Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biology (Basel) ; 10(5)2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33922704

RESUMEN

Molecular hydrogen (MH) reportedly exerts therapeutic effects against inflammatory diseases as a suppressor of free radical chain reactions. Here, the cardiovascular protective effects of the intake of molecular hydrogen water (MHW) were investigated using high-fat diet-induced obesity (DIO) mice. MHW was prepared using supplier sticks and degassed water as control. MHW intake for 2 weeks did not improve blood sugar or body weight but decreased heart weight in DIO mice. Moreover, MHW intake improved cardiac hypertrophy, shortened the width of cardiomyocytes, dilated the capillaries and arterioles, activated myocardial eNOS-Ser-1177 phosphorylation, and restored left ventricular function in DIO mice. MHW intake promoted the histological conversion of hypertrophy to hyperplasia in white and brown adipose tissues (WAT and BAT) with the upregulation of thermogenic and cardiovascular protective genes in BAT (i.e., Ucp-1, Vegf-a, and eNos). Furthermore, the results of a colony formation assay of bone-marrow-derived endothelial progenitor cells (EPCs) indicated that MHW activated the expansion, differentiation, and mobilization of EPCs to maintain vascular homeostasis. These findings indicate that the intake of MHW exerts cardiovascular protective effects in DIO mice. Hence, drinking MHW is a potential prophylactic strategy against cardiovascular disorders in metabolic syndrome.

2.
Biocontrol Sci ; 18(2): 83-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23796639

RESUMEN

The oxidative power of hydroxyl radicals has been applied to disinfection systems for the purpose of oral hygiene by utilizing blue light-induced photolysis of hydrogen peroxide (H2O2) in our laboratory. In the present study, the bactericidal potential of blue light-irradiated oxydol products via hydroxyl radical generation was compared with that of 3% (w/v) H2O2. Eleven commercially available oxydol products were used in the present study. Even though a few of the products that contained ethanol, a hydroxyl radical scavenger, as an additive showed slightly lower hydroxyl radical yield as compared with 3% (w/v) H2O2, the blue-light irradiation of each oxydol product for 3 min showed similar or superior bactericidal activity against Staphylococcus aureus to that of 3% (w/v) H2O2. The results strongly suggest that any of the oxydol products tested in the present study can be used as a source of hydroxyl radicals for the disinfection technique developed in our laboratory.


Asunto(s)
Antibacterianos/farmacología , Peróxido de Hidrógeno/farmacología , Radical Hidroxilo/farmacología , Antibacterianos/química , Peróxido de Hidrógeno/química , Radical Hidroxilo/química , Rayos Láser , Luz , Staphylococcus aureus/efectos de los fármacos
3.
PLoS One ; 6(9): e24872, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21969865

RESUMEN

RATIONALE: Despite preclinical success in regenerating and revascularizing the infarcted heart using angiogenic growth factors or bone marrow (BM) cells, recent clinical trials have revealed less benefit from these therapies than expected. OBJECTIVE: We explored the therapeutic potential of myocardial gene therapy of placental growth factor (PlGF), a VEGF-related angiogenic growth factor, with progenitor-mobilizing activity. METHODS AND RESULTS: Myocardial PlGF gene therapy improves cardiac performance after myocardial infarction, by inducing cardiac repair and reparative myoangiogenesis, via upregulation of paracrine anti-apoptotic and angiogenic factors. In addition, PlGF therapy stimulated Sca-1(+)/Lin(-) (SL) BM progenitor proliferation, enhanced their mobilization into peripheral blood, and promoted their recruitment into the peri-infarct borders. Moreover, PlGF enhanced endothelial progenitor colony formation of BM-derived SL cells, and induced a phenotypic switch of BM-SL cells, recruited in the infarct, to the endothelial, smooth muscle and cardiomyocyte lineage. CONCLUSIONS: Such pleiotropic effects of PlGF on cardiac repair and regeneration offer novel opportunities in the treatment of ischemic heart disease.


Asunto(s)
Células de la Médula Ósea/citología , Isquemia Miocárdica/metabolismo , Neovascularización Patológica , Proteínas Gestacionales/metabolismo , Células Madre/citología , Animales , Proliferación Celular , Electrocardiografía/métodos , Femenino , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/química , Hemodinámica , Humanos , Masculino , Ratones , Ratones Transgénicos , Isquemia Miocárdica/terapia , Miocardio/patología , Fenotipo , Factor de Crecimiento Placentario , Ratas , Ratas Sprague-Dawley , Regeneración
4.
J Exp Med ; 207(10): 2207-23, 2010 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-20855498

RESUMEN

The therapeutic potential of hematopoietic stem cells/endothelial progenitor cells (HSCs/EPCs) for fracture healing has been demonstrated with evidence for enhanced vasculogenesis/angiogenesis and osteogenesis at the site of fracture. The adaptor protein Lnk has recently been identified as an essential inhibitor of stem cell factor (SCF)-cKit signaling during stem cell self-renewal, and Lnk-deficient mice demonstrate enhanced hematopoietic reconstitution. In this study, we investigated whether the loss of Lnk signaling enhances the regenerative response during fracture healing. Radiological and histological examination showed accelerated fracture healing and remodeling in Lnk-deficient mice compared with wild-type mice. Molecular, physiological, and morphological approaches showed that vasculogenesis/angiogenesis and osteogenesis were promoted in Lnk-deficient mice by the mobilization and recruitment of HSCs/EPCs via activation of the SCF-cKit signaling pathway in the perifracture zone, which established a favorable environment for bone healing and remodeling. In addition, osteoblasts (OBs) from Lnk-deficient mice had a greater potential for terminal differentiation in response to SCF-cKit signaling in vitro. These findings suggest that inhibition of Lnk may have therapeutic potential by promoting an environment conducive to vasculogenesis/angiogenesis and osteogenesis and by facilitating OB terminal differentiation, leading to enhanced fracture healing.


Asunto(s)
Curación de Fractura , Osteogénesis , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Diferenciación Celular , Fracturas Óseas/metabolismo , Fracturas Óseas/fisiopatología , Fracturas Óseas/terapia , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Neovascularización Patológica , Neovascularización Fisiológica , Osteoblastos/metabolismo , Osteoblastos/patología , Osteogénesis/genética , Proteínas/genética , Transducción de Señal , Células Madre/metabolismo , Células Madre/patología
5.
Arterioscler Thromb Vasc Biol ; 29(11): 1830-5, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19679830

RESUMEN

OBJECTIVE: Unrestricted somatic stem cells (USSCs) were successfully identified from human cord blood. However, the efficacy of USSC transplantation for improving left ventricular (LV) function post myocardial infarction (MI) is still controversial. METHODS AND RESULTS: PBS, 1x10(6) human fibroblasts (Fbr), 1x10(5) USSCs (LD), or 1x10(6) USSCs (HD) were transplanted intramyocardially 20 minutes after ligating the LAD of nude rats. Echocardiography and a microtip conductance catheter at day 28 revealed a dose-dependent improvement of LV function after USSC transplantation. Necropsy examination revealed dose-dependent augmentation of capillary density and inhibition of LV fibrosis. Dual-label immunohistochemistry for cardiac troponin-I and human nuclear antigen (HNA) demonstrated that human cardiomyocytes (CMCs) were dose-dependently generated in ischemic myocardium 28 days after USSC transplantation. Similarly, dual-label immunostaining for smooth muscle actin and class I human leukocyte antigen or that for von Willebrand factor and HNA also revealed a dose-dependent vasculogenesis after USSC transplantation. RT-PCR indicated that expression of human-specific genes of CMCs, smooth muscle cells, and endothelial cell markers in infarcted myocardium were significantly augmented in USSC-treated animals compared with control groups. CONCLUSIONS: USSC transplantation leads to functional improvement and recovery from MI and exhibits a significant and dose-dependent potential for concurrent cardiomyogenesis and vasculogenesis.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Circulación Coronaria/fisiología , Infarto del Miocardio/terapia , Células Madre Pluripotentes/trasplante , Remodelación Ventricular/fisiología , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Ecocardiografía , Femenino , Sangre Fetal/citología , Humanos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Ratas Desnudas , Función Ventricular/fisiología
6.
PLoS One ; 3(3): e1789, 2008 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-18335059

RESUMEN

BACKGROUND: Cellular cardiomyoplasty for myocardial infarction has been developed using various cell types. However, complete differentiation and/or trans-differentiation into cardiomyocytes have never occurred in these transplant studies, whereas functional contributions were reported. METHODS AND RESULTS: Skeletal muscle interstitium-derived CD34(+)/CD45(-) (Sk-34) cells were purified from green fluorescent protein transgenic mice by flowcytometory. Cardiac differentiation of Sk-34 cells was examined by in vitro clonal culture and co-culture with embryonic cardiomyocytes, and in vivo transplantation into a nude rat myocardial infarction (MI) model (left ventricle). Lower relative expression of cardiomyogenic transcription factors, such as GATA-4, Nkx2-5, Isl-1, Mef2 and Hand2, was seen in clonal cell culture. However, vigorous expression of these factors was seen on co-culture with embryonic cardiomyocytes, together with formation of gap-junctions and synchronous contraction following sphere-like colony formation. At 4 weeks after transplantation of freshly isolated Sk-34 cells, donor cells exhibited typical cardiomyocyte structure with formation of gap-junctions, as well as intercalated discs and desmosomes, between donor and recipient and/or donor and donor cells. Fluorescence in situ hybridization (FISH) analysis detecting the rat and mouse genomic DNA and immunoelectron microscopy using anti-GFP revealed donor-derived cells. Transplanted Sk-34 cells were incorporated into infarcted portions of recipient muscles and contributed to cardiac reconstitution. Significant improvement in left ventricular function, as evaluated by transthoracic echocardiography and micro-tip conductance catheter, was also observed. CONCLUSIONS AND SIGNIFICANCE: Skeletal muscle-derived multipotent Sk-34 cells that can give rise to skeletal and smooth muscle cells as reported previously, also give rise to cardiac muscle cells as multi-myogenic stem cells, and thus are a potential source for practical cellular cardiomyoplasty.


Asunto(s)
Trasplante de Células , Músculo Esquelético/citología , Miocardio/citología , Células Madre/citología , Animales , Diferenciación Celular , Técnicas de Cocultivo , Proteínas Fluorescentes Verdes/genética , Inmunofenotipificación , Hibridación Fluorescente in Situ , Ratones , Ratones Transgénicos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Cell Physiol ; 215(1): 234-42, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18205179

RESUMEN

We recently reported that systemic administration of peripheral blood (PB) CD34+ cells, an endothelial progenitor cell (EPC)-enriched population, contributed to fracture healing via vasculogenesis/angiogenesis. However, pathophysiological role of EPCs in fracture healing process has not been fully clarified. Therefore, we investigated the hypothesis whether mobilization and incorporation of bone marrow (BM)-derived EPCs may play a pivotal role in appropriate fracture healing. Serial examinations of Laser doppler perfusion imaging and histological capillary density revealed that neovascularization activity at the fracture site peaked at day 7 post-fracture, the early phase of endochondral ossifification. Fluorescence-activated cell sorting (FACS) analysis demonstrated that the frequency of BM cKit+Sca1+Lineage- (Lin-) cells and PB Sca1+Lin- cells, which are EPC-enriched fractions, significantly increased post-fracture. The Sca1+ EPC-derived vasuculogenesis at the fracture site was confirmed by double immunohistochemistry for CD31 and Sca1. BM transplantation from transgenic donors expressing LacZ transcriptionally regulated by endothelial cell-specific Tie-2 promoter into wild type also provided direct evidence that EPCs contributing to enhanced neovascularization at the fracture site were specifically derived from BM. Animal model of systemic administration of PB Sca1+Lin- Green Fluorescent Protein (GFP)+ cells further confirmed incorporation of the mobilized EPCs into the fracture site for fracture healing. These findings indicate that fracture may induce mobilization of EPCs from BM to PB and recruitment of the mobilized EPCs into fracture sites, thereby augment neovascularization during the process of bone healing. EPCs may play an essential role in fracture healing by promoting a favorable environment through neovascularization in damaged skeletal tissue.


Asunto(s)
Células de la Médula Ósea/patología , Huesos/patología , Movimiento Celular , Células Endoteliales/patología , Fracturas Óseas/patología , Células Madre/patología , Cicatrización de Heridas , Animales , Ataxina-1 , Ataxinas , Huesos/irrigación sanguínea , Fracturas Óseas/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Fenotipo
8.
Arterioscler Thromb Vasc Biol ; 27(6): 1326-33, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17363693

RESUMEN

BACKGROUND: Therapeutic effect of stem cell transplantation (SCTx) for myocardial neovascularization has been evaluated by histological capillary density in small animals. However, it has been technically difficult to obtain imaging evidence of collateral formation by conventional angiography. METHODS AND RESULTS: Peripheral blood CD34+ and CD34- cells were isolated from patients with critical limb ischemia. PBS, CD34- cells, or CD34+ cells were intramyocardially transplanted after ligating LAD of nude rats. Coronary angiography of ex vivo beating hearts 5 and 28 days after the treatment was performed using the third generation synchrotron radiation microangiography (SRM), which has potential to visualize vessels as small as 20 microm in diameter. The SRM was performed pre and post sodium nitroprusside (SNP) to examine vascular physiology at each time point. Diameter of most collateral vessels was 20 to 120 microm, apparently invisible size in conventional angiography. Rentrop scores at day 28 pre and post SNP were significantly greater in CD34+ cell group than other groups (P<0.01). To quantify the extent of collateral formation, angiographic microvessel density (AMVD) in the occluded LAD area was analyzed. AMVD on day 28 post SNP, not pre SNP, was significantly augmented in CD34+ cell group than other groups (P<0.05). AMVD post SNP closely correlated with histological capillary density (R=0.82, P<0.0001). CONCLUSIONS: The SRM, capable of visualizing microvessels, may be useful for morphometric and physiological evaluation of coronary collateral formation by SCTx. The novel imaging system may be an essential tool in future preclinical/translational research of stem cell biology.


Asunto(s)
Angiografía Coronaria/métodos , Vasos Coronarios/patología , Infarto del Miocardio/diagnóstico por imagen , Revascularización Miocárdica/métodos , Neovascularización Fisiológica , Trasplante de Células Madre de Sangre Periférica , Sincrotrones , Anciano , Animales , Procedimientos Quirúrgicos Cardíacos , Circulación Colateral , Circulación Coronaria , Vasos Coronarios/fisiopatología , Enfermedad Crítica , Modelos Animales de Enfermedad , Células Endoteliales/patología , Extremidades/irrigación sanguínea , Femenino , Humanos , Isquemia/patología , Masculino , Microcirculación/diagnóstico por imagen , Microcirculación/fisiopatología , Persona de Mediana Edad , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/cirugía , Ratas , Ratas Desnudas , Células Madre/patología , Factores de Tiempo , Trasplante Heterólogo
9.
Circulation ; 114(20): 2163-9, 2006 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-17075009

RESUMEN

BACKGROUND: We compared the therapeutic potential of purified mobilized human CD34+ cells with that of mobilized total mononuclear cells (tMNCs) for the preservation/recovery of myocardial tissue integrity and function after myocardial infarction (MI). METHODS AND RESULTS: CD34+ cells were purified from peripheral blood tMNCs of healthy volunteers by magnetic cell sorting after a 5-day administration of granulocyte colony-stimulating factor. Phosphate-buffered saline (PBS), 5x10(5) CD34+ cells/kg, 5x10(5) tMNCs/kg (low-dose MNCs [loMNCs]), or a higher dose of tMNCs (hiMNCs) containing 5x10(5) CD34+ cells/kg was transplanted intramyocardially 10 minutes after the induction of MI in athymic nude rats. Hematoxylin and eosin staining revealed that moderate to severe hemorrhagic MI on day 3 was more frequent in the hiMNC group than in the PBS and CD34+ cell groups. Immunostaining for human-specific CD45 revealed abundant distribution of hematopoietic/inflammatory cells derived from transplanted cells in the ischemic myocardium of the hiMNC group. Capillary density on day 28 was significantly greater in the CD34+ cell group (721.1+/-19.9 per 1 mm2) than in the PBS, loMNC, and hiMNC groups (384.7+/-11.0, 372.5+/-14.1, and 497.5+/-24.0 per 1 mm2) (P<0.01). Percent fibrosis area on day 28 was less in the CD34(+) cell group (15.6+/-0.9%) than in the PBS, loMNC, and hiMNC groups (26.3+/-1.2%, 27.5+/-1.8%, and 22.2+/-1.8%) (P<0.05). Echocardiographic fractional shortening on day 28 was significantly higher in the CD34+ cell group (30.3+/-0.9%) than in the PBS, loMNC, and hiMNC groups (22.7+/-1.5%, 23.4+/-1.1%, and 24.9+/-1.7%; P<0.05). Echocardiographic regional wall motion score was better preserved in the CD34+ cell group (21.8+/-0.5) than in the PBS, loMNC, and hiMNC groups (25.4+/-0.4, 24.9+/-0.4, and 24.1+/-0.6; P<0.05). CONCLUSIONS: CD34+ cells exhibit superior efficacy for preserving myocardial integrity and function after MI than unselected circulating MNCs.


Asunto(s)
Antígenos CD34/metabolismo , Monocitos/metabolismo , Monocitos/trasplante , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/cirugía , Neovascularización Fisiológica , Animales , Diferenciación Celular , Ecocardiografía , Femenino , Hemorragia/etiología , Humanos , Monocitos/patología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Miocardio/patología , Periodo Posoperatorio , Ratas , Ratas Desnudas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Factores de Tiempo , Trasplante Heterólogo , Función Ventricular Izquierda , Remodelación Ventricular
10.
Am J Pathol ; 169(4): 1440-57, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17003498

RESUMEN

Failures in fracture healing are mainly caused by a lack of vascularization. Adult human circulating CD34+ cells, an endothelial/hematopoietic progenitor-enriched cell population, have been reported to differentiate into osteoblasts in vitro; however, the therapeutic potential of CD34+ cells for fracture healing is still unclear. Therefore, we performed a series of experiments to test our hypothesis that functional fracture healing is supported by vasculogenesis and osteogenesis via regenerative plasticity of CD34+ cells. Peripheral blood CD34+ cells, isolated from total mononuclear cells of adult human volunteers, showed gene expression of osteocalcin in 4 of 20 freshly isolated cells by single cell reverse transcriptase-polymerase chain reaction analysis. Phosphate-buffered saline, mononuclear cells, or CD34+ cells were intravenously transplanted after producing nonhealing femoral fractures in nude rats. Reverse transcriptase-polymerase chain reaction and immunohistochemical staining at the peri-fracture site demonstrated molecular and histological expression of human-specific markers for endothelial cells and osteoblasts at week 2. Functional bone healing assessed by biomechanical as well as radiological and histological examinations was significantly enhanced by CD34+ cell transplantation compared with the other groups. Our data suggest circulating human CD34+ cells have therapeutic potential to promote an environment conducive to neovascularization and osteogenesis in damaged skeletal tissue, allowing the complete healing of fractures.


Asunto(s)
Antígenos CD34/análisis , Fracturas del Fémur/terapia , Fémur/irrigación sanguínea , Leucocitos Mononucleares/trasplante , Neovascularización Fisiológica , Osteogénesis , Inhibidores de la Angiogénesis/farmacología , Animales , Movimiento Celular , Células Cultivadas , Células Endoteliales , Femenino , Fracturas del Fémur/patología , Fémur/patología , Expresión Génica , Humanos , Leucocitos Mononucleares/química , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Osteocalcina/genética , Osteogénesis/genética , ARN Mensajero/análisis , Ratas , Ratas Desnudas
11.
Circulation ; 113(10): 1311-25, 2006 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-16534028

RESUMEN

BACKGROUND: Multilineage developmental capacity of the CD34+ cells, especially into cardiomyocytes and smooth muscle cells (SMCs), is still controversial. In the present study we performed a series of experiments to prove our hypothesis that vasculogenesis and cardiomyogenesis after myocardial infarction (MI) may be dose-dependently enhanced after CD34+ cell transplantation. METHODS AND RESULTS: Peripheral blood CD34+ cells were isolated from total mononuclear cells of patients with limb ischemia by apheresis after 5-day administration of granulocyte colony-stimulating factor. PBS and 1x10(3) (low), 1x10(5) (mid), or 5x10(5) (high) CD34+ cells were intramyocardially transplanted after ligation of the left anterior descending coronary artery of nude rats. Functional assessments with the use of echocardiography and a microtip conductance catheter at day 28 revealed dose-dependent preservation of left ventricular function by CD34+ cell transplantation. Necropsy examination disclosed dose-dependent augmentation of capillary density and dose-dependent inhibition of left ventricular fibrosis. Immunohistochemistry for human-specific brain natriuretic peptide demonstrated that human cardiomyocytes were dose-dependently observed in ischemic myocardium at day 28 (high, 2480+/-149; mid, 1860+/-141; low, 423+/-9; PBS, 0+/-0/mm2; P<0.05 for high versus mid and mid versus low). Immunostaining for smooth muscle actin and human leukocyte antigen or Ulex europaeus lectin type 1 also revealed dose-dependent vasculogenesis by endothelial cell and SMC development after CD34+ cell transplantation. Reverse transcriptase-polymerase chain reaction indicated that human-specific gene expression of cardiomyocyte (brain natriuretic peptide, cardiac troponin-I, myosin heavy chain, and Nkx 2.5), SMC (smooth muscle actin and sm22alpha), and endothelial cell (CD31 and KDR) markers were dose-dependently augmented in MI tissue. CONCLUSIONS: Human CD34+ cell transplantation may have significant and dose-dependent potential for vasculogenesis and cardiomyogenesis with functional recovery from MI.


Asunto(s)
Antígenos CD34 , Trasplante de Células Madre Hematopoyéticas/métodos , Desarrollo de Músculos , Infarto del Miocardio/terapia , Miocardio/citología , Neovascularización Fisiológica , Animales , Recuento de Células , Electrocardiografía , Células Endoteliales/citología , Fibrosis/patología , Humanos , Infarto del Miocardio/patología , Isquemia Miocárdica/patología , Miocitos Cardíacos/patología , Miocitos del Músculo Liso/citología , Ratas , Ratas Desnudas , Trasplante Heterólogo , Resultado del Tratamiento , Función Ventricular Izquierda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...