Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 495(1): 904-910, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29154826

RESUMEN

W9 is a peptide that abrogates osteoclast differentiation via blockade of nuclear factor-κB ligand (RANKL)-RANK signaling, which activates bone formation. However, W9 stimulated osteogenesis in osteoblasts and mesenchymal stem cells. The present study demonstrated that the W9 peptide promoted osteogenic differentiation of human adipose-derived stem cells (hAdSCs) even under non-osteogenic differentiation culture conditions. W9-treated hAdSCs exhibited several osteocalcin-expressing cells and great mineralization compared to the BMP2-treated hAdSCs, which suggests that the W9 peptide had potent osteogenic potential in hAdSCs. W9 treatment also markedly enhanced the phosphorylation of p38, JNK, Erk1/2, and Akt, and BMP2 treatment only enhanced the phosphorylation of p38 and Erk1/2 in hAdSCs. hAdSCs did not express the RANKL gene, but W9 treatment upregulated Runx2, Collagen type 1A1 and TGF receptor genes and increased Akt phosphorylation. These results suggest that the W9-induced potent osteogenic induction was attributed to activation of TGF and the PI3 kinase/Akt signaling pathway in hAdSCs.


Asunto(s)
Adipocitos/citología , Diferenciación Celular/fisiología , Osteoblastos/citología , Osteoblastos/fisiología , Osteogénesis/fisiología , Péptidos Cíclicos/administración & dosificación , Células Madre/fisiología , Adipocitos/fisiología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos
2.
J Surg Res ; 196(1): 49-55, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25813142

RESUMEN

BACKGROUND: In patients with chronic severe aortic regurgitation (AR), aortic valve replacement (AVR) has been proved to promote left ventricular (LV) remodeling, especially LV end-diastolic dimension (LVEDD) reduction. However, there is little research whether postoperative LVEDD could return to normal parameter after AVR. The objective of this study was to determine predictors for the recovery of dilated LVEDD early after AVR. METHODS: The echocardiographic data of 105 patients, who underwent AVR for chronic pure AR between January 2005 and December 2011, were analyzed at the preoperative (3-7 d), early (6-8 mo), and late (2-y) postoperative stages, retrospectively. According to the baseline level, LVEDD >70 mm or LV end-systolic dimension (LVESD) >50 mm or LVESD index >25 mm/m(2) were defined as severe LV dilation. Patients were then categorized into two groups (group 1: severe LV dilation; group 2: nonsevere LV dilation). RESULTS: In all patients, four-fifth of the reduction in LV dimension occurred at early (6-8 mo) postoperative stage. The patients in both groups had significant decreases in the LVEDD and LVESD early after AVR, with an additional but insignificant reduction at late postoperative stage. The ejection fraction (EF) in both groups significantly increased at either early or late stage. However, the LVEDD and LVESD in group 1 were larger than those in group 2, and the EF in group 1 was lower than that in group 2 at early postoperative stage. By multivariate analysis, we found that the preoperative EF was a good predictor for the recovery of dilated LVEDD early after AVR (P = 0.009). Receiver-operating characteristics analysis showed that EF >52% was the best cut-off value for the recovery of LVEDD. CONCLUSIONS: In patients with chronic pure AR, preoperative EF may be a good predictor for successful recovery of dilated LVEDD early after AVR.


Asunto(s)
Insuficiencia de la Válvula Aórtica/cirugía , Válvula Aórtica/cirugía , Implantación de Prótesis de Válvulas Cardíacas , Volumen Sistólico , Función Ventricular Izquierda , Adulto , Anciano , Insuficiencia de la Válvula Aórtica/fisiopatología , Enfermedad Crónica , Diástole/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad
3.
J Clin Biochem Nutr ; 57(3): 244-53, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26566312

RESUMEN

The beneficial effect of dipeptidyl peptidase-4 inhibition on diet-induced extra-pancreatic effects, especially on liver tissue remains poorly understood. Thus, we made the experimental designs as follows; five-week-old male ob/ob mice, which develop type 2 diabetic mellitus and nonalcoholic fatty liver disease by taking a high-carbohydrate diet (HCD), were divided into a group in which a HCD was given for 8 weeks as control, and another in which a HCD added with 0.0018% sitagliptin was given for 8 weeks. Hepatic steatosis was seen in all mice, but the mean grade of steatosis in the sitagliptin-administrated mice was significantly decreased. The acetyl-CoA concentrations were lower in sitagliptin-administrated mice, although the differences were not significant. However, the malonyl-CoA concentrations were significantly lower in sitagliptin-administrated mice. The expression of acetyl-CoA carboxylase 1 was inhibited in sitagliptin-administrated mice, irrespective of expressions of carbohydrate responsive element-binding protein (ChREBP) or sterol regulatory element-binding protein (SREBP)-1c. In conclusion, sitagliptin may affect the development of nonalcoholic fatty liver disease by inhibiting the production of malonyl-CoA and thus synthesis of fatty acids in the liver.

4.
Circulation ; 127(1): 63-73, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23204107

RESUMEN

BACKGROUND: CXC-chemokine receptor 4 (CXCR4) regulates the retention of stem/progenitor cells in the bone marrow (BM), and the CXCR4 antagonist AMD3100 improves recovery from coronary ligation injury by mobilizing stem/progenitor cells from the BM to the peripheral blood. Thus, we investigated whether AMD3100 also improves recovery from ischemia/reperfusion injury, which more closely mimics myocardial infarction in patients, because blood flow is only temporarily obstructed. METHODS AND RESULTS: Mice were treated with single subcutaneous injections of AMD3100 (5 mg/kg) or saline after ischemia/reperfusion injury. Three days later, histological measurements of the ratio of infarct area to area at risk were smaller in AMD3100-treated mice than in mice administered saline, and echocardiographic measurements of left ventricular function were greater in the AMD3100-treated mice at week 4. CXCR4(+) cells were mobilized for just 1 day in both groups, but the mobilization of sca1(+)/flk1(+) cells endured for 7 days in AMD3100-treated mice compared with just 1 day in the saline-treated mice. AMD3100 upregulated BM levels of endothelial nitric oxide synthase (eNOS) and 2 targets of eNOS signaling, matrix metalloproteinase-9 and soluble Kit ligand. Furthermore, the loss of BM eNOS expression abolished the benefit of AMD3100 on sca1(+)/flk1(+) cell mobilization without altering the mobilization of CXCR4(+) cells, and the cardioprotective effects of AMD3100 were retained in eNOS-knockout mice that had been transplanted with BM from wild-type mice but not in wild-type mice with eNOS-knockout BM. CONCLUSIONS: AMD3100 prolongs BM progenitor mobilization and improves recovery from ischemia/reperfusion injury, and these benefits appear to occur through a previously unidentified link between AMD3100 and BM eNOS expression.


Asunto(s)
Compuestos Heterocíclicos/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Animales , Bencilaminas , Trasplante de Médula Ósea , Cardiotónicos/farmacología , Ciclamas , Modelos Animales de Enfermedad , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Recuperación de la Función/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
5.
Biochem Biophys Res Commun ; 446(4): 1102-7, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24680678

RESUMEN

Mesenchymal stem cells (MSCs) have generated a great deal of interest in the field of regenerative medicine. Adipose-derived stromal cells (AdSCs) are known to exhibit extensive proliferation potential and can undergo multilineage differentiation, sharing similar characteristics to bone marrow-derived MSCs. However, as the effect of AdSCs on tumor growth has not been studied sufficiently, we assessed the degree to which AdSCs affect the proliferation of prostate cancer (PCa) cell. Human AdSCs exerted an inhibitory effect on the proliferation of androgen-responsive (LNCaP) and androgen-nonresponsive (PC3) human PCa cells, while normal human dermal fibroblasts (NHDFs) did not, and in fact promoted PCa cell proliferation to a degree. Moreover, AdSCs induced apoptosis of LNCaP cells and PC3 cells, activating the caspase3/7 signaling pathway. cDNA microarray analysis suggested that AdSC-induced apoptosis in both LNCaP and PC3 cells was related to the TGF-ß signaling pathway. Consistent with our in vitro observations, local transplantation of AdSCs delayed the growth of tumors derived from both LNCaP- and PC3-xenografts in immunodeficient mice. This is the first preclinical study to have directly demonstrated that AdSC-induced PCa cell apoptosis may occur via the TGF-ß signaling pathway, irrespective of androgen-responsiveness. Since autologous AdSCs can be easily isolated from adipose tissue without any ethical concerns, we suggest that therapy with these cells could be a novel approach for patients with PCa.


Asunto(s)
Tejido Adiposo/citología , Apoptosis , Proliferación Celular , Próstata/patología , Neoplasias de la Próstata/terapia , Células del Estroma/trasplante , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Células del Estroma/citología , Células del Estroma/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba
6.
J Gastroenterol Hepatol ; 29(10): 1830-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24731186

RESUMEN

BACKGROUND AND AIM: Preclinical studies in rodent models of chronic liver fibrosis have shown that transplantation of peripheral blood (PB) CD34(+) cells leads to hepatic regeneration and a reduction of liver fibrosis by suppressing hepatic stellate cell activity and increasing matrix metalloproteinase activity. The aim of this study was to examine the safety and clinical efficacy of intrahepatic transplantation of autologous granulocyte colony-stimulating factor (G-CSF)-mobilized PB-CD34(+) cells in patients with decompensated liver cirrhosis. METHODS: PB-CD34(+) cells were isolated from G-CSF-mobilized apheresis products. Ten patients were treated with G-CSF-mobilized PB-CD34(+) cells (treatment group) and seven patients were treated with standard medical therapy. For mobilization, patients in the treatment group received subcutaneous injections of 10 µg G-CSF/kg/day for 5 days. The cells were then injected at three different doses (5 × 10(5) , 1 × 10(6) and 2 × 10(6) cells/kg) through the hepatic artery. Thereafter, all patients were followed up for 24 months. RESULTS: G-CSF treatment and leukapheresis were well tolerated, and no serious adverse events were observed. Patients in the treatment group had a significant but transient splenomegaly. After 24 weeks, serum albumin was significantly increased in patients who had received middle or high doses of CD34(+) cells compared with baseline. Doppler ultrasound showed a significant increase in hepatic blood flow velocity and blood flow volume after CD34(+) cell therapy. The hepatic vein pressure gradient decreased in two patients who received high-dose CD34(+) cells at week 16. CONCLUSIONS: CD34(+) cell therapy is feasible, safe and effective in slowing the decline of hepatic reserve function.


Asunto(s)
Antígenos CD34 , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Cirrosis Hepática/terapia , Trasplante de Células Madre de Sangre Periférica/métodos , Anciano , Autoinjertos , Estudios de Factibilidad , Femenino , Estudios de Seguimiento , Factor Estimulante de Colonias de Granulocitos/farmacología , Arteria Hepática , Células Estrelladas Hepáticas/parasitología , Venas Hepáticas/fisiopatología , Humanos , Inyecciones Subcutáneas , Circulación Hepática , Cirrosis Hepática/enzimología , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , Regeneración Hepática , Masculino , Metaloproteinasas de la Matriz/metabolismo , Persona de Mediana Edad , Estudios Prospectivos , Terapéutica , Factores de Tiempo , Presión Venosa
7.
Lab Invest ; 93(9): 1036-53, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23897412

RESUMEN

Lnk, an intracellular adapter protein, is expressed in hematopoietic cell lineages, which has recently been proved as an essential inhibitory signaling molecule for stem cell self-renewal in the stem cell factor-c-Kit signaling pathway with enhanced hematopoietic and osteogenic reconstitution in Lnk-deficient mice. Moreover, the therapeutic potential of hematopoietic stem/endothelial progenitor cells (EPCs) for fracture healing has been demonstrated with mechanistic insight into vasculogenesis/angiogenesis and osteogenesis enhancement in the fracture sites. We report here, Lnk siRNA-transfected endothelial commitment of c-kit+/Sca-1+/lineage- subpopulations of bone marrow cells have high EPC colony-forming capacity exhibiting endothelial markers, VE-Cad, VEGF and Ang-1. Lnk siRNA-transfected osteoblasts also show highly osteoblastic capacity. In vivo, locally transfected Lnk siRNA could successfully downregulate the expression of Lnk at the fracture site up to 1 week, and radiological and histological examination showed extremely accelerated fracture healing in Lnk siRNA-transfected mice. Moreover, Lnk siRNA-transfected mice exhibited sufficient therapeutic outcomes with intrinstic enhancement of angiogenesis and osteogenesis, specifically, the mice demonstrated better blood flow recovery in the sites of fracture. In our series of experiments, we clarified that a negatively regulated Lnk system contributed to a favorable circumstance for fracture healing by enhancing vasculogenesis/angiogenesis and osteogenesis. These findings suggest that downregulation of Lnk system may have the clinical potential for faster fracture healing, which contributes to the reduction of delayed unions or non-unions.


Asunto(s)
Fracturas Óseas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neovascularización Fisiológica/fisiología , ARN Interferente Pequeño/metabolismo , Cicatrización de Heridas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Células de la Médula Ósea/metabolismo , Proliferación Celular , Distribución de Chi-Cuadrado , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Histocitoquímica , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/genética , Flujometría por Láser-Doppler , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/genética , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/genética , Osteogénesis/fisiología , Fenotipo , ARN Interferente Pequeño/genética , Flujo Sanguíneo Regional , Estadísticas no Paramétricas , Transfección , Cicatrización de Heridas/genética , Microtomografía por Rayos X
8.
Angiogenesis ; 16(1): 45-58, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22941227

RESUMEN

Neural stem cells (NSCs) differentiate into endothelial cells (ECs) and neuronal cells. Estradiol (E2) is known to exhibit proangiogenic effects on ischemic tissues via EC activation. Therefore, we hypothesized that E2 can promote the therapeutic potential of NSC transplantation for injured nerve repair via the differentiation of NSCs into ECs during neovascularization. NSCs isolated from newborn mouse brains were transplanted into injured sciatic nerves with (NSC/E2 group) or without E2-conjugated gelatin hydrogel (E2 group). The NSC/E2 group exhibited the greatest recovery in motor nerve conduction velocity, voltage amplitude, and exercise tolerance. Histological analyses revealed increased intraneural vascularity and blood perfusion as well as striking NSC recruitment to the neovasculature in the injured nerves in the NSC/E2 group. In vitro, E2 enhanced the NSC migration and proliferation inhibiting apoptosis. Fluorescence-activated cell sorting analysis also revealed that E2 significantly increased the percentage of CD31 in NSCs, and the effect of E2 was completely neutralized by the estrogen receptor antagonist ICI. The combination of E2 administration and NSC transplantation cooperatively improved the functional recovery of injured peripheral nerves, at least in part, via E2-associated NSC differentiation into ECs. These findings provide a novel mechanistic insight into both NSC biology and the biological effects of endogenous E2.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Células Endoteliales/citología , Estradiol/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Células-Madre Neurales/citología , Traumatismos de los Nervios Periféricos/terapia , Animales , Proliferación Celular/efectos de los fármacos , Terapia Combinada , Células Endoteliales/efectos de los fármacos , Estradiol/sangre , Estradiol/uso terapéutico , Femenino , Ácido Láctico/administración & dosificación , Ácido Láctico/farmacología , Ratones , Ratones Endogámicos C57BL , Compresión Nerviosa , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/trasplante , Traumatismos de los Nervios Periféricos/sangre , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Traumatismos de los Nervios Periféricos/fisiopatología , Ácido Poliglicólico/administración & dosificación , Ácido Poliglicólico/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Recuperación de la Función/efectos de los fármacos , Nervio Ciático/efectos de los fármacos , Nervio Ciático/lesiones , Nervio Ciático/patología , Nervio Ciático/fisiopatología , Trasplante de Células Madre
9.
Int Wound J ; 10(5): 527-33, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22738265

RESUMEN

Impaired wound healing leading to skin ulceration is a serious complication of diabetes and may be caused by defective angiogenesis. Endothelial progenitor cells (EPCs) can augment neovascularisation in the ischaemic tissue. Experiments were performed to test the hypothesis that locally administered EPCs can promote wound healing in diabetes. Full-thickness skin wounds were created on the dorsum of diabetic mice. EPCs were obtained from bone marrow mononuclear cells (BMMNCs) and applied topically to the wound immediately after surgery. Vehicle and non-selective BMMNCs were used as controls. Wound size was measured on days 5, 10 and 14 after treatment, followed by resection, histological analysis and quantification of vascularity. Topical application of EPCs significantly promoted wound healing, as assessed by closure rate and wound vascularity. Immunostaining revealed that transplanted EPCs induced increased expression of vascular endothelial growth factor and basic fibroblast growth factor. Few EPCs were observed in the neovasculature based on in vivo staining of the functional vasculature. Ex vivo expanded EPCs promote wound healing in diabetic mice via mechanisms involving increased local cytokine expression and enhanced neovascularisation of the wound. This strategy exploiting the therapeutic capacity of autologously derived EPCs may be a novel approach to skin repair in diabetes.


Asunto(s)
Diabetes Mellitus Experimental , Pie Diabético/terapia , Endotelio Vascular/citología , Neovascularización Fisiológica/fisiología , Trasplante de Células Madre/métodos , Cicatrización de Heridas/fisiología , Animales , Células Cultivadas , Pie Diabético/patología , Modelos Animales de Enfermedad , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL
10.
J Cell Physiol ; 227(4): 1538-52, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21678408

RESUMEN

We investigated whether transplantation of purified human peripheral blood CD34(+) cells could reduce established liver fibrosis and up-regulate therapeutic regeneration. Human peripheral blood CD34(+) cells were isolated from total mononuclear cells of healthy volunteers by magnetic cell sorting. Recipient nude rats were injected intraperitoneally with carbon tetrachloride (CCl(4)) twice weekly for 3 weeks before single administration of CD34(+) cells. CCl(4) was then re-administered twice weekly for 3 more weeks, and the nude rats were sacrificed. Saline (control group), 1 × 10(5) (low-dose group), 5 × 10(5) (middle-dose group), or 2 × 10(6) (high-dose group) CD34(+) cells/kg body weight were intrasplenically transplanted after CCl(4) treatment for 3 weeks. Reverse transcriptase-polymerase chain reaction analysis of the freshly isolated CD34(+) cells revealed the expression of CD31, keratin19, α-smooth muscle actin (α-SMA), and epithelial growth factor, but not other liver related markers. The transplanted cells differentiated into vascular and sinusoidal endothelial cells, and vascular smooth muscle cells. CD34(+) cell transplantation reduced liver fibrosis in a dose-dependent fashion, with decreased collagen type-I and α-SMA-positive cells after 6 weeks of CCl(4) treatment by Mallory's Azan and immunohistochemical staining. Gelatin zymography showed that the expression levels of active matrix metalloproteinase-2 and -9 in CD34(+) cell transplanted livers were significantly stronger than those in saline-infused livers. In recipients of high-doses of CD34(+) cells, the number of PCNA-positive hepatocyte increased 6 weeks after CCl(4) treatment compared with saline-infused livers. We conclude that human peripheral blood CD34(+) cell transplantation halts established liver fibrosis and promotes hepatic regeneration in CCl(4)-induced chronic liver injury.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/terapia , Regeneración Hepática , Trasplante de Células Madre de Sangre Periférica , Animales , Antígenos CD34/metabolismo , Secuencia de Bases , Tetracloruro de Carbono/toxicidad , Diferenciación Celular , Supervivencia Celular , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/patología , Cartilla de ADN/genética , Células Endoteliales/patología , Expresión Génica , Células Estrelladas Hepáticas/patología , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/patología , Cirrosis Hepática/terapia , Regeneración Hepática/genética , Regeneración Hepática/fisiología , Masculino , Miocitos del Músculo Liso/patología , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Trasplante Heterólogo
11.
Lab Invest ; 92(4): 532-42, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22330336

RESUMEN

Both estradiol (E2) and Sonic Hedgehog (Shh) contribute to angiogenesis and nerve regeneration. Here, we investigated whether E2 improves the recovery of injured nerves by downregulating the Shh inhibitor hedgehog-interacting protein (HIP) and increasing Shh-induced angiogenesis. Mice were treated with local injections of E2 or placebo one week before nerve-crush injury; 28 days after injury, nerve conduction velocity, exercise duration, and vascularity were significantly greater in E2-treated mice than in placebo-treated mice. E2 treatment was also associated with higher mRNA levels of Shh, the Shh receptor Patched-1, and the Shh transcriptional target Gli1, but with lower levels of HIP. The E2-induced enhancement of nerve vascularity was abolished by the Shh inhibitor cyclopamine, and the effect of E2 treatment on Shh, Gli1, and HIP mRNA expression was abolished by the E2 inhibitor ICI. Gli-luciferase activity in human umbilical-vein endothelial cells (HUVECs) increased more after treatment with E2 and Shh than after treatment with E2 alone, and E2 treatment reduced HIP expression in HUVECs and Schwann cells without altering Shh expression. Collectively, these findings suggest that E2 improves nerve recovery, at least in part, by reducing HIP expression, which subsequently leads to an increase in Shh signaling and Shh-induced angiogenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Estradiol/metabolismo , Proteínas Hedgehog/metabolismo , Glicoproteínas de Membrana/metabolismo , Neovascularización Fisiológica , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/metabolismo , Animales , Regulación hacia Abajo , Estradiol/administración & dosificación , Femenino , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores Patched , Receptor Patched-1 , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Receptores de Superficie Celular/metabolismo , Recuperación de la Función , Transducción de Señal , Proteína con Dedos de Zinc GLI1
12.
J Exp Med ; 203(1): 153-63, 2006 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-16401693

RESUMEN

The cell surface receptor alpha4 integrin plays a critical role in the homing, engraftment, and maintenance of hematopoietic progenitor cells (HPCs) in the bone marrow (BM). Down-regulation or functional blockade of alpha4 integrin or its ligand vascular cell adhesion molecule-1 mobilizes long-term HPCs. We investigated the role of alpha4 integrin in the mobilization and homing of BM endothelial progenitor cells (EPCs). EPCs with endothelial colony-forming activity in the BM are exclusively alpha4 integrin-expressing cells. In vivo, a single dose of anti-alpha4 integrin antibody resulted in increased circulating EPC counts for 3 d. In hindlimb ischemia and myocardial infarction, systemically administered anti-alpha4 integrin antibody increased recruitment and incorporation of BM EPCs in newly formed vasculature and improved functional blood flow recovery and tissue preservation. Interestingly, BM EPCs that had been preblocked with anti-alpha4 integrin ex vivo or collected from alpha4 integrin-deficient mice incorporated as well as control cells into the neovasculature in ischemic sites, suggesting that alpha4 integrin may be dispensable or play a redundant role in EPC homing to ischemic tissue. These data indicate that functional disruption of alpha4 integrin may represent a potential angiogenic therapy for ischemic disease by increasing the available circulating supply of EPCs.


Asunto(s)
Integrina alfa4/metabolismo , Isquemia Miocárdica/fisiopatología , Neovascularización Fisiológica , Células Madre/fisiología , Animales , Médula Ósea , Movimiento Celular , Células Endoteliales , Integrina alfa4/genética , Masculino , Ratones , Ratones Noqueados
13.
J Neurosci Res ; 90(12): 2281-92, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22996658

RESUMEN

Spinal cord injury causes initial mechanical damage, followed by ischemia-induced, secondary degeneration, worsening the tissue damage. Although endothelial progenitor cells (EPCs) have been reported to play an important role for pathophysiological neovascularization in various ischemic tissues, the EPC kinetics following spinal cord injury have never been elucidated. In this study, we therefore assessed the in vivo kinetics of bone marrow-derived EPCs by EPC colony-forming assay and bone marrow transplantation from Tie2/lacZ transgenic mice into wild-type mice with spinal cord injury. The number of circulating mononuclear cells and EPC colonies formed by the mononuclear cells peaked at day 3 postspinal cord injury. Bone marrow transplantation study revealed that bone marrow-derived EPCs recruited into the injured spinal cord markedly increased at day 7, when neovascularization and astrogliosis drastically occurred in parallel with axon growth in the damaged tissue. To elucidate further the contribution of EPCs to recovery after spinal cord injury, exogenous EPCs were systemically infused immediately after the injury. The administered EPCs were incorporated into the injured spinal cord and accelerated neovascularization and astrogliosis. These findings suggest that bone marrow-derived EPCs may contribute to the tissue repair by augmenting neovascularization and astrogliosis following spinal cord injury.


Asunto(s)
Astrocitos/patología , Trasplante de Médula Ósea , Células Endoteliales/fisiología , Gliosis/etiología , Células Madre Hematopoyéticas/fisiología , Neovascularización Fisiológica , Traumatismos de la Médula Espinal/cirugía , Cicatrización de Heridas/fisiología , Animales , Linaje de la Célula , Movimiento Celular , Ensayo de Unidades Formadoras de Colonias , Genes Reporteros , Gliosis/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Quimera por Radiación , Recuperación de la Función , Traumatismos de la Médula Espinal/patología , Isquemia de la Médula Espinal/patología
14.
Circ J ; 76(1): 204-12, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22033345

RESUMEN

BACKGROUND: The survival of stem cells upon transplantation into ischemic myocardium is a major concern in cell-based therapy. In this study, we tested the hypothesis that activation of opioid receptors would enhance the survival of mesenchymal stem cells (MSCs) upon exposure to an injury stimulus. METHODS AND RESULTS: MSCs were obtained from rat bone marrow and cultured in basal DMEM cell culture medium. Delta-opioid receptor (DOR) was present in MSCs as examined by reverse transcription-polymerase chain reaction and immunochemistry. Activation of DOR with 5µmol/L SNC80 (DOR agonist) for 24h significantly enhanced MSC viability upon exposure to 5µg/ml actinomycin D as determined by TUNEL and MTT assays. The cytoprotection was abolished with 20µmol/L naltrindole hydrochloride (a DOR antagonist). Treatment of the cells with 1.5µmol/L chelerythrine (protein kinase C inhibitor) and 1.25µmol/L WP1066 (signal transducer and activator of transcription 3 (STAT3) inhibitor) blocked SNC80-induced cytoprotection. Furthermore, treatment of the cells with chelerythrine also blocked STAT3-phosphorylation and Mcl-1 gene expression. CONCLUSIONS: Taken together, the results indicate that DOR plays a critical role in MSC survival upon exposure to actinomycin D through activation of protein kinase C and its downstream signaling molecules STAT3 and Mcl-1. DOR may be a novel therapeutic target for stem cell survival during cell-based therapy.


Asunto(s)
Células Madre Mesenquimatosas/citología , Proteína Quinasa C/fisiología , Receptores Opioides delta/fisiología , Factor de Transcripción STAT3/fisiología , Transducción de Señal/fisiología , Animales , Benzamidas/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Dactinomicina/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/fisiología , Modelos Animales , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Naltrexona/análogos & derivados , Naltrexona/farmacología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Endogámicas F344 , Receptores Opioides delta/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
15.
Nat Med ; 11(11): 1197-204, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16244652

RESUMEN

Sonic hedgehog (Shh) is a crucial regulator of organ development during embryogenesis. We investigated whether intramyocardial gene transfer of naked DNA encoding human Shh (phShh) could promote a favorable effect on recovery from acute and chronic myocardial ischemia in adult animals, not only by promoting neovascularization, but by broader effects, consistent with the role of this morphogen in embryogenesis. After Shh gene transfer, the hedgehog pathway was upregulated in mammalian fibroblasts and cardiomyocytes. This resulted in preservation of left ventricular function in both acute and chronic myocardial ischemia by enhanced neovascularization, and reduced fibrosis and cardiac apoptosis. Shh gene transfer also enhanced the contribution of bone marrow-derived endothelial progenitor cells to myocardial neovascularization. These data suggest that Shh gene therapy may have considerable therapeutic potential in individuals with acute and chronic myocardial ischemia by triggering expression of multiple trophic factors and engendering tissue repair in the adult heart.


Asunto(s)
Terapia Genética , Corazón/embriología , Miocardio/metabolismo , Transducción de Señal , Transactivadores/uso terapéutico , Enfermedad Aguda , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Enfermedad Crónica , Modelos Animales de Enfermedad , Ecocardiografía , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog , Humanos , Ratones , Ratones Mutantes , Isquemia Miocárdica/etiología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/terapia , Miocardio/citología , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Porcinos , Función Ventricular Izquierda/fisiología
16.
Circulation ; 121(9): 1104-12, 2010 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-20176991

RESUMEN

BACKGROUND: Little is known about the role of endothelial progenitor cells (EPCs) in atherosclerosis. Accordingly, we performed a series of assessments with hypercholesterolemic (apolipoprotein E-null [ApoE(-/-)]) and wild-type (WT) mice to evaluate how cholesterol influences reendothelialization, atherosclerosis, and EPC function after arterial injury. METHODS AND RESULTS: Unexpectedly, reendothelialization (assessed by resistance to Evans blue staining) and circulating EPC counts (EPC culture assay) were greater in ApoE(-/-) mice than in WT mice, and transplantation of ApoE(-/-) bone marrow in WT mice accelerated endothelial recovery and increased recruitment of bone marrow-derived EPCs to the neoendothelium. Cholesterol concentration-dependently promoted the proliferation (MTS assay) of both ApoE(-/-) and WT EPCs, and the concentration dependence of EPC adhesion (to vitronectin-, collagen type I-, fibronectin-, and laminin-coated plates), migration (modified Boyden chamber assay), and antiapoptotic (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling stain) activity was biphasic. Cholesterol enhanced the messenger RNA expression (quantitative, real-time reverse-transcription polymerase chain reaction) of vascular endothelial growth factor and inhibited Notch1 messenger RNA expression in both ApoE(-/-) and WT EPCs, whereas endothelial nitric oxide synthase messenger RNA expression increased in ApoE(-/-) EPCs and declined in WT EPCs after cholesterol exposure. EPC activity was greater in Notch1(+/-) EPCs than in WT EPCs, and transplantation of Notch1(+/-) bone marrow accelerated endothelial recovery after arterial injury in WT mice. CONCLUSIONS: The results presented here provide novel insights into the role of EPCs during atherosclerosis and suggest that cholesterol and Notch1 may be involved in the regulation of EPC activity.


Asunto(s)
Traumatismos de las Arterias Carótidas/patología , Células Endoteliales/patología , Hipercolesterolemia/patología , Células Madre Mesenquimatosas/patología , Receptor Notch1/fisiología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/patología , Trasplante de Médula Ósea , Traumatismos de las Arterias Carótidas/complicaciones , Movimiento Celular , Colesterol/sangre , Regulación de la Expresión Génica , Genotipo , Hipercolesterolemia/complicaciones , Hipercolesterolemia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Óxido Nítrico Sintasa de Tipo III/genética , ARN Mensajero/biosíntesis , Quimera por Radiación , Receptor Notch1/biosíntesis , Receptor Notch1/deficiencia , Receptor Notch1/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
17.
Lab Invest ; 91(4): 539-52, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21135814

RESUMEN

Human multipotent adipose-derived stem cells (hMADSCs) have recently been isolated featuring extensive expansion capacity ex vivo. We tested the hypothesis that hMADSC transplantation might contribute to cardiac functional recovery by its direct or indirect effect on myocardial infarction (MI). Nude rats were either transplanted with hMADSCs or PBS (control) in ischemic myocardium immediately following MI. Echocardiographical assessment of cardiac function after MI with hMADSCs showed significant improvement of each parameter compared to that with PBS. Histological analysis also showed significantly reduced infarct size and increased capillary density in peri-infarct myocardium by hMADSC treatment. However, remarkable transdifferentiation of hMADSCs into cardiac or vascular lineage cells was not observed. Despite the less transdifferentiation capacity, hMADSCs produced robust multiple pro-angiogenic growth factors and chemokines, such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and stromal cell-derived factor-1α (SDF-1α). Specifically, hMADSC-derived SDF-1α had a crucial role for cooperative angiogenesis, with the paracrine effect of hMADSCs and Tie2-positive bone marrow (BM) progenitor recruitment in ischemic myocardium. hMADSCs exhibit a therapeutic effect on cardiac preservation following MI, with the production of VEGF, bFGF, and SDF-1α showing paracrine effects and endogenous BM stem/progenitor recruitment to ischemic myocardium rather than its direct contribution to tissue regeneration.


Asunto(s)
Adipocitos/trasplante , Isquemia Miocárdica/cirugía , Trasplante de Células Madre , Animales , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Capilares/patología , Diferenciación Celular , Quimiocina CXCL12 , Vasos Coronarios/patología , Ecocardiografía , Factores de Crecimiento de Fibroblastos , Corazón/fisiopatología , Humanos , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/etiología , Isquemia Miocárdica/fisiopatología , Miocardio/patología , Neovascularización Fisiológica , Comunicación Paracrina , Ratas , Ratas Desnudas , Células Madre/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Stem Cells ; 28(2): 365-75, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19859984

RESUMEN

Lnk is an intracellular adaptor protein reported as a negative regulator of proliferation in c-Kit positive, Sca-1 positive, lineage marker-negative (KSL) bone marrow cells. The KSL fraction in mouse bone marrow is believed to represent a population of hematopoietic and endothelial progenitor cells (EPCs). We report here that, in vitro, Lnk(-/-) KSL cells form more EPC colonies than Lnk(+/+) KSL cells and show higher expression levels of endothelial marker genes, including CD105, CD144, Tie-1, and Tie2, than their wild-type counterparts. In vivo, the administration of Lnk(+/+) KSL cells to a mouse spinal cord injury model promoted angiogenesis, astrogliosis, axon growth, and functional recovery following injury, with Lnk(-/-) KSL being significantly more effective in inducing and promoting these regenerative events. At day 3 following injury, large vessels could be observed in spinal cords treated with KSL cells, and reactive astrocytes were found to have migrated along these large vessels. We could further show that the enhancement of astrogliosis appears to be caused in conjunction with the acceleration of angiogenesis. These findings suggest that Lnk deletion reinforces the commitment of KSL cells to EPCs, promoting subsequent repair of injured spinal cord through the acceleration of angiogenesis and astrogliosis.


Asunto(s)
Astrocitos/fisiología , Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/fisiología , Neovascularización Fisiológica/fisiología , Proteínas/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Proteínas Adaptadoras Transductoras de Señales , Animales , Astrocitos/citología , Astrocitos/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Ratones Noqueados , Neovascularización Fisiológica/genética , Proteínas/genética , Traumatismos de la Médula Espinal/metabolismo
19.
Circ Res ; 105(9): 860-8, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19762683

RESUMEN

RATIONALE: Recent reports have demonstrated that signals from vascular endothelial cells are necessary for organogenesis that may precede vasculogenesis. However, the origin of these neovascular cells in regenerating tissue has not been clarified. OBJECTIVE: Here we tested the hypothesis that adult neural stem cells (NSCs) can differentiate into vascular lineage, as well as neural lineage, in the process of collaborative organogenesis. METHODS AND RESULTS: NSCs, clonally isolated from mouse brain, were shown to develop endothelial and smooth muscle phenotypes in vitro. To elucidate whether NSCs can simultaneously differentiate into vascular and neural cells in vivo, genetically labeled NSCs were administered to mice with unilateral sciatic nerve crush injury or operatively induced brain and myocardial ischemia. Two weeks later, necropsy examination disclosed recruitment of the labeled NSCs to sites of injury differentiating into vascular cells (endothelial cells and vascular smooth muscle cells) and Schwann cells in regenerating nerve. Similarly, NSC-derived vascular cells/astrocytes and endothelial cells were identified in ischemic brain tissue and capillaries in myocardium 2 weeks following transplantation, respectively. CONCLUSIONS: These findings, concurrent vasculogenesis and neurogenesis from a common stem cell, suggest that certain somatic stem cells are capable of differentiating into not only somatic cells of identity but also into vascular cells for tissue regeneration.


Asunto(s)
Células Madre Adultas/metabolismo , Isquemia Encefálica/fisiopatología , Células Endoteliales/metabolismo , Infarto del Miocardio/fisiopatología , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica , Neurogénesis , Neuronas/metabolismo , Neuropatía Ciática/fisiopatología , Células Madre Adultas/trasplante , Animales , Biomarcadores/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/cirugía , Comunicación Celular , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Modelos Animales de Enfermedad , Células Endoteliales/trasplante , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/cirugía , Miocitos del Músculo Liso/trasplante , Neuronas/trasplante , Neuropatía Ciática/metabolismo , Neuropatía Ciática/patología , Neuropatía Ciática/cirugía , Transducción de Señal , Esferoides Celulares , Trasplante de Células Madre , Factores de Tiempo
20.
Circ Res ; 104(8): 969-77, 2009 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-19325148

RESUMEN

Despite the fact that endothelial progenitor cells (EPCs) are important for postnatal neovascularization, their origins, differentiation, and modulators are not clear. Here, we demonstrate that Lnk, a negative regulator of hematopoietic stem cell proliferation, controls endothelial commitment of c-kit(+)/Sca-1(+)/Lineage(-) (KSL) subpopulations of bone marrow cells. The results of EPC colony-forming assays reveal that small (primitive) EPC colony formation by CD34(-) KSLs and large (definitive) EPC colony formation by CD34((dim)) KSLs are more robust in lnk(-/-) mice. In hindlimb ischemia, perfusion recovery is augmented in lnk(-/-) mice through enhanced proliferation and mobilization of EPCs via c-Kit/stem cell factor. We found that Lnk-deficient EPCs are more potent actors than resident cells in hindlimb perfusion recovery and ischemic neovascularization, mainly via the activity of bone marrow-EPCs. Similarly, lnk(-/-) mice show augmented retinal neovascularization and astrocyte network maturation without an increase in indicators of pathogenic angiogenesis in an in vivo model of retinopathy. Taken together, our results provide strong evidence that Lnk regulates bone marrow-EPC kinetics in vascular regeneration. Selective targeting of Lnk may be a safe and effective strategy to augment therapeutic neovascularization by EPC transplantation.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Endoteliales/trasplante , Isquemia/cirugía , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Proteínas/metabolismo , Regeneración , Células Madre/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos CD34/metabolismo , Astrocitos/metabolismo , Trasplante de Médula Ósea , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Miembro Posterior , Péptidos y Proteínas de Señalización Intracelular , Isquemia/metabolismo , Isquemia/fisiopatología , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Proteínas/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/fisiopatología , Transducción de Señal , Factor de Células Madre/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA