin, caldesmon) and late (myosin heavy chain, MHC) SMC markers by RT-PCR, qPCR and Western blot, and (2) contraction upon plating on collagen gel. Differentiated ASCs were seeded onto a vascular graft (decellularized saphenous vein) within a bioreactor, and cell attachment was determined using confocal microscopy. RESULTS: Prior to differentiation, ASC expressed low levels of all three molecular markers. After culture in each differentiating medium, the extent of up-regulation of calponin, caldesmon, and MHC was variable across all cell lines. After seeding onto collagen gel, ASCs differentiated in SPC and TGFß1 exhibit contractile properties, similar to smooth muscle cell controls. Differentiated stem cells adhered and proliferated on the vascular graft. CONCLUSION: These data suggest that human adipose-derived stem cells (1) exhibit variable expression of SMC molecular markers after differentiation, (2) exhibit a contractile phenotype after differentiation with SPC and TGFß1, and (3) proliferate on a vascular graft scaffold. Thus, ASCs are potentially useful in the construction of autologous arteries.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Angiotensina II/farmacología , Diferenciación Celular/efectos de los fármacos , Miocitos del Músculo Liso/citología , Fosforilcolina/análogos & derivados , Esfingosina/análogos & derivados , Factor de Crecimiento Transformador beta/farmacología , Anciano , Anciano de 80 o más Años , Prótesis Vascular , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Técnicas de Cultivo de Célula , Colágeno , Femenino , Humanos , Masculino , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Miocitos del Músculo Liso/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Fenotipo , Fosforilcolina/farmacología , Esfingosina/farmacología , Ingeniería de Tejidos , Andamios del Tejido , Calponinas
9.
J Surg Res ; 165(1): 112-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20080246

RESUMEN

Stem cells differentiate into a variety of cell lines, making them attractive for tissue engineering and regenerative medicine. Specific microenvironmental cues regulate self-renewal and differentiation capabilities. Oxygen is an important component of the cellular microenvironment, serving as both metabolic substrate and signaling molecule. Oxygen has been shown to have a variety of effects on embryonic and adult stem cells. This review examines the role of hypoxia in regulating stem cell biology, specifically focusing on growth, maintenance of pluripotency, differentiation, and production of growth factors. Particular attention is paid to hypoxia and stem cells in relation to therapeutic angiogenesis. We conclude that further study is needed to optimize the use of hypoxia as a stimulus for various stem cell functions, including its potential role in therapeutic angiogenesis.


Asunto(s)
Hipoxia de la Célula , Neovascularización Fisiológica , Células Madre/citología , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Células Madre/fisiología
10.
J Surg Res ; 163(2): e105-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20638677

RESUMEN

BACKGROUND: Most research evaluating adipose-derived stem cells (ASC) uses tissue obtained from young, healthy patients undergoing plastic surgical procedures. Given the propensity of other adult stem cell lines to diminish with increasing patient age and co-morbidities, we assess the availability of ASC in elderly patients undergoing vascular surgical procedures, and evaluate their acquisition of endothelial cell (EC) traits to define their potential use in vascular tissue engineering. METHODS AND METHODS: Adipose tissue obtained by liposuction from patients undergoing vascular procedures (n = 50) was digested with collagenase and centrifuged to remove mature adipocytes. The resultant number of cells, defined as the stromal-vascular (SV) pellet, was quantified. Following a 7-d culture period and negative selection for CD31 and CD45, the resultant number of ASC was quantified. After culture in differentiating media (EMG-2), ASCs were tested for the acquisition of endothelial-specific traits (expression of CD31, realignment in shear, cord formation on Matrigel). RESULTS: The SV pellet contained 2.87 ± 0.34 × 10(5) cells/g fat, and the resultant number of ASCs obtained was 1.41 ± 0.18 × 10(5) cells/g fat. Flow cytometry revealed a homogeneous ASC population (>98% positive for CD13, 29, 90). Advanced age or co-morbidity (obesity, diabetes, renal or peripheral vascular disease) did not significantly alter yield of ASC. After culture in differentiating media (EMG-2), ASCs acquired each of the endothelial-specific traits. CONCLUSION: ASC isolation appears independent of age and co-morbidities, and ASCs harvested from patients with vascular disease retain their ability to differentiate into endothelial-like cells. Adipose tissue, therefore, is a practical source of autologous, adult stem cells for vascular tissue engineering.


Asunto(s)
Tejido Adiposo/citología , Células Madre Adultas/citología , Procedimientos Quirúrgicos Vasculares , Adulto , Factores de Edad , Anciano , Diferenciación Celular , Separación Celular , Comorbilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ingeniería de Tejidos
11.
J Surg Res ; 152(1): 157-66, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19883577

RESUMEN

BACKGROUND: Adipose tissue is a readily available source of multipotent adult stem cells for use in tissue engineering/regenerative medicine. Various growth factors have been used to stimulate acquisition of endothelial characteristics by adipose-derived stem cells (ASC). Herein we study the effects of endothelial cell growth supplement (ECGS) and physiological shear force on the differentiation of ASC into endothelial cells. MATERIALS AND METHODS: Human ASC (CD13(+)29(+)90(+)31(-)45(-)) were isolated from periumbilical fat, cultured in ECGS media (for up to 3 wk), and exposed to physiological shear force (12 dynes for up to 8 d) in vitro. Endothelial phenotype was defined by cord formation on Matrigel, acetylated-low density lipoprotein (acLDL) uptake, and expression of nitric oxide synthase (eNOS), von Willebrand factor (vWF), and CD31 (platelet endothelial cell adhesion molecule, PECAM). Additionally, cell thrombogenicity was evaluated by seeding canine autologous ASC onto vascular grafts implanted within the canine arterial circulation for 2 wk. RESULTS: We found that undifferentiated ASC did not display any of the noted endothelial characteristics. After culture in ECGS, ASC formed cords in Matrigel but failed to take up acLDL or express the molecular markers. Subsequent exposure to shear resulted in stem cell realignment, acLDL uptake, and expression of CD31; eNOS and vWF expression was still not observed. Grafts seeded with cells grown in ECGS (+/- shear) remained patent (six of seven) at 2 wk but had a thin coat of fibrin along the luminal surfaces. CONCLUSIONS: This study suggests that (1) ECGS and shear promote the expression of several endothelial characteristics in human adipose-derived stem cells, but not eNOS or vWF; (2) their combined effects appear synergistic; and (3) stem cells differentiated in ECGS appear mildly thrombogenic in vitro, possibly related, in part, to insufficient eNOS expression. Thus, while the acquisition of several endothelial characteristics by adult stem cells derived from adipose tissue suggests these cells are a viable source of autologous cells for cardiovascular regeneration, further stimulation/modifications are necessary prior to using them as a true endothelial cell replacement.


Asunto(s)
Tejido Adiposo/citología , Células Madre Adultas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Endoteliales/citología , Células Madre Adultas/metabolismo , Células Madre Adultas/trasplante , Animales , Células Cultivadas , Medios de Cultivo , Perros , Células Endoteliales/metabolismo , Femenino , Humanos , Lipoproteínas LDL/metabolismo , Fenotipo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Resistencia al Corte , Trasplante de Células Madre/efectos adversos , Estrés Mecánico , Trombosis/etiología
14.
Arterioscler Thromb Vasc Biol ; 25(10): 2054-61, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16037567

RESUMEN

Observational studies of necrotic core progression identify intraplaque hemorrhage as a critical factor in atherosclerotic plaque growth and destabilization. The rapid accumulation of erythrocyte membranes causes an abrupt change in plaque substrate characterized by increased free cholesterol within the lipid core and excessive macrophage infiltration. Neoangiogenesis is associated closely with plaque progression, and microvascular incompetence is a likely source of intraplaque hemorrhage. Intimal neovascularization is predominantly thought to arise from the adventitia, where there are a plethora of pre-existing vasa vasorum. In lesions that have early necrotic cores, the majority of vessels invading from the adventitia occur at specific sites of medial wall disruption. A breech in the medial wall likely facilitates the rapid in-growth of microvessels from the adventitia, and exposure to an atherosclerotic environment stimulates abnormal vascular development characterized by disorganized branching and immature endothelial tubes with "leaky" imperfect linings. This network of immature blood vessels is a viable source of intraplaque hemorrhage providing erythrocyte-derived phospholipids and free cholesterol. The rapid change in plaque substrate caused by the excessive accumulation of erythrocytes may promote the transition from a stable to an unstable lesion. This review discusses the potential role of intraplaque vasa vasorum in lesion instability as it relates to plaque rupture.


Asunto(s)
Enfermedad de la Arteria Coronaria/patología , Hemorragia/patología , Neovascularización Patológica/patología , Animales , Enfermedad de la Arteria Coronaria/complicaciones , Hemorragia/etiología , Humanos , Necrosis , Neovascularización Patológica/complicaciones , Rotura
15.
Chem Phys Lipids ; 142(1-2): 33-42, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16584719

RESUMEN

The subendothelial retention of low density lipoproteins (LDL) is believed to be the central pathogenic event in atherosclerosis, as stated by the response-to-retention hypothesis. Sphingomyelinase, an enzyme present in the arteries, has been proven to promote LDL aggregation. This study investigates the hypothesis that the extent of LDL aggregation is determined by the molar ratio of sphingomyelinase (SMase)-to-LDL, rather than the absolute concentrations. A mass action model is used to describe the aggregation process, and binding and dissociation rate constants are determined by fitting of dynamic light scattering data. The model predicts aggregate sizes that agree well with experimental observations. This study also tests the hypothesis that monocyte uptake of LDL correlates with aggregate size. LDL aggregates of three specific sizes (75, 100, and 150 nm) were incubated with J774A.1 cells and the net accumulation of LDL was monitored by measuring changes in the cellular cholesterol and protein content. Relative to a control sample, cholesterol accumulation was enhanced for aggregate sizes of 75 and 150 nm. The intermediate size aggregates, 100 nm, led to a very striking result demonstrating that cholesterol accumulation was markedly greater than the other samples, and was sufficient to cause cell death. These results underscore an important role of colloidal aggregation, and the influence of LDL aggregate size, in atherosclerosis.


Asunto(s)
Lipoproteínas LDL/química , Esfingomielina Fosfodiesterasa/química , Animales , Aterosclerosis/patología , Sitios de Unión , Muerte Celular/efectos de los fármacos , Células Cultivadas , Colesterol/química , Colesterol/metabolismo , Cinética , Metabolismo de los Lípidos , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacología , Tamaño de la Partícula , Proteínas/química , Proteínas/metabolismo , Dispersión de Radiación , Esfingomielina Fosfodiesterasa/metabolismo
16.
Biochim Biophys Acta ; 1610(2): 198-207, 2003 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-12648774

RESUMEN

This review will discuss the use of small-angle X-ray diffraction approaches to study the organization of lipids in plasma membranes derived from two distinct mammalian cell types: arterial smooth muscle cells and ocular lens fiber cells. These studies indicate that cholesterol at an elevated concentration can self-associate and form immiscible domains in the plasma membrane, a phenomenon that contributes to both physiologic and pathologic cellular processes, depending on tissue source. In plasma membrane samples isolated from atherosclerotic smooth muscle cells, the formation of sterol-rich domains is associated with loss of normal cell function, including ion transport activity and control of cell replication. Analysis of meridional diffraction patterns from intact and reconstituted plasma membrane samples indicates the presence of an immiscible cholesterol domain with a unit cell periodicity of 34 A, consistent with a cholesterol monohydrate tail-to-tail bilayer, under disease conditions. These cholesterol domains were observed in smooth muscle cells enriched with cholesterol in vitro as well as from cells obtained ex vivo from an animal model of atherosclerosis. By contrast, well-defined cholesterol domains appear to be essential to the normal physiology of fiber cell plasma membranes of the human ocular lens. The organization of cholesterol into separate domains underlies the role of lens fiber cell plasma membranes in maintaining lens transparency. These domains may also interfere with cataractogenic aggregation of soluble lens proteins at the membrane surface. Taken together, these analyses provide examples of both physiologic and pathologic roles that sterol-rich domains may have in mammalian plasma membranes. These findings support a model of the membrane in which cholesterol aggregates into structurally distinct regions that regulate the function of the cell membrane.


Asunto(s)
Membrana Celular/patología , Colesterol/química , Arterias , Membrana Celular/química , Cristalización , Humanos , Cristalino/citología , Cristalino/patología , Microdominios de Membrana/química , Microdominios de Membrana/patología , Músculo Liso Vascular/citología , Músculo Liso Vascular/patología , Difracción de Rayos X/métodos
17.
J Tissue Eng Regen Med ; 9(11): 1277-85, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23319464

RESUMEN

This study evaluates the durability of a novel tissue engineered blood vessel (TEBV) created by seeding a natural vascular tissue scaffold (decellularized human saphenous vein allograft) with autologous adipose-derived stem cells (ASC) differentiated into endothelial-like cells. Previous work with this model revealed the graft to be thrombogenic, likely due to inadequate endothelial differentiation as evidenced by minimal production of nitric oxide (NO). To evaluate the importance of NO expression by the seeded cells, we created TEBV using autologous ASC transfected with the endothelial nitric oxide synthase (eNOS) gene to produce NO. We found that transfected ASC produced NO at levels similar to endothelial cell (EC) controls in vitro which was capable of causing vasorelaxation of aortic specimens ex vivo. TEBV (n = 5) created with NO-producing ASC and implanted as interposition grafts within the aorta of rabbits remained patent for two months and demonstrated a non-thrombogenic surface compared to unseeded controls (n = 5). Despite the xenograft nature of the scaffold, the TEBV structure remained well preserved in seeded grafts. In sum, this study demonstrates that upregulation of NO expression within adult stem cells differentiated towards an endothelial-like lineage imparts a non-thrombogenic phenotype and highlights the importance of NO production by cells to be used as endothelial cell substitutes in vascular tissue engineering applications.


Asunto(s)
Adipocitos/citología , Células Madre Adultas/citología , Óxido Nítrico Sintasa de Tipo III/química , Óxido Nítrico/química , Ingeniería de Tejidos/métodos , Tejido Adiposo/citología , Adulto , Animales , Aorta/patología , Aorta Abdominal/patología , Bioensayo , Diferenciación Celular , Células Endoteliales/citología , Vectores Genéticos , Xenoinjertos , Humanos , Microscopía Confocal , Fenotipo , Conejos , Ratas , Trombosis , Andamios del Tejido , Regulación hacia Arriba
18.
Prev Cardiol ; 7(4): 176-81, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15539964

RESUMEN

Combination therapy with a statin and niacin may provide optimal therapy for patients with combined hyperlipidemia and low levels of high-density lipoprotein (HDL) cholesterol. The authors assessed the efficacy and safety of rosuvastatin monotherapy, extended-release (ER) niacin monotherapy, or rosuvastatin and ER niacin combined therapy in patients with atherogenic dyslipidemia. In a 24-week, open-label, multicenter trial, men and women aged > or =18 years with fasting levels of total cholesterol > or =200 mg/dL, HDL cholesterol > or =45 mg/dL, triglycerides 200-800 mg/dL, and apolipoprotein B > or =110 mg/dL were randomly assigned to one of four treatment groups: rosuvastatin 10-40 mg, ER niacin 0.5-2 g, rosuvastatin 40 mg plus ER niacin 0.5-1 g, or rosuvastatin 10 mg plus ER niacin 0.5-2 g. Daily doses of rosuvastatin 40 mg monotherapy reduced low-density lipoprotein (LDL) cholesterol and non-HDL cholesterol levels significantly more than did either ER niacin 2 g monotherapy or rosuvastatin 10 mg combined with ER niacin 2 g. Addition of ER niacin 1 g to rosuvastatin 40 mg did not further reduce total or non-HDL cholesterol. Triglyceride reductions were similar among the four treatment groups. ER niacin mono- and combined therapy produced significantly greater rises in HDL cholesterol and apolipoprotein A-1 than did rosuvastatin monotherapy. Rosuvastatin monotherapy was better tolerated than ER niacin taken either alone or with rosuvastatin. In this study, rosuvastatin very effectively improved the three major lipoprotein-lipid abnormalities of combined hyperlipidemia.


Asunto(s)
Fluorobencenos/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hiperlipidemias/tratamiento farmacológico , Niacina/uso terapéutico , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Adulto , Apolipoproteínas/sangre , Colesterol/sangre , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Preparaciones de Acción Retardada , Quimioterapia Combinada , Femenino , Fluorobencenos/administración & dosificación , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Hiperlipidemias/sangre , Masculino , Persona de Mediana Edad , Niacina/administración & dosificación , Pirimidinas/administración & dosificación , Rosuvastatina Cálcica , Sulfonamidas/administración & dosificación , Resultado del Tratamiento , Triglicéridos/sangre , Estados Unidos
19.
J Colloid Interface Sci ; 279(1): 109-16, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15380418

RESUMEN

The response-to-retention hypothesis in atherosclerosis states that subendothelial retention of cholesterol-rich, atherogenic lipoproteins is the central pathogenic event that is both necessary and sufficient to provoke lesion initiation in an otherwise normal artery. Sphingomyelinase-induced aggregation of low density lipoproteins (LDL) is known to facilitate LDL retention, and the only available measurements of LDL aggregates suggest LDL aggregate size is approximately 100 nm. This study investigates the hypothesis that LDL aggregate size is determined by the relative rates of sphingomyelinase hydrolysis and LDL collisions. Using a combination of dynamic light scattering and UV-vis absorbance spectroscopy to measure aggregation kinetics and particle sizes, a mass action model was developed to describe the aggregation process. It is found that LDL aggregation is sensitive to the relative amounts of sphingomyelinase and LDL and to pH. Model rate parameters were fit to experimental data in vitro and used to predict LDL aggregate sizes in vivo. The value of 100 nm in vivo does not appear to be fixed; rather, it is the value expected for the prevailing enzyme-to-LDL molar ratio.


Asunto(s)
Lipoproteínas LDL/química , Esfingomielina Fosfodiesterasa/química , Cinética , Propiedades de Superficie , Factores de Tiempo
20.
Mol Genet Metab Rep ; 1: 431-442, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25405082

RESUMEN

Smith-Lemli-Opitz syndrome (SLOS) is a congenital, autosomal recessive metabolic and developmental disorder caused by mutations in the enzyme which catalyzes the reduction of 7-dehydrocholesterol (7DHC) to cholesterol. Herein we show that dermal fibroblasts obtained from SLOS children display increased basal levels of LC3B-II, the hallmark protein signifying increased autophagy. The elevated LC3B-II is accompanied by increased beclin-1 and cellular autophagosome content. We also show that the LC3B-II concentration in SLOS cells is directly proportional to the cellular concentration of 7DHC, suggesting that the increased autophagy is caused by 7DHC accumulation secondary to defective DHCR7. Further, the increased basal LC3B-II levels were decreased significantly by pretreating the cells with antioxidants implicating a role for oxidative stress in elevating autophagy in SLOS cells. Considering the possible source of oxidative stress, we examined mitochondrial function in the SLOS cells using JC-1 assay and found significant mitochondrial dysfunction compared to mitochondria in control cells. In addition, the levels of PINK1 which targets dysfunctional mitochondria for removal by the autophagic pathway are elevated in SLOS cells, consistent with mitochondrial dysfunction as a stimulant of mitophagy in SLOS. This suggests the increase in autophagic activity may be protective, i.e., to remove dysfunctional mitochondria. Taken together, these studies are consistent with a role for mitochondrial dysfunction leading to increased autophagy in SLOS pathophysiology.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA