Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
EBioMedicine ; 57: 102862, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32629392

RESUMEN

BACKGROUND: Bone marrow stem cell clonal dysfunction by somatic mutation is suspected to affect post-infarction myocardial regeneration after coronary bypass surgery (CABG). METHODS: Transcriptome and variant expression analysis was studied in the phase 3 PERFECT trial post myocardial infarction CABG and CD133+ bone marrow derived hematopoetic stem cells showing difference in left ventricular ejection fraction (∆LVEF) myocardial regeneration Responders (n=14; ∆LVEF +16% day 180/0) and Non-responders (n=9; ∆LVEF -1.1% day 180/0). Subsequently, the findings have been validated in an independent patient cohort (n=14) as well as in two preclinical mouse models investigating SH2B3/LNK antisense or knockout deficient conditions. FINDINGS: 1. Clinical: R differed from NR in a total of 161 genes in differential expression (n=23, q<0•05) and 872 genes in coexpression analysis (n=23, q<0•05). Machine Learning clustering analysis revealed distinct RvsNR preoperative gene-expression signatures in peripheral blood acorrelated to SH2B3 (p<0.05). Mutation analysis revealed increased specific variants in RvsNR. (R: 48 genes; NR: 224 genes). 2. Preclinical:SH2B3/LNK-silenced hematopoietic stem cell (HSC) clones displayed significant overgrowth of myeloid and immune cells in bone marrow, peripheral blood, and tissue at day 160 after competitive bone-marrow transplantation into mice. SH2B3/LNK-/- mice demonstrated enhanced cardiac repair through augmenting the kinetics of bone marrow-derived endothelial progenitor cells, increased capillary density in ischemic myocardium, and reduced left ventricular fibrosis with preserved cardiac function. 3. VALIDATION: Evaluation analysis in 14 additional patients revealed 85% RvsNR (12/14 patients) prediction accuracy for the identified biomarker signature. INTERPRETATION: Myocardial repair is affected by HSC gene response and somatic mutation. Machine Learning can be utilized to identify and predict pathological HSC response. FUNDING: German Ministry of Research and Education (BMBF): Reference and Translation Center for Cardiac Stem Cell Therapy - FKZ0312138A and FKZ031L0106C, German Ministry of Research and Education (BMBF): Collaborative research center - DFG:SFB738 and Center of Excellence - DFG:EC-REBIRTH), European Social Fonds: ESF/IV-WM-B34-0011/08, ESF/IV-WM-B34-0030/10, and Miltenyi Biotec GmbH, Bergisch-Gladbach, Germany. Japanese Ministry of Health : Health and Labour Sciences Research Grant (H14-trans-001, H17-trans-002) TRIAL REGISTRATION: ClinicalTrials.gov NCT00950274.


Asunto(s)
Antígeno AC133/genética , Trasplante de Médula Ósea/métodos , Enfermedad de la Arteria Coronaria/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Isquemia Miocárdica/terapia , Adolescente , Adulto , Anciano , Células de la Médula Ósea/citología , Senescencia Celular/genética , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/fisiopatología , Femenino , Corazón/crecimiento & desarrollo , Corazón/fisiopatología , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Persona de Mediana Edad , Isquemia Miocárdica/genética , Isquemia Miocárdica/patología , Regeneración/genética , Adulto Joven
2.
N Engl J Med ; 376(11): 1038-1046, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28296613

RESUMEN

We assessed the feasibility of transplanting a sheet of retinal pigment epithelial (RPE) cells differentiated from induced pluripotent stem cells (iPSCs) in a patient with neovascular age-related macular degeneration. The iPSCs were generated from skin fibroblasts obtained from two patients with advanced neovascular age-related macular degeneration and were differentiated into RPE cells. The RPE cells and the iPSCs from which they were derived were subject to extensive testing. A surgery that included the removal of the neovascular membrane and transplantation of the autologous iPSC-derived RPE cell sheet under the retina was performed in one of the patients. At 1 year after surgery, the transplanted sheet remained intact, best corrected visual acuity had not improved or worsened, and cystoid macular edema was present. (Funded by Highway Program for Realization of Regenerative Medicine and others; University Hospital Medical Information Network Clinical Trials Registry [UMIN-CTR] number, UMIN000011929 .).


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Degeneración Macular/terapia , Epitelio Pigmentado de la Retina/citología , Anciano , Técnicas de Cultivo de Célula , Diferenciación Celular , Estudios de Factibilidad , Femenino , Fibroblastos , Humanos , Masculino , Epitelio Pigmentado de la Retina/trasplante , Trasplante Autólogo
3.
Stem Cell Res ; 14(2): 165-76, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25644021

RESUMEN

Identification of pivotal factors potentially present in the in situ environment and capable of influencing the function of CD34(+) cells, which can be used for autologous cell therapy, is of paramount interest. SHh is one of the morphogens essential for embryonic vascular development as well as postnatal neovascularization, and the activation of SHh signaling with angiogenic and vascular differentiation responses in CD34(+) cells by SHh treatment differed depending on the G-CSF treatment or the background disease. SHh enhanced the migration, proliferation, adhesion, and EPC colony forming capacities of G-CSF mobilized CD34(+) cells, increasing the vasculogenic/angiogenic potential for neovascularization. An increase in the differentiation potential of CD34(+) cells toward vascular lineages was demonstrated with SHh treatment involving TGFß signaling pathway. The SHh-activated G-CSF mobilized CD34(+) cells directly contributed to vascular regeneration while non-activated CD34(+) cells showed a lower regenerative capacity in a mouse ischemic hindlimb model. SHh signaling regulates human CD34(+) cell fate and function, and may potentiate the therapeutic effect of G-CSF mobilized CD34(+) cells on ischemic diseases.


Asunto(s)
Antígenos CD34/sangre , Proteínas Hedgehog/metabolismo , Adulto , Animales , Diferenciación Celular/fisiología , Factor Estimulante de Colonias de Granulocitos , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/sangre , Isquemia/metabolismo , Masculino , Ratones , Ratones Desnudos , Proteínas Recombinantes/química , Transducción de Señal , Proteínas Smad/metabolismo , Factores de Crecimiento Transformadores/metabolismo
4.
J Bone Miner Res ; 30(1): 95-105, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25130304

RESUMEN

CXC chemokine receptor 4 (CXCR4) is a specific receptor for stromal-derived-factor 1 (SDF-1). SDF-1/CXCR4 interaction is reported to play an important role in vascular development. On the other hand, the therapeutic potential of endothelial progenitor cells (EPCs) in fracture healing has been demonstrated with mechanistic insight of vasculogenesis/angiogenesis and osteogenesis enhancement at sites of fracture. The purpose of this study was to investigate the influence of the SDF-1/CXCR4 pathway in Tie2-lineage cells (including EPCs) in bone formation. We created CXCR4 gene conditional knockout mice using the Cre/loxP system and set two groups of mice: Tie2-Cre(ER) CXCR4 knockout mice (CXCR4(-/-) ) and wild-type mice (WT). We report here that in vitro, EPCs derived from of CXCR4(-/-) mouse bone marrow demonstrated severe reduction of migration activity and EPC colony-forming activity when compared with those derived from WT mouse bone marrow. In vivo, radiological and morphological examinations showed fracture healing delayed in the CXCR4(-/-) group and the relative callus area at weeks 2 and 3 was significantly smaller in CXCR4(-/-) group mice. Quantitative analysis of capillary density at perifracture sites also showed a significant decrease in the CXCR4(-/-) group. Especially, CXCR4(-/-) group mice demonstrated significant early reduction of blood flow recovery at fracture sites compared with the WT group in laser Doppler perfusion imaging analysis. Real-time RT-PCR analysis showed that the gene expressions of angiogenic markers (CD31, VE-cadherin, vascular endothelial growth factor [VEGF]) and osteogenic markers (osteocalcin, collagen 1A1, bone morphogenetic protein 2 [BMP2]) were lower in the CXCR4(-/-) group. In the gain-of-function study, the fracture in the SDF-1 intraperitoneally injected WT group healed significantly faster with enough callus formation compared with the SDF-1 injected CXCR4(-/-) group. We demonstrated that an EPC SDF-1/CXCR4 axis plays an important role in bone fracture healing using Tie2-Cre(ER) CXCR4 conditional knockout mice.


Asunto(s)
Regeneración Ósea , Quimiocina CXCL12/metabolismo , Células Progenitoras Endoteliales/metabolismo , Fracturas Óseas/metabolismo , Receptor TIE-2/metabolismo , Receptores CXCR4/metabolismo , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/farmacología , Células Progenitoras Endoteliales/patología , Fracturas Óseas/dietoterapia , Fracturas Óseas/genética , Fracturas Óseas/patología , Ratones , Ratones Noqueados , Receptor TIE-2/genética , Receptores CXCR4/genética
5.
Cell Transplant ; 24(7): 1379-93, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24800622

RESUMEN

We recently demonstrated that the local transplantation of human peripheral blood (PB) CD34(+) cells, an endothelial/hematopoietic progenitor cell-rich population, contributes to fracture repair via vasculogenesis/angiogenesis and osteogenesis. Human PB mononuclear cells (MNCs) are also considered a potential cell fraction for neovascularization. We have previously shown the feasibility of human PB MNCs to enhance fracture healing. However, there is no report directly comparing the efficacy for fracture repair between CD34(+) cells and MNCs. In addition, an unhealing fracture model, which does not accurately resemble a clinical setting, was used in our previous studies. To overcome these issues, we compared the capacity of human granulocyte colony-stimulating factor-mobilized PB (GM-PB) CD34(+) cells and human GM-PB MNCs in a nonunion model, which more closely resembles a clinical setting. First, the effect of local transplantation of 1 × 10(5) GM-PB CD34(+) cells (CD34(+) group), 1 × 10(7) GM-PB MNCs (containing approximately 1 × 10(5) GM-PB CD34(+) cells) (MNC group), and phosphate-buffered saline (PBS) (PBS group) on nonunion healing was compared. Similar augmentation of blood flow recovery at perinonunion sites was observed in the CD34(+) and MNC groups. Meanwhile, a superior effect on nonunion repair was revealed by radiological, histological, and functional assessment in the CD34(+) group compared with the other groups. Moreover, through in vivo and in vitro experiments, excessive inflammation induced by GM-PB MNCs was confirmed and believed to be one of the mechanisms underlying this potency difference. These results strongly suggest that local transplantation of GM-PB CD34(+) cells is a practical and effective strategy for treatment of nonunion after fracture.


Asunto(s)
Antígenos CD34/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Leucocitos Mononucleares/metabolismo , Acondicionamiento Pretrasplante/métodos , Cicatrización de Heridas/efectos de los fármacos , Diferenciación Celular , Fracturas Óseas , Humanos
6.
J Am Heart Assoc ; 3(3): e000743, 2014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-24965023

RESUMEN

BACKGROUND: Cell-based therapies involving mononuclear cells (MNCs) have been developed for vascular regeneration to treat ischemic diseases; however, quality control of therapeutic MNCs has not been evaluated. We investigated the therapeutic potential of peripheral blood (PB) MNCs, operated by recently developed quality and quantity (QQ) culture of endothelial progenitor cells (EPCs). METHODS AND RESULTS: PBs were collected from healthy volunteers; peripheral blood mononuclear cells (PBMNCs) isolated from these PBs were subjected to QQ culture for 7 days with medium containing stem cell factor, thrombopoietin, Flt-3 ligand, vascular endothelial growth factor, and interleukin-6. The resulting cells (QQMNCs) in EPC colony-forming assay generated significantly more definitive EPC colonies than PBMNCs. In flow cytometry, macrophages and helper T lymphocytes of QQMNCs became phenotypically polarized into angiogenic, anti-inflammatory, and regenerative subsets: classical M1 to alternative M2; T helper (Th)1 to Th2; angiogenic or regulatory T-cell expansion. Quantitative real-time polymerase chain reaction (qRT-PCR) assay revealed the predominant proangiogenic gene expressions in QQMNCs versus PBMNCs. Using murine ischemic hindlimb models, the efficacy of QQMNC intramuscular transplantation (Tx) was compared to that of PBMNCTx, cultured "early EPC" Tx (eEPCTx), and granulocyte colony-stimulating factor mobilized CD34(+) cell Tx (GmCD34Tx). Laser Doppler imaging revealed the blood perfusion recovery in ischemic hindlimbs after QQMNCTx superior to after PBMNCTx and eEPCTx, but also earlier than after GmCD34Tx. Histological evaluations and qRT-PCR assays in ischemic hindlimbs demonstrated that QQMNCTx, similarly to GmCD34Tx, enhanced angiovasculogenesis and myogenesis, whereas it preponderantly inhibited inflammation and fibrosis versus PBMNCTx and eEPCTx. CONCLUSIONS: QQ culture potentiates the ability of PBMNCs to promote regeneration of injured tissue; considering the feasible cell preparation, QQ culture-treated PBMNCs may provide a promising therapeutic option for ischemic diseases. CLINICAL TRIAL REGISTRATION URL: irb.med.u-tokai.ac.jp/d/2/monthly/2010.html; IRB No.: 10R-020.URL: irb.med.u-tokai.ac.jp/d/2/monthly/201312.html; IRB No.: 13R228.


Asunto(s)
Células Progenitoras Endoteliales/fisiología , Leucocitos Mononucleares/fisiología , Macrófagos/fisiología , Linfocitos T/fisiología , Animales , Vasos Sanguíneos/fisiología , Células Cultivadas , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/efectos de los fármacos , Citometría de Flujo , Humanos , Interleucina-6/farmacología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Activación de Linfocitos/fisiología , Macrófagos/citología , Masculino , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos BALB C , Neovascularización Fisiológica/fisiología , Regeneración/fisiología , Factor de Células Madre/farmacología , Linfocitos T/citología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/fisiología , Trombopoyetina/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología
7.
Physiol Rep ; 2(3): e00261, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24760515

RESUMEN

Abstract Endothelial progenitor cells (EPCs) have been demonstrated to be effective for the treatment of cardiovascular diseases. However, the differentiation process from circulation to adhesion has not been clarified because circulating EPCs rarely attached to dishes in EPC cultures previously. Here we investigated whether immature circulating EPCs differentiate into mature adhesive EPCs in response to dextran. When floating-circulating EPCs derived from ex vivo expanded human cord blood were cultured with 5% and 10% dextran, they attached to fibronectin-coated dishes and grew exponentially. The bioactivities of adhesion, proliferation, migration, tube formation, and differentiated type of EPC colony formation increased in EPCs exposed to dextran. The surface protein expression rate of the endothelial markers vascular endothelial growth factor (VEGF)-R1/2, VE-cadherin, Tie2, ICAM1, VCAM1, and integrin αv/ß3 increased in EPCs exposed to dextran. The mRNA levels of VEGF-R1/2, VE-cadherin, Tie2, endothelial nitric oxide synthase, MMP9, and VEGF increased in EPCs treated with dextran. Those of endothelium-related transcription factors ID1/2, FOXM1, HEY1, SMAD1, FOSL1, NFkB1, NRF2, HIF1A, EPAS1 increased in dextran-treated EPCs; however, those of hematopoietic- and antiangiogenic-related transcription factors TAL1, RUNX1, c-MYB, GATA1/2, ERG, FOXH1, HHEX, SMAD2/3 decreased in dextran-exposed EPCs. Inhibitor analysis showed that PI3K/Akt, ERK1/2, JNK, and p38 signal transduction pathways are involved in the differentiation in response to dextran. In conclusion, dextran induces differentiation of circulating EPCs in terms of adhesion, migration, proliferation, and vasculogenesis. The differentiation mechanism in response to dextran is regulated by multiple signal transductions including PI3K/Akt, ERK1/2, JNK, and p38. These findings indicate that dextran is an effective treatment for EPCs in regenerative medicines.

8.
Circ J ; 78(1): 222-31, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24107361

RESUMEN

BACKGROUND: Because human cardiac stem cells (CSC) have regeneration potential in damaged cardiac tissue, there is increasing interest in using them in cell-based therapies for cardiac failure. However, culture conditions, by which CSCs are expanded while maintaining their therapeutic potential, have not been optimized. We hypothesized that the plating cell-density would affect proliferation activity, differentiation and therapeutic potential of CSCs through the Notch signaling pathway. METHODS AND RESULTS: Human CSCs were plated at 4 different densities. The population doubling time, C-KIT positivity, and dexamethasone-induced multidifferentiation potential were examined in vitro. The therapeutic potential of CSCs was assessed by transplanting them into a rat acute myocardial infarction (AMI) model. The low plating density (340cells/cm(2)) maintained the multidifferentiation potential with greater proliferation activity and C-KIT positivity in vitro. On the other hand, the high plating density (5,500cells/cm(2)) induced autonomous differentiation into endothelial cells by activating Notch signaling in vitro. CSCs cultured at low or high density with Notch signal inhibitor showed significantly greater therapeutic potential in vivo compared with those cultured at high density. CONCLUSIONS: CSCs cultured with reduced Notch signaling showed better cardiomyogenic differentiation and therapeutic potentials in a rat AMI model. Thus, reducing Notch signaling is important when culturing CSCs for clinical applications.


Asunto(s)
Infarto del Miocardio , Miocardio , Receptores Notch/metabolismo , Transducción de Señal , Trasplante de Células Madre , Células Madre , Adulto , Animales , Células Cultivadas , Niño , Femenino , Xenoinjertos , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Desnudas , Células Madre/metabolismo , Células Madre/patología
9.
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
10.
Stem Cells Transl Med ; 1(2): 160-71, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23197763

RESUMEN

Quantitative and qualitative impairment of endothelial progenitor cells (EPCs) limits the efficacy of autologous cell therapy in patients with cardiovascular diseases. Here, we developed a serum-free quality and quantity control culture system for colony-forming EPCs to enhance their regenerative potential. A culture with serum-free medium containing stem cell factor, thrombopoietin, vascular endothelial growth factor, interleukin-6, and Flt-3 ligand was determined as optimal quality and quantity culture (QQc) in terms of the most vasculogenic colony-forming EPC expansion, evaluated by the newly established EPC colony formation assay. The QQc of umbilical cord blood-CD133(+) cells for 7 days produced a 52.9-fold increase in total cell number and 3.28-fold frequency in definitive EPC colony development, resulting in a 203.9-fold increase in estimated total definitive EPC colony number in vitro. Pre- or post-QQc cells were intramyocardially transplanted into nude rats with myocardial infarction (MI). Echocardiographic and micromanometer-tipped conductance catheter examinations 28 days post-MI revealed significant preservation of left ventricular (LV) function in rats receiving pre- or post-QQc cells compared with those receiving phosphate-buffered saline. Assessments of global LV contractility indicated a dose-dependent effect of pre- or post-QQc cells and the superior potency of post-QQc cells over pre-QQc cells. Furthermore, immunohistochemistry showed more abundant formation of both human and rat endothelial cells and cardiomyocytes in the infarcted myocardium following transplantation of post-QQc cells compared with pre-QQc cells. Our optimal serum-free quality and quantity culture may enhance the therapeutic potential of EPCs in both quantitative and qualitative aspects for cardiovascular regeneration.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Ensayo de Unidades Formadoras de Colonias/métodos , Medio de Cultivo Libre de Suero/metabolismo , Células Endoteliales/citología , Neovascularización Fisiológica , Células Madre/citología , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Tampones (Química) , Recuento de Células , Técnicas de Cultivo de Célula/normas , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/normas , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias/normas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ecocardiografía , Células Endoteliales/metabolismo , Células Endoteliales/trasplante , Sangre Fetal/citología , Sangre Fetal/metabolismo , Glicoproteínas/metabolismo , Humanos , Inmunohistoquímica , Contracción Miocárdica , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/trasplante , Péptidos/metabolismo , Control de Calidad , Ratas , Ratas Desnudas , Células Madre/metabolismo , Función Ventricular Izquierda
11.
Cell Transplant ; 21(12): 2689-709, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22943882

RESUMEN

Transplantation of bone marrow (BM) CD34(+) cells, an endothelial/hematopoietic progenitor-enriched cell population, has shown therapeutic efficiency in the treatment of ischemic diseases enhancing neovascularization. However, the number of CD34(+) cells obtained from bone marrow is not sufficient for routine clinical application. To overcome this issue, we developed a more efficient and clinically applicable CD34(+) cell expansion method. Seven-day ex vivo expansion culture of BM CD34(+) cells with a cocktail of five growth factors containing VEGF, SCF, IL-6, Flt-3 ligand, and TPO resulted in reproducible more than 20-fold increase in cell number. The favorable effect of the local transplantation of culture expanded (cEx)-BM CD34(+) cells on rat unhealing fractures was equivalent or higher than that of nonexpanded (fresh) BM CD34(+) cells exhibiting sufficient therapeutic outcome with frequent vasculogenic/osteogenic differentiation of transplanted cEx-BM CD34(+) cells and fresh BM CD34(+) cells as well as intrinsic enhancement of angiogenesis/osteogenesis at the treated fracture sites. Specifically, cEx-BM CD34(+) cell treatment demonstrated the best blood flow recovery at fracture sites compared with the nonexpanded BM CD34(+) cells. In vitro, cEx-BM CD34(+) cells showed higher colony/tube-forming capacity than nonexpanded BM CD34(+) cells. Both cells demonstrated differentiation potential into osteoblasts. Since fresh BM CD34(+) cells can be easily collected from fracture sites at the time of primary operation and stored for future use, autologous cEx-BM CD34(+) cell transplantation would be not only a simple but also a promising therapeutic strategy for unhealing fractures in the field of orthopedic trauma surgery.


Asunto(s)
Antígenos CD34/metabolismo , Células de la Médula Ósea/citología , Fracturas del Fémur/terapia , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Animales , Velocidad del Flujo Sanguíneo , Huesos/irrigación sanguínea , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Fracturas del Fémur/diagnóstico por imagen , Fracturas del Fémur/patología , Células Madre Hematopoyéticas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Neovascularización Patológica , Osteogénesis , Ratas , Ratas Desnudas , Tomografía Computarizada por Rayos X , Adulto Joven
12.
J Bone Miner Res ; 27(5): 1118-31, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22275312

RESUMEN

Several reports have shown the therapeutic effect of statins on bone formation and neovascularization. However, the effect of the systemic administration of statins is limited due to its metabolism in the liver and clearance in the digestive system. In addition, high-dose administration may cause adverse side effects. To avoid low-efficacy/frequent side effects of high-dose statin treatment, we utilized biodegradable gelatin hydrogel as a drug delivery system of statin for fracture healing. A femoral fracture was created in rats with periosteum cauterization leading to nonunion at 8 weeks postfracture. Rats received local administration of either simvastatin-conjugated gelatin hydrogel (ST-Gel group) or gelatin hydrogel alone (Gel group). Approximately 70% of animals in the ST-Gel group achieved fracture union radiographically and histologically, while only 7% of animals achieved fracture healing in the Gel group. Functional bone healing was also significantly greater with increased angiogenesis- and osteogenesis-related growth factor expressions in periosteal granulation tissue in the ST-Gel group than in the Gel group. Simvastatin locally applied with gelatin hydrogel to fracture sites at a dose similar to that used in clinical settings successfully induced fracture union in a rat unhealing bone fracture model via its effect on both angiogenesis and osteogenesis.


Asunto(s)
Curación de Fractura/efectos de los fármacos , Gelatina/farmacología , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Simvastatina/administración & dosificación , Simvastatina/farmacología , Adyuvantes Farmacéuticos/farmacología , Administración Tópica , Animales , Trasplante de Médula Ósea , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Sistemas de Liberación de Medicamentos , Femenino , Fémur/irrigación sanguínea , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Radiografía , Ratas
13.
PLoS One ; 6(5): e20219, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21655289

RESUMEN

BACKGROUND: Endothelial progenitor cells (EPCs) were shown to have angiogenic potential contributing to neovascularization. However, a clear definition of mouse EPCs by cell surface markers still remains elusive. We hypothesized that CD34 could be used for identification and isolation of functional EPCs from mouse bone marrow. METHODOLOGY/PRINCIPAL FINDINGS: CD34(+) cells, c-Kit(+)/Sca-1(+)/Lin(-) (KSL) cells, c-Kit(+)/Lin(-) (KL) cells and Sca-1(+)/Lin(-) (SL) cells were isolated from mouse bone marrow mononuclear cells (BMMNCs) using fluorescent activated cell sorting. EPC colony forming capacity and differentiation capacity into endothelial lineage were examined in the cells. Although CD34(+) cells showed the lowest EPC colony forming activity, CD34(+) cells exhibited under endothelial culture conditions a more adherent phenotype compared with the others, demonstrating the highest mRNA expression levels of endothelial markers vWF, VE-cadherin, and Flk-1. Furthermore, a dramatic increase in immediate recruitment of cells to the myocardium following myocardial infarction and systemic cell injection was observed for CD34(+) cells comparing with others, which could be explained by the highest mRNA expression levels of key homing-related molecules Integrin ß2 and CXCR4 in CD34(+) cells. Cell retention and incorporation into the vasculature of the ischemic myocardium was also markedly increased in the CD34(+) cell-injected group, giving a possible explanation for significant reduction in fibrosis area, significant increase in neovascularization and the best cardiac functional recovery in this group in comparison with the others. CONCLUSION: These findings suggest that mouse CD34(+) cells may represent a functional EPC population in bone marrow, which could benefit the investigation of therapeutic EPC biology.


Asunto(s)
Antígenos CD34/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Células Cultivadas , Ecocardiografía , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Circ Res ; 109(1): 20-37, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21566217

RESUMEN

The precise and conceptual insight of circulating endothelial progenitor cell (EPC) kinetics is hampered by the absence of an assay system capable of evaluating the EPC differentiation cascade. An assay system for EPC colony formation was developed to delineate circulating EPC differentiation. EPC colony-forming assay using semisolid medium and single or bulk CD133(+) cells from umbilical cord blood exhibited the formation of two types of attaching cell colonies made of small or large cells featuring endothelial lineage potential and properties, termed small EPC colony-forming units and large EPC colony-forming units, respectively. In vitro and in vivo assays of each EPC colony-forming unit cell revealed a differentiation hierarchy from small EPC to large EPC colonies, indicating a primitive EPC stage with highly proliferative activity and a definitive EPC stage with vasculogenic properties, respectively. Experimental comparison with a conventional EPC culture assay system disclosed EPC colony-forming unit cells differentiate into noncolony-forming early EPC. The fate analysis of single CD133(+) cells into the endothelial and hematopoietic lineage was achieved by combining this assay system with a hematopoietic progenitor assay and demonstrated the development of colony-forming EPC and hematopoietic progenitor cells from a single hematopoietic stem cell. EPC colony-forming assay permits the determination of circulating EPC kinetics from single or bulk cells, based on the evaluation of hierarchical EPC colony formation. This assay further enables a proper exploration of possible links between the origin of EPC and hematopoietic stem cells, representing a novel and powerful tool to investigate the molecular signaling pathways involved in EPC biology.


Asunto(s)
Ensayo de Unidades Formadoras de Colonias/métodos , Células Endoteliales/citología , Células Madre/citología , Antígeno AC133 , Adulto , Animales , Antígenos CD/análisis , Diferenciación Celular , Células Cultivadas , Glicoproteínas/análisis , Células Madre Hematopoyéticas/citología , Humanos , Receptores de Lipopolisacáridos/análisis , Ratones , Ratones Endogámicos BALB C , Péptidos/análisis , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/farmacología
15.
PLoS One ; 5(12): e15365, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21179535

RESUMEN

Hedgehog (Hh) signalling plays an important role in various developmental processes by activating the Cubitus interruptus (Ci)/Glioblastoma (Gli) family of transcription factors. In the process of proper pattern formation, Ci activity is regulated by multiple mechanisms, including processing, trafficking, and degradation. However, it remains elusive how Ci distinctly recognizes the strong and moderate Hh signals. Roadkill (Rdx) induces Ci degradation in the anterior region of the Drosophila wing disc. Here, we report that Rdx inhibited Ci activity by two different mechanisms. In the region abutting the anterior/posterior boundary, which receives strong Hh signal, Rdx inhibited the nuclear import of Ci by releasing importin α3 from Ci. In this region, Rdx negatively regulated the expression of transcription factor Knot/Collier. In farther anterior regions receiving moderate levels of Hh signal, Rdx induced Ci degradation, as reported previously. Thus, two different mechanisms by which Rdx negatively regulates Ci may play an important role in the fine-tuning of Hh responses.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Animales , Cruzamientos Genéticos , Drosophila melanogaster , Genotipo , Modelos Genéticos , Mutación , Plásmidos/metabolismo , Isoformas de Proteínas , Interferencia de ARN , Técnicas del Sistema de Dos Híbridos
16.
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
17.
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
18.
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
19.
J Mol Cell Cardiol ; 43(5): 627-35, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17900610

RESUMEN

Therapeutic angiogenesis can be induced by the implantation of bone marrow cells (BMCs). However, the mechanism of BMC-mediated neovascularization remains to be clarified. We investigated the differential activities of bone marrow subpopulations in angiogenesis and cytokine production. BMCs were separated into positive and negative fractions by surface expression of Mac-1, Gr-1, CD19, and c-kit, respectively. After 7 days of culture in the presence of vascular endothelial growth factor (VEGF), the cells produced adherent cells which incorporate acetylated low-density lipoprotein (acLDL). Mac-1(+) and Mac-1(-) cells produced almost equal numbers of acLDL(+) cells, but only Mac-1(-) cells expressed endothelial markers, including Flk-1, vWF, and CD31. Similarly, the expression of endothelial markers was detected in Gr-1(-), CD19(-), and c-kit(+) BMC fractions at 7-day cultures, but not in Gr-1(+), CD19(+), or c-kit(-) cells. In contrast, freshly isolated Mac-1(+) and Gr-1(+) BMCs expressed higher levels of mRNAs for angiogenic cytokines (including VEGF-A, FGF-2, and HGF) than Mac-1(-) and Gr-1(-) cells, respectively. Moreover, Mac-1(+)/c-kit(+) BMC subpopulation expressed higher levels of VEGF-A and SDF-1 mRNAs than other subpopulations. These data demonstrate that a relatively small proportion of VEGF-cultured adherent cells are true endothelial cells with a Flk-1(+)/vWF(+)/CD31(+) phenotype. Moreover, endothelial stem/progenitor cells (EPCs) are limited primarily to Mac-1(-), Gr-1(-), and c-kit(+) BMC populations. In contrast, angiogenic cytokine mRNAs were also produced by Mac-1(+), Gr-1(+), and c-kit(-) BMCs, suggesting the heterogeneity of effector cell types for neovasculatization therapy.


Asunto(s)
Células de la Médula Ósea/citología , Citocinas/genética , Endotelio Vascular/citología , Células Madre/citología , Animales , Células de la Médula Ósea/fisiología , Técnicas de Cultivo de Célula , Separación Celular , Endotelio Vascular/fisiología , Fémur , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Stem Cells ; 25(7): 1664-74, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17446562

RESUMEN

Although there is a criticism that embryonic stem (ES) cell differentiation does not always reflect the differentiation process involved in mouse development, it is a suitable model system to dissect the specific differentiation pathway. We established the culture conditions that selectively differentiated mouse ES cells into three germ layers containing mesendoderm, definitive endoderm (DE), visceral endoderm (VE), mesoderm, and neuroectoderm. However, the molecular mechanisms of differentiation under each specific condition still remain unclear. Here, in combination with the RNA interference-mediated gene knockdown (KD) method, we show that Eomesodermin (Eomes), Mixl1, Brachyury (T), and GATA6 are major molecular determinants in the differentiation of mesendoderm, DE, VE, and mesoderm. Eomes plays a pivotal role in an early stage of mesendoderm differentiation, whereas Mixl1 does the same in the later stage where mesendoderm differentiates into DE. Further analyses of quantitative reverse transcription polymerase chain reaction and overexpression of Mixl1 demonstrated that Mixl1 is genetically a downstream molecule of Eomes. In addition, both Eomes and Mixl1 act as negative regulators of T expression. This strategy also reveals that Eomes and T play cell-autonomous roles in platelet-derived growth factor receptor alpha (PDGFRalpha)+ vascular endothelial growth factor receptor 2 (VEGFR2)+ and PDGFRalpha+ mesoderm generations, respectively. Our results obtained from this study are fully consistent with previous knockout studies of those genes. The present study, therefore, demonstrates that the major molecular mechanism underlying in vitro ES cell differentiation largely recapitulates that in actual embryogenesis, and the combination of our culture system and RNAi-mediated gene KD is an useful tool to elucidate the molecular hierarchy in in vitro ES cell differentiation. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Endodermo/citología , Mesodermo/citología , Interferencia de ARN , Activinas/metabolismo , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Factor de Transcripción GATA6/metabolismo , Expresión Génica , Proteínas de Homeodominio/metabolismo , Ratones , Modelos Biológicos , Proteínas de Dominio T Box/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...