Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Circulation ; 136(20): 1939-1954, 2017 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-28972000

RESUMEN

BACKGROUND: Human pluripotent stem cell (hPSC)-derived endothelial cells (ECs) have limited clinical utility because of undefined components in the differentiation system and poor cell survival in vivo. Here, we aimed to develop a fully defined and clinically compatible system to differentiate hPSCs into ECs. Furthermore, we aimed to enhance cell survival, vessel formation, and therapeutic potential by encapsulating hPSC-ECs with a peptide amphiphile (PA) nanomatrix gel. METHODS: We induced differentiation of hPSCs into the mesodermal lineage by culturing on collagen-coated plates with a glycogen synthase kinase 3ß inhibitor. Next, vascular endothelial growth factor, endothelial growth factor, and basic fibroblast growth factor were added for endothelial lineage differentiation, followed by sorting for CDH5 (VE-cadherin). We constructed an extracellular matrix-mimicking PA nanomatrix gel (PA-RGDS) by incorporating the cell adhesive ligand Arg-Gly-Asp-Ser (RGDS) and a matrix metalloproteinase-2-degradable sequence. We then evaluated whether the encapsulation of hPSC-CDH5+ cells in PA-RGDS could enhance long-term cell survival and vascular regenerative effects in a hind-limb ischemia model with laser Doppler perfusion imaging, bioluminescence imaging, real-time reverse transcription-polymerase chain reaction, and histological analysis. RESULTS: The resultant hPSC-derived CDH5+ cells (hPSC-ECs) showed highly enriched and genuine EC characteristics and proangiogenic activities. When injected into ischemic hind limbs, hPSC-ECs showed better perfusion recovery and higher vessel-forming capacity compared with media-, PA-RGDS-, or human umbilical vein EC-injected groups. However, the group receiving the PA-RGDS-encapsulated hPSC-ECs showed better perfusion recovery, more robust and longer cell survival (> 10 months), and higher and prolonged angiogenic and vascular incorporation capabilities than the bare hPSC-EC-injected group. Surprisingly, the engrafted hPSC-ECs demonstrated previously unknown sustained and dynamic vessel-forming behavior: initial perivascular concentration, a guiding role for new vessel formation, and progressive incorporation into the vessels over 10 months. CONCLUSIONS: We generated highly enriched hPSC-ECs via a clinically compatible system. Furthermore, this study demonstrated that a biocompatible PA-RGDS nanomatrix gel substantially improved long-term survival of hPSC-ECs in an ischemic environment and improved neovascularization effects of hPSC-ECs via prolonged and unique angiogenic and vessel-forming properties. This PA-RGDS-mediated transplantation of hPSC-ECs can serve as a novel platform for cell-based therapy and investigation of long-term behavior of hPSC-ECs.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/trasplante , Isquemia/terapia , Metaloproteinasa 2 de la Matriz/administración & dosificación , Nanoestructuras/administración & dosificación , Oligopéptidos/administración & dosificación , Células Madre Pluripotentes/trasplante , Animales , Diferenciación Celular/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Células Endoteliales/fisiología , Células Endoteliales/trasplante , Miembro Posterior/irrigación sanguínea , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Isquemia/fisiopatología , Masculino , Ratones , Ratones Desnudos , Células Madre Pluripotentes/fisiología , Distribución Aleatoria , Resultado del Tratamiento
2.
Circ Res ; 107(5): 602-14, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20634489

RESUMEN

RATIONALE: Bone marrow (BM) cells play an important role in physiological and therapeutic neovascularization. However, it remains unclear whether any specific uncultured BM cell populations have higher angiogenic and vasculogenic activities. Moreover, there has been controversy regarding the vasculogenic ability of BM cells. OBJECTIVE: Preliminary flow cytometric analysis showed that CD31, traditionally a marker for endothelial cells, is expressed in certain nonendothelial BM mononuclear cells in both human and mouse. Based on the conserved CD31 expression in the axis of hematopoietic stem/progenitor cells (HSC/HPCs) to endothelial cells, we further sought to determine the comprehensive vasculogenic and angiogenic characteristics of human and mouse BM-derived CD31(+) cells. METHODS AND RESULTS: Flow cytometric analysis demonstrated that all CD31(+) cells derived from BM were CD45(+) and expressed markers for both HSC/HPCs and endothelial cells. Comprehensive gene expression analyses revealed that BM-CD31(+) cells expressed higher levels of angiogenic genes than CD31(-) cells. Endothelial progenitor cells, as well as HSC/HPCs, were almost exclusively confined to the CD31(+) cell fraction, and culture of CD31(+) cells under defined conditions gave rise to endothelial cells. Finally, injection of CD31(+) cells into ischemic hindlimb repaired ischemia, increased expression of angiogenic and chemoattractive factors, and, in part, directly contributed to vasculogenesis, as demonstrated by both 3D confocal microscopy and flow cytometry. CONCLUSIONS: These data indicate that BM-CD31(+) cells represent highly angiogenic and vasculogenic cells and can be a novel and highly promising source of cells for cell therapy to treat ischemic cardiovascular diseases.


Asunto(s)
Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Isquemia/cirugía , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteínas Angiogénicas/metabolismo , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Factores Quimiotácticos/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Miembro Posterior , Humanos , Inmunofenotipificación , Isquemia/inmunología , Isquemia/metabolismo , Isquemia/fisiopatología , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Microscopía Confocal , Neovascularización Fisiológica/genética , Fenotipo , Factores de Tiempo
3.
Sci Rep ; 11(1): 3630, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33574435

RESUMEN

Preeclampsia (PE) is a prevalent pregnancy disorder that leads to high maternal and fetal morbidity and mortality. While defective vascular development and angiogenesis in placenta are known as crucial pathological findings, its pathophysiological mechanism remains elusive. To better understand the effects of PE on angio-vasculogenesis and inflammatory networks in the fetus and to identify their biological signatures, we investigated the quantitative and functional characteristics of cord blood-derived mononuclear cells (CB-MNCs) and CD31-positive MNCs. Flow cytometry analysis demonstrated that the CB-MNCs from the severe PE group had significantly decreased number of cells expressing CD3, CD11b, CD14, CD19, KDR, and CD31 compared with the normal group. Quantitative real time PCR (qRT-PCR) shows down-regulation of the major angiogenic factor VEGFA in MNCs and CD31+ MNCs in severe PE. The major inflammatory cytokines IL1 was highly upregulated in CD31+ CB-MNCs in the severe PE patients. Mild PE patients, however, did not display any significant difference in expression of all measured angiogenic genes and most inflammatory genes. These findings show distinct angiogenic and inflammatory signatures from severe PE, and they may play a significant role in the pathogenesis of vascular defects in placenta of severe PE.


Asunto(s)
Sangre Fetal/citología , Inflamación/patología , Neovascularización Fisiológica , Preeclampsia/patología , Adulto , Femenino , Feto/patología , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Masculino , Neovascularización Fisiológica/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo
4.
Toxicon ; 51(3): 406-17, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18155118

RESUMEN

To examine the toxicological effect of saxatilin, a disintegrin isolated from the venom of a Korean snake (Gloydius saxatilis), recombinant saxatilin was highly expressed as a biologically active form in Pichia pastoris, and was successfully purified to homogeneity from the culture broth supernatant. The molecular and biological properties of the recombinant protein were the same as those of its natural form. Plasma half-life of the protein in rat was determined to 13.8 min. The maximum tolerated dose of the recombinant saxatilin was examined in ICR mice. The determined LD(50) values were 400 and 600 mg/kg of the body weight of a male and female mouse, respectively. To investigate the repeated dose toxicity of saxatilin in mice, the test item was intravenously administered to groups of ICR mice every day for 4 weeks. We observed a decrease in locomotor activity, piloerection, and crouching in clinical findings, a decrease of red blood cells (RBCs) in hematology, and hyperplasia of the spleen in histology related to administration of the test item. These results suggest that the target organ of intravenous administration of the test item is the spleen. The no adverse effect level (NOAEL) in this test for both males and females is considered to be 3mg/kg. Our results also indicate that recombinant saxatilin is non-toxic at an administration dose with an anti-platelet effect, and might be a potential anti-adhesion therapeutic agent for thrombosis, cancer, restenosis, cataract, and osteoporosis.


Asunto(s)
Desintegrinas/administración & dosificación , Desintegrinas/toxicidad , Viperidae , Animales , Desintegrinas/química , Desintegrinas/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Regulación de la Expresión Génica , Concentración de Iones de Hidrógeno , Inyecciones Intravenosas , Masculino , Ratones , Ratones Endogámicos ICR , Tamaño de los Órganos/efectos de los fármacos , Organismos Modificados Genéticamente , Pichia/genética , Pichia/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidad , Bazo/efectos de los fármacos , Bazo/patología , Orina , Pérdida de Peso/efectos de los fármacos
5.
Toxicon ; 52(3): 474-80, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18625263

RESUMEN

RGD-peptides can inhibit the binding of ligands to certain beta3 integrins, alphaIIbbeta3 and alphavbeta3, both of which are involved in neointimal hyperplasia that contributes to atherosclerosis and restenosis of arterial walls. Saxatilin, a disintegrin from a Korean snake (Gloydius saxatilis), interacts with integrins alphaIIbbeta3 and alphavbeta3. It suppressed the adhesion of human coronary artery smooth muscle cells (HCASMCs) to vitronectin with an IC(50) of 2.5 microM, and growth factor (PDGF-BB or bFGF)-induced proliferation was inhibited at an IC(50) of 25 microM. Saxatilin disassembled the actin cytoskeleton of focal adhesion and induced cell detachment. This disassembly of focal adhesion in saxatilin-treated HCASMCs involved caspase-induced paxillin degradation. Saxatilin temporally phosphorylated FAK and ERKs and affected the cell cycle of HCASMCs by increasing CDK inhibitors (p21 and p27) and reducing cyclins (D1/2 and E). These results may have significant implications for integrin antagonistic therapy used for the treatment of atherosclerosis and restenosis.


Asunto(s)
Desintegrinas/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Viperidae/fisiología , Animales , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Venenos de Serpiente/química
6.
Cell Transplant ; 24(8): 1571-84, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25008576

RESUMEN

The purpose of this study was to investigate the effects of diabetes on mesenchymal stem cells (MSCs) in terms of their angiogenic and therapeutic potential for repairing tissue ischemia. We culture-isolated MSCs from streptozotocin-induced diabetic rats (D-MSCs) and compared their proliferation, differentiation, and angiogenic effects with those from normal rats (N-MSCs). The angiogenic effects of MSCs were evaluated by real-time PCR, in vitro tube formation assay, and transplantation of the MSCs into a hindlimb ischemia model followed by laser Doppler perfusion imaging. The number of MSCs derived from diabetic rats was smaller, and their proliferation rate was slower than N-MSCs. Upon induction of differentiation, the osteogenic and angiogenic differentiation of D-MSCs were aberrant compared to N-MSCs. The expression of angiogenic factors was lower in D-MSCs than N-MSCs. D-MSCs cocultured with endothelial cells resulted in decreased tube formation compared to N-MSCs. D-MSCs were ineffective to improve hindlimb ischemia and showed lower capillary density and angiogenic gene expression in ischemic limbs than N-MSCs. D-MSCs have defective proliferation and angiogenic activities and are ineffective for repairing hindlimb ischemia. Newer measures are needed before MSCs can be employed as a source for autologous cell therapy.


Asunto(s)
Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Animales , Vasos Sanguíneos/fisiología , Capilares/fisiopatología , Diferenciación Celular , Proliferación Celular , Técnicas de Cocultivo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Células Endoteliales/citología , Miembro Posterior/irrigación sanguínea , Miembro Posterior/metabolismo , Humanos , Isquemia/patología , Masculino , Células Madre Mesenquimatosas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
7.
Insect Biochem Mol Biol ; 34(3): 239-50, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14871620

RESUMEN

A novel serine protease, named as scolonase, was purified and characterized from the tissue of the Korean centipede, Scolopendra subspinipes mutilans. Purified scolonase showed an apparent molecular weight of 25 kDa on sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis and an isoelectric point of 4.8 on isoelectric focusing gel. Scolonase was able to preferentially hydrolyze arginine over lysine at the cleavage site among the several synthetic peptide substrates. Scolonase has also a potent fibrinolytic activity by converting human Glu-plasminogen to activated plasmin due to the specific cleavage of the molecule at the peptide bond Arg(561)-Val(562). The enzyme activity of scolonase was completely inhibited by phenylmethanesulfonyl fluoride and difluorophosphate. The cDNA encoding scolonase was cloned from the cDNA library of the centipede constructed with oligonucleotide probe, which was designed on the basis of the N-terminal amino acid sequence of scolonase. The deduced complete amino acid sequence of scolonase demonstrated that the protein is composed of 277 amino acids including 33 amino acids as a leader sequence, and that it has significant sequence homology with other serine proteases.


Asunto(s)
Artrópodos/enzimología , Artrópodos/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , ADN Complementario/genética , Fibrinólisis , Humanos , Datos de Secuencia Molecular , Peso Molecular , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Homología de Secuencia de Aminoácido , Serina Endopeptidasas/metabolismo , Especificidad por Sustrato , Activador de Tejido Plasminógeno/metabolismo
8.
Toxicon ; 40(7): 947-57, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12076649

RESUMEN

We report upon the isolation, characterization, and cDNA cloning of an anticoagulant protein, halyxin from Agkistrodon halys brevicaudus venom. The protein exists as a 29kDa protein, and is separated into three chains on SDS-PAGE under reducing conditions. However, we cloned only two cDNAs encoding halyxin from the cDNA library of the snake venom gland, on the basis of the determined amino acid sequences. The complete amino acid sequences were deduced from their nucleotide sequences and named halyxin A (129 amino acid residues) and B chain (123 amino acid residues). The deduced amino acid sequence of halyxin A chain corresponds to the two smaller chains. Thus, it is considered that halyxin A chain could be synthesized as a single-chain protein that is subsequently cleaved to yield the mature two-chain protein. The amino acid sequence of halyxin is similar to that of other snake venom proteins of the C-type lectin superfamily, and prolongs plasma-clotting time. In the presence of Ca(2+) ions, halyxin binds to coagulation factors IX, X, IXa, and Xa, but not to other vitamin K-dependent coagulation factors. It also inhibits factor Xa in a non-competitive manner but does not affect other activated coagulation factors.


Asunto(s)
Agkistrodon , Anticoagulantes/aislamiento & purificación , Venenos de Crotálidos/genética , Venenos de Crotálidos/aislamiento & purificación , ADN Complementario/aislamiento & purificación , Endopeptidasas/aislamiento & purificación , Proteínas de Insectos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Factores de Coagulación Sanguínea/metabolismo , Clonación Molecular , Endopeptidasas/genética , Glándulas Exocrinas/metabolismo , Humanos , Datos de Secuencia Molecular , Proteínas y Péptidos Salivales/antagonistas & inhibidores , Homología de Secuencia de Aminoácido
9.
J Pharm Pharmacol ; 55(11): 1577-82, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14713370

RESUMEN

We have investigated the inhibitory effect of salmosin on integrin-mediated human tumour cell proliferation. SK-Mel-2 human melanoma cell adhesion to denatured collagen or vitronectin was found to be significantly and statistically inhibited by salmosin in a dose-dependent manner (P<0.05). Moreover, the binding of SK-Mel-2 cells to salmosin-coated plates was specifically disrupted by anti-integrin alphav monoclonal antibody at 8 microg mL(-1), but not by anti-integrin monoclonal antibody. These findings indicated that salmosin inhibited the adhesion of SK-Mel-2 cells to denatured collagen by specifically blocking integrin alphav. The proliferation of SK-Mel-2 cells on a denatured collagen-coated plate was statistically and significantly inhibited by salmosin induced apoptosis in a dose-dependent manner (P<0.05). Anti-integrin alphav monoclonal antibody, anti-integrin alphavbeta3 monoclonal antibody, and synthetic RGD peptide also suppressed SK-Mel-2 cell proliferation. Several lines of experimental evidence strongly suggested that the inhibition of SK-Mel-2 cell proliferation by salmosin was due to the induction of apoptosis via the blocking of integrin alphav-mediated cell survival.


Asunto(s)
Venenos de Crotálidos/farmacología , Integrina alfaV/efectos de los fármacos , Melanoma/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Humanos , Melanoma/metabolismo , Células Tumorales Cultivadas
10.
Biomaterials ; 34(17): 4235-41, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23489923

RESUMEN

Since the successful generation of induced pluripotent stem cells (iPSC) from adult somatic cells using integrating-viral methods, various methods have been tried for iPSC generation using non-viral and non-integrating technique for clinical applications. Recently, various non-viral approaches such as protein, mRNA, microRNA, and small molecule transduction were developed to avoid genomic integration and generate stem cell-like cells from mouse and human fibroblasts. Despite these successes, there has been no successful generation of iPSC from bone marrow (BM)-derived hematopoietic cells derived using non-viral methods to date. Previous reports demonstrate the ability of polymeric micro and nanoparticles made from polyketals to deliver various molecules to macrophages. MicroRNA-loaded nanoparticles were created using the polyketal polymer PK3 (PK3-miR) and delivered to somatic cells for 6 days, resulting in the formation of colonies. Isolated cells from these colonies were assayed and substantial induction of the pluripotency markers Oct4, Sox2, and Nanog were detected. Moreover, colonies transferred to feeder layers also stained positive for pluripotency markers including SSEA-1. Here, we demonstrate successful activation of pluripotency-associated genes in mouse BM-mononuclear cells using embryonic stem cell (ESC)-specific microRNAs encapsulated in the acid sensitive polyketal PK3. These reprogramming results demonstrate that a polyketal-microRNA delivery vehicle can be used to generate various reprogrammed cells without permanent genetic manipulation in an efficient manner.


Asunto(s)
Acetales/farmacología , Células de la Médula Ósea/citología , Técnicas de Transferencia de Gen , Células Madre Pluripotentes Inducidas/citología , Leucocitos Mononucleares/citología , MicroARNs/metabolismo , Nanopartículas/química , Polímeros/farmacología , Acetales/química , Adulto , Animales , Forma de la Célula/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/metabolismo , Cinética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Antígeno Lewis X/metabolismo , Ratones , Nanopartículas/ultraestructura , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Polímeros/química
11.
Tissue Eng Part C Methods ; 19(5): 375-85, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23126402

RESUMEN

Endothelial progenitor cell (EPC)-capturing techniques have led to revolutionary strategies that can improve the performance of cardiovascular implant devices and engineered tissues by enhancing re-endothelialization and angiogenesis. However, these strategies are limited by controversies regarding the phenotypic identities of EPCs as well as their inability to target and prevent the other afflictions associated with current therapies, namely, thrombosis and neointimal hyperplasia. Therefore, the goal of this study was to study the efficacy of a bioinspired multifunctional nanomatrix in recruiting and promoting the differentiation of EPCs toward an endothelial lineage. The bioinspired nanomatrix combines multiple components, including self-assembled peptide amphiphiles (PAs) that include cell adhesive ligands, nitric oxide (NO)-producing donors, and enzyme-mediated degradable sequences to achieve an endothelium-mimicking character. In this study, human peripheral blood mononuclear cells (PBMNCs) were isolated and cultured on the bioinspired multifunctional nanomatrix. Initial cell adhesion, lectin staining, acetylated low-density lipoprotein uptake, and expression of endothelial markers, including CD31, CD34, von Willebrand Factor, and VEGFR2, were analyzed. The results from this study indicate that the NO releasing bioinspired multifunctional nanomatrix promotes initial adhesion of EPCs when compared to control surfaces. The expression of endothelial markers is also increased on the bioinspired multifunctional nanomatrix, suggesting that it directs the differentiation of EPCs toward an endothelial phenotype. The bioinspired nanomatrix therefore provides a novel biomaterial-based platform for capturing as well as directing EPC behavior. Therefore, this study has the potential to positively impact the patency of cardiovascular devices such as stents and vascular grafts as well as enhanced angiogenesis for ischemic or engineered tissues.


Asunto(s)
Materiales Biocompatibles/farmacología , Diferenciación Celular/efectos de los fármacos , Células Endoteliales/citología , Nanopartículas/química , Células Madre/citología , Andamios del Tejido/química , Secuencia de Aminoácidos , Biomarcadores/metabolismo , Adhesión Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Citometría de Flujo , Humanos , Ligandos , Datos de Secuencia Molecular , Óxido Nítrico/metabolismo , Péptidos/química , Coloración y Etiquetado , Células Madre/efectos de los fármacos , Células Madre/metabolismo
12.
Prog Mol Biol Transl Sci ; 111: 1-26, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22917224

RESUMEN

The technology for generation of induced pluripotent stem cell (iPSC) from somatic cells emerged to circumvent the ethical and immunological limitations of embryonic stem cell (ESC). The recent progress of iPSC technology offers an unprecedented tool for regenerative medicine; however, integrating viral-driven iPSCs prohibits clinical applications by their genetic alterations and tumorigenicity. Various approaches including nonintegrating, nonviral, and nongenetic methods have been developed for generating clinically compatible iPSCs. In addition, approaches for using more clinically convenient or compatible source cells replacing fibroblasts have been actively pursued. While iPSC and ESC closely resemble in genomic, cell biologic, and phenotypic characteristics, these two pluripotent stem cells are not identical in terms of differentiation capacity and epigenetic features. In this chapter, we deal with the current techniques of generating iPSCs and their various characteristics.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Reprogramación Celular/genética , Epigénesis Genética , Técnicas de Transferencia de Gen , Humanos , Modelos Biológicos , Factores de Transcripción/metabolismo
13.
Acta Biomater ; 7(1): 225-33, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20728588

RESUMEN

Current cardiovascular therapies are limited by the loss of endothelium, restenosis and thrombosis. The goal of this study was to develop a biomimetic hybrid nanomatrix that combined the unique properties of electrospun polycaprolactone (ePCL) nanofibers with self-assembled peptide amphiphiles (PAs). ePCL nanofibers have interconnected nanoporous structures, but are hampered by a lack of surface bioactivity to control cellular behavior. It has been hypothesized that PAs could self-assemble onto the surface of ePCL nanofibers and endow them with the characteristic properties of native endothelium. The PAs, which comprised hydrophobic alkyl tails attached to functional hydrophilic peptide sequences, contained enzyme-mediated degradable sites coupled to either endothelial cell-adhesive ligands (YIGSR) or polylysine (KKKKK) nitric oxide (NO) donors. Two different PAs (PA-YIGSR and PA-KKKKK) were successfully synthesized and mixed in a 90:10 (YK) ratio to obtain PA-YK. PA-YK was reacted with pure NO to develop PA-YK-NO, which was then self-assembled onto ePCL nanofibers to generate a hybrid nanomatrix, ePCL-PA-YK-NO. Uniform coating of self-assembled PA nanofibers on ePCL was confirmed by transmission electron microscopy. Successful NO release from ePCL-PA-YK-NO was observed. ePCL-YK and ePCL-PA-YK-NO showed significantly increased adhesion of human umbilical vein endothelial cells (HUVECs). ePCL-PA-YK-NO also showed significantly increased proliferation of HUVECs and reduced smooth muscle cell proliferation. ePCL-PA-YK-NO also displayed significantly reduced platelet adhesion compared with ePCL, ePCL-PA-YK and a collagen control. These results indicate that this hybrid nanomatrix has great potential application in cardiovascular implants.


Asunto(s)
Materiales Biocompatibles/farmacología , Prótesis Vascular , Nanopartículas/química , Péptidos/farmacología , Poliésteres/farmacología , Tensoactivos/farmacología , Ingeniería de Tejidos/métodos , Secuencia de Aminoácidos , Adhesión Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Datos de Secuencia Molecular , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Nanopartículas/ultraestructura , Óxido Nítrico/metabolismo , Péptidos/química , Adhesividad Plaquetaria/efectos de los fármacos , Venas Umbilicales/citología
14.
Biochem Biophys Res Commun ; 293(1): 530-6, 2002 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-12054633

RESUMEN

Saxatilin is a 7.7 kDa disintegrin that belongs to a family of homologous protein found in several snake venoms. Six disulfide bond locations of the disintegrin were determined by enzymatic cleavage and matrix-assisted-laser-desorption-ionization time-of-flight mass spectrometry (MALDI-TOF). Functional implications of the disulfide bonds related to the biological activity of saxatilin were investigated with recombinant protein species produced by site-directed mutagenesis of saxatilin. Several lines of experimental evidence indicated that three disulfide bonds, Cys21-Cys35, Cys29-Cys59, and Cys47-Cys67, of the disintegrin are closely associated with its biological function such as its ability to block the binding of integrin GPIIb-IIIa and alpha(v)beta(3) with fibrinogen and extracellular matrix. Those disulfide linkages were also revealed to be important for maintaining the functional structure of the protein molecule. On the other hand, the disulfide bridges of Cys6-Cys15 and Cys8-Cys16 do not appear to be critical for the molecular structure and function of saxatilin.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Desintegrinas/química , Desintegrinas/farmacología , Disulfuros/química , Secuencia de Aminoácidos , Animales , Células Cultivadas , Dicroismo Circular , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Humanos , Datos de Secuencia Molecular , Peso Molecular , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/química , Conformación Proteica , Proteínas Recombinantes/farmacología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Venenos de Serpiente , Venas Umbilicales
15.
Protein Expr Purif ; 36(1): 1-10, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15177278

RESUMEN

Angiogenesis, the formation of new capillaries from preexisting blood vessels, is involved in many pathological conditions, for example, tumorigenesis, diabetic retinopathy, and rheumatoid arthritis. Angiostatin, which contains the kringle 1-4 domains of plasminogen, is known to be a potent inhibitor of angiogenesis and a strong suppressor of various solid tumors. In this study, we expressed recombinant protein containing the kringle 1-3 domains of human plasminogen in Escherichia coli and investigated its biological activities. The protein was successfully refolded from inclusion bodies and purified at a 30% overall yield, as a single peak by HPLC. The purified recombinant protein had biochemical properties that were similar to those of the native form, which included molecular size, lysine-binding capacity, and immunoreactivity with a specific antibody. The recombinant protein was also found to strongly inhibit the proliferation of bovine capillary endothelial cells in vitro, and the formation of new capillaries on chick embryos. In addition, it suppressed the growth of primary Lewis lung carcinoma and B16 melanoma in an in vivo mouse model. Our findings suggest that the recombinant kringle 1-3 domains in a prokaryote expression system have anti-angiogenic activities, which may be useful in clinical and basic research in the field of angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Escherichia coli/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Plasminógeno/química , Plasminógeno/farmacología , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/aislamiento & purificación , Angiostatinas/farmacología , Animales , Antineoplásicos/aislamiento & purificación , Carcinoma Pulmonar de Lewis/metabolismo , Bovinos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Peso Molecular , Fragmentos de Péptidos/genética , Plasminógeno/genética , Pliegue de Proteína , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA