Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Front Med (Lausanne) ; 9: 681316, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35360717

RESUMEN

This study was conducted to evaluate the safety and efficacy of human peripheral blood CD34 positive (CD34+) cells transplanted into a murine chronic stroke model to obtain pre-clinical proof of concept, prior to clinical testing. Granulocyte colony stimulating factor (G-CSF) mobilized human CD34+ cells [1 × 104 cells in 50 µl phosphate-buffered saline (PBS)] were intravenously (iv) or intra-carotid arterially (ia) transplanted 4 weeks after the induction of stroke (chronic stage), and neurological function was evaluated. In this study, severe combined immune deficiency (SCID) mice were used to prevent excessive immune response after cell therapy. Two weeks post cell therapy, the ia CD34+ cells group demonstrated a significant improvement in neurological functions compared to the PBS control. The therapeutic effect was maintained 8 weeks after the treatment. Even after a single administration, ia transplantation of CD34+ cells had a significant therapeutic effect on chronic stroke. Based on the result of this pre-clinical proof of concept study, a future clinical trial of autologous peripheral blood CD34+ cells administration in the intra-carotid artery for chronic stroke patients is planned.

2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Cell Transplant ; 20(9): 1491-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21176407

RESUMEN

Although implantation of crude bone marrow cells has been applied in a small number of patients for fracture healing, transplantation of peripheral blood CD34(+) cells, the hematopoietic/endothelial progenitor cell-enriched population, in patients with fracture has never been reported. Here, we report the first case of tibial nonunion receiving autologous, granulocyte colony stimulating factor mobilized CD34(+) cells accompanied with autologous bone grafting. No serious adverse event occurred, and the novel therapy performed 9 months after the primary operation resulted in bone union 3 months later without any symptoms including pain and gait disturbance.


Asunto(s)
Antígenos CD34/metabolismo , Fracturas no Consolidadas/terapia , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Fracturas de la Tibia/terapia , Adulto , Fracturas no Consolidadas/diagnóstico por imagen , Humanos , Masculino , Atención Perioperativa , Fracturas de la Tibia/diagnóstico por imagen , Tomografía Computarizada por Rayos X
10.
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
11.
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
12.
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
13.
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
14.
Stem Cells ; 27(11): 2857-64, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19711453

RESUMEN

A number of preclinical studies have indicated the therapeutic potential of endothelial progenitor cells for vascular regeneration in ischemic diseases. A phase I/IIa clinical trial of transplantation of autologous CD34(+) cells, the endothelial and hematopoietic progenitor-enriched fraction, was performed in no-option patients with atherosclerotic peripheral artery disease or Buerger's disease with critical limb ischemia (CLI). CD34(+) cells were isolated from the G-CSF-mobilized apheresis product using a magnetic cell sorting system. CD34(+) cells (10(5)/kg, n = 6; 5 x 10(5)/kg, n = 8; or 10(6)/kg, n = 3) were injected i.m. into the leg with more severe ischemia. The Efficacy Score, representing changes in the toe brachial pressure index (TBPI), Wong-Baker FACES pain rating scale, and total walking distance 12 weeks after cell transplantation, the primary endpoint, was positive, indicating improvement in limb ischemia in all patients, although no significant dose-response relationship was observed. During the 12-week observation after cell therapy, the Wong-Baker FACES pain rating scale, TBPI, transcutaneous partial oxygen pressure, total or pain-free walking distance, and ulcer size serially improved in all patients. No death or major amputation occurred, and severe adverse events were rare, although mild to moderate events relating to G-CSF and leukapheresis were frequent during the 12-week follow-up. In conclusion, the outcomes of this prospective clinical study indicate the safety and feasibility of CD34(+) cell therapy in patients with CLI. Favorable trends in efficacy parameters encourage a randomized and controlled trial in the future.


Asunto(s)
Antígenos CD34/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Factor Estimulante de Colonias de Granulocitos/metabolismo , Isquemia/terapia , Pierna/patología , Células Madre/citología , Adulto , Anciano , Anciano de 80 o más Años , Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Femenino , Humanos , Inyecciones Intramusculares , Masculino , Trasplante de Células Madre , Células Madre/metabolismo , Trasplante Autólogo , Resultado del Tratamiento
15.
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
16.
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
17.
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
18.
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
19.
Arterioscler Thromb Vasc Biol ; 25(7): 1388-94, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15860746

RESUMEN

OBJECTIVE: Previous studies from our laboratory have shown therapeutic potential of ex vivo expanded endothelial progenitor cells (EPCs) for myocardial ischemia. Our purpose was to investigate the mechanisms regulating EPC contribution to myocardial regeneration. METHODS AND RESULTS: To evaluate niche-dependent expression profiles of EPCs in vitro, we performed coculture using cultured EPCs derived from human peripheral blood and rat cardiac myoblast cell line (H9C2). Reverse-transcription polymerase chain reaction (PCR) disclosed the expression of human-specific cardiac markers as well as human-specific smooth muscle markers. Cytoimmunochemistry presented several cocultured cells stained with human specific cardiac antibody. To prove this translineage differentiation in vivo, human cultured EPCs were injected into nude rat myocardial infarction model. Reverse-transcription PCR as well as immunohistochemistry of rat myocardial samples demonstrated the expression of human specific cardiac, vascular smooth muscle, and endothelial markers. We observed the distribution of colors (Qtracker; Quantum Dot Corp) in coculture to detect the fused cells, and the frequency of cell fusion was <1%. CONCLUSIONS: EPCs can contribute to not only vasculogenesis but also myogenesis in the ischemic myocardium in vivo. Transdifferentiation, not cell fusion, is dominant for EPCs commitment to myocardial lineage cells. Ex vivo expanded EPCs transplantation might have enhanced therapeutic potential for myocardial regeneration.


Asunto(s)
Fusión Celular , Células Endoteliales/citología , Células Endoteliales/trasplante , Mioblastos Cardíacos/citología , Trasplante de Células Madre , Células Madre/citología , Animales , Especificidad de Anticuerpos , Biomarcadores , Linaje de la Célula/fisiología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/inmunología , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Mioblastos Cardíacos/inmunología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/inmunología , Ratas , Ratas Desnudas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...