Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Med ; 26(1): 15, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-32005100

RESUMEN

BACKGROUND: Sca-1+ cardiac stem cells and their limited proliferative potential were major limiting factors for use in various studies. METHODS: Therefore, the effects of sphere genetically engineered cardiac stem cells (S-GECS) inserted with telomerase reverse transcriptase (TERT) were investigated to examine cardiomyocyte survival under hypoxic conditions. GECS was obtained from hTERT-immortalized Sca-1+ cardiac stem cell (CSC) lines, and S-GECS were generated using poly-HEMA. RESULTS: The optimal conditions for S-GECS was determined to be 1052 GECS cells/mm2 and a 48 h culture period to produce spheroids. Compared to adherent-GECS (A-GECS) and S-GECS showed significantly higher mRNA expression of SDF-1α and CXCR4. S-GECS conditioned medium (CM) significantly reduced the proportion of early and late apoptotic cardiomyoblasts during CoCl2-induced hypoxic injury; however, gene silencing via CXCR4 siRNA deteriorated the protective effects of S-GECS against hypoxic injury. As downstream pathways of SDF-1α/CXCR4, the Erk and Akt signaling pathways were stimulated in the presence of S-GECS CM. S-GECS transplantation into a rat acute myocardial infarction model improved cardiac function and reduced the fibrotic area. These cardioprotective effects were confirmed to be related with the SDF-1α/CXCR4 pathway. CONCLUSIONS: Our findings suggest that paracrine factors secreted from transplanted cells may protect host cardiomyoblasts in the infarcted myocardium, contributing to beneficial left ventricle (LV) remodeling after acute myocardial infarction (AMI).


Asunto(s)
Ataxina-1/metabolismo , Miocitos Cardíacos/citología , Esferoides Celulares/citología , Células Madre/citología , Telomerasa/genética , Animales , Ataxina-1/genética , Adhesión Celular , Técnicas de Cultivo de Célula , Hipoxia de la Célula , Línea Celular , Proliferación Celular , Supervivencia Celular , Quimiocina CXCL12/genética , Cobalto/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Ingeniería Genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Comunicación Paracrina , Regiones Promotoras Genéticas , Ratas , Receptores CXCR4/genética , Esferoides Celulares/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo
2.
Soft Matter ; 16(2): 428-434, 2020 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-31799582

RESUMEN

Maturation and synchronisation of heart cells, including cardiomyocytes and fibroblasts, are essential to develop functional biomimetic cardiac tissues for regenerative medicine and drug discovery. Synchronisation of cells in the biomimetic cardiac tissue requires the structural integrity and functional maturation of cardiomyocytes with other cell types. However, it is challenging to synchronise the beating of macroscale cardiac tissues and induce maturation of cardiomyocytes derived from stem cells. Here, we developed a simple assembly technology to modulate cell-cell interactions by combining layer-by-layer (LBL) deposition and centrifugation of cells with collagen type I to control cell-cell interactions for the preparation of cardiac macro tissues (CMTs). We found that maturation of cardiomyocytes in CMTs was largely enhanced by growth factors FGF-4 and ascorbic acid, but synchronisation of cardiac beating required LBL deposition of cardiomyocytes and cardiac fibroblasts in addition to the growth factors during the maturation process. Our findings have important implications because incorporation of cardiac fibroblasts into the cardiomyocyte layer is a prerequisite for synchronised beating of macroscale cardiac tissues in addition to growth factors to facilitate maturation of stem cell-derived cardiomyocytes.


Asunto(s)
Fibroblastos/citología , Miocitos Cardíacos/citología , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Biomimética , Bioimpresión , Comunicación Celular , Colágeno Tipo I/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Factor 4 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/metabolismo , Humanos , Miocitos Cardíacos/metabolismo
3.
Int J Mol Sci ; 21(6)2020 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-32245208

RESUMEN

Thymosin ß4 (Tß4) is a G-actin sequestering protein that contributes to diverse cellular activities, such as migration and angiogenesis. In this study, the beneficial effects of combined cell therapy with Tß4 and human adipose-derived stem cells (hASCs) in a mouse ischemic hindlimb model were investigated. We observed that exogenous treatment with Tß4 enhanced endogenous TMSB4X mRNA expression and promoted morphological changes (increased cell length) in hASCs. Interestingly, Tß4 induced the active state of hASCs by up-regulating intracellular signaling pathways including the PI3K/AKT/mTOR and MAPK/ERK pathways. Treatment with Tß4 significantly increased cell migration and sprouting from microbeads. Moreover, additional treatment with Tß4 promoted the endothelial differentiation potential of hASCs by up-regulating various angiogenic genes. To evaluate the in vivo effects of the Tß4-hASCs combination on vessel recruitment, dorsal window chambers were transplanted, and the co-treated mice were found to have a significantly increased number of microvessel branches. Transplantation of hASCs in combination with Tß4 was found to improve blood flow and attenuate limb or foot loss post-ischemia compared to transplantation with hASCs alone. Taken together, the therapeutic application of hASCs combined with Tß4 could be effective in enhancing endothelial differentiation and vascularization for treating hindlimb ischemia.


Asunto(s)
Miembro Posterior/metabolismo , Isquemia/metabolismo , Células Madre Mesenquimatosas/metabolismo , Timosina/metabolismo , Timosina/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Trasplante de Células , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/genética , Isquemia/terapia , Sistema de Señalización de MAP Quinasas/genética , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Desnudos , Neovascularización Fisiológica/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Timosina/genética , Timosina/uso terapéutico , Cicatrización de Heridas/genética
4.
Mol Med ; 25(1): 33, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31307370

RESUMEN

BACKGROUND: The beneficial effects of angiotensin II type 1 receptor blockers (ARBs) on atherosclerosis have been demonstrated in numerous studies. We investigated the effects of fimasartan on reducing neointimal formation and systemic inflammation after carotid artery (CA) injury in Apolipoprotein E knockout (ApoE KO) mice. METHODS: ApoE KO mice were randomly allocated to Group I (without CA injury), Group II (without CA injury + Fimasartan), Group III (CA injury), and Group IV (CA injury + Fimasartan). Fimasartan was orally administered everyday starting 3 days before iatrogenic left CA injury. RESULTS: At 28 days, neointimal hyperplasia and the inflammatory cytokines including TNFα, IL-6, ICAM, and MMP-9 in the peripheral blood were significantly reduced in Groups II and IV compared to Groups I and III, respectively. All fimasartan-administered groups revealed significant increases of CD4+CD25+Foxp3+ regulatory T (Treg) cells with increased plasma levels of IL-10 and TGFß. In addition, increased CD8+ T cells by fimasartan were correlated with reduced smooth muscle cell (SMC) proliferation in the neointima in Groups II and IV. Furthermore, the populations of Treg and CD8+ T cells in total splenocytes were increased in Groups II and IV compared to Groups I and III, respectively. The enlargement of spleens due to CA injury in the Group III was attenuated by fimasartan, as shown in the Group IV. These data indicate that fimasartan significantly reduced SMC proliferation in neointima and increased Treg cells in ApoE KO CA injury mice. CONCLUSIONS: This study suggests fimasartan could be an efficient strategy for reduction of atherosclerotic progression, with a decrease in immune response and systemic inflammation.


Asunto(s)
Compuestos de Bifenilo/farmacocinética , Compuestos de Bifenilo/uso terapéutico , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Inflamación/sangre , Inflamación/tratamiento farmacológico , Neointima/sangre , Neointima/tratamiento farmacológico , Pirimidinas/farmacocinética , Pirimidinas/uso terapéutico , Tetrazoles/farmacocinética , Tetrazoles/uso terapéutico , Antagonistas de Receptores de Angiotensina/uso terapéutico , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Interleucina-6/sangre , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Ratones , Ratones Noqueados , Linfocitos T Reguladores/efectos de los fármacos , Factor de Necrosis Tumoral alfa/sangre
5.
Int J Mol Sci ; 18(1)2017 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-28117756

RESUMEN

Talin is a focal adhesion protein that activates integrins and recruits other focal adhesion proteins. Talin regulates the interactions between integrins and the extracellular matrix, which are critical for endothelial cells during angiogenesis. In this study, we successfully synthesized a novel talin modulator, N-((2-(1H-indol-3-yl)ethyl)carbamoyl)-2-(benzo[d][1,3]dioxol-5-yloxy)acetamide, referred to as KCH-1521. KCH-1521 was determined to bind talin and modulate downstream signaling molecules of talin. After 24 h of treatment, KCH-1521 changed the cell morphology of human umbilical vein endothelial cells (HUVECs) and reduced focal adhesion protein expression including vinculin and paxillin. Talin downstream signaling is regulated via focal adhesion kinase (FAK), kinase B (AKT), and extracellular signal-regulated kinase (ERK) pathways, however, treatment with KCH-1521 decreased phosphorylation of FAK, AKT, and ERK, leading to reduction of cell proliferation, survival, and angiogenesis. Interestingly, the expression of various angiogenic genes was significantly decreased after treatment with KCH-1521. Also, in vitro tube forming assay revealed that KCH-1521 reduced angiogenic networks in a time-dependent manner. To investigate the reversibility of its effects, KCH-1521 was removed after treatment. HUVECs recovered their morphology through rearrangement of the cytoskeleton and the expression of angiogenic genes was also recovered. By further optimization and in vivo studies of KCH-1521, a novel drug of talin modulation could be used to achieve therapeutic anti-angiogenesis for vascular diseases and cancers.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Talina/metabolismo , Urea/farmacología , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos , Urea/química
6.
Biol Chem ; 397(3): 249-55, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26733157

RESUMEN

Key regulatory genes in pluripotent stem cells are of interest not only as reprogramming factors but also as regulators driving tumorigenesis. Nanog is a transcription factor involved in the maintenance of embryonic stem cells and is one of the reprogramming factors along with Oct4, Sox2, and Lin28. Nanog expression has been detected in different types of tumors, and its expression is a poor prognosis for cancer patients. However, there is no clear evidence that Nanog is functionally involved in tumorigenesis. In this study, we induced overexpression of Nanog in mouse embryonic fibroblast cells and subsequently assessed their morphological changes, proliferation rate, and tumor formation ability. We found that Nanog overexpression induced immortalization of mouse embryonic fibroblast cells (MEFs) and increased their proliferation rate in vitro. We also found that formation of tumors after subcutaneous injection of retroviral-Nanog infected MEFs (N-MEFs) into athymic mouse. Cancer-related genes such as Bmi1 were expressed at high levels in N-MEFs. Hence, our results demonstrate that Nanog is able to transform normal somatic cells into tumor cells.


Asunto(s)
Carcinogénesis/genética , Transformación Celular Neoplásica/genética , Fibroblastos/patología , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Regulación hacia Arriba , Animales , Carcinogénesis/patología , Transformación Celular Neoplásica/patología , Células Cultivadas , Fibroblastos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Proteína Homeótica Nanog
7.
Int J Mol Sci ; 17(6)2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27231894

RESUMEN

Cardiac stem cells (CSCs) were known to secrete diverse paracrine factors leading to functional improvement and beneficial left ventricular remodeling via activation of the endogenous pro-survival signaling pathway. However, little is known about the paracrine factors secreted by CSCs and their roles in cardiomyocyte survival during hypoxic condition mimicking the post-myocardial infarction environment. We established Sca-1+/CD31- human telomerase reverse transcriptase-immortalized CSCs (Sca-1+/CD31- CSCs(hTERT)), evaluated their stem cell properties, and paracrine potential in cardiomyocyte survival during hypoxia-induced injury. Sca-1+/CD31- CSCs(hTERT) sustained proliferation ability even after long-term culture exceeding 100 population doublings, and represented multi-differentiation potential into cardiomyogenic, endothelial, adipogenic, and osteogenic lineages. Dominant factors secreted from Sca-1+/CD31- CSCs(hTERT) were EGF, TGF-ß1, IGF-1, IGF-2, MCP-1, HGF R, and IL-6. Among these, MCP-1 was the most predominant factor in Sca-1+/CD31- CSCs(hTERT) conditioned medium (CM). Sca-1+/CD31- CSCs(hTERT) CM increased survival and reduced apoptosis of HL-1 cardiomyocytes during hypoxic injury. MCP-1 silencing in Sca-1+/CD31- CSCs(hTERT) CM resulted in a significant reduction in cardiomyocyte apoptosis. We demonstrated that Sca-1+/CD31- CSCs(hTERT) exhibited long-term proliferation capacity and multi-differentiation potential. Sca-1+/CD31- CSCs(hTERT) CM protected cardiomyocytes from hypoxic injury partly via MCP-1-dependent mechanism. Thus, they are valuable sources for in vitro and in vivo studies in the cardiovascular field.


Asunto(s)
Quimiocina CCL2/metabolismo , Miocitos Cardíacos/citología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Células Madre/metabolismo , Telomerasa/genética , Animales , Diferenciación Celular , Hipoxia de la Célula , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Humanos , Ratones , Modelos Biológicos , Células Madre/citología
8.
J Cell Physiol ; 230(8): 1807-21, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25521758

RESUMEN

Dimethyl sulfoxide (DMSO) is widely used to induce multilineage differentiation of embryonic and adult progenitor cells. To date, little is known about the mechanisms underlying DMSO-induced mesodermal specification. In this study, we investigated the signaling pathways and lineage-determining genes involved in DMSO-induced mesodermal specification in P19 cells. Wnt/ß-catenin and TGF-ß superfamily signaling pathways such as BMP, TGF-ß and GDF1 signaling were significantly activated during DMSO-induced mesodermal specification. In contrast, Nodal/Cripto signaling pathway molecules, required for endoderm specification, were severely downregulated. DMSO significantly upregulated the expression of cardiac mesoderm markers but inhibited the expression of endodermal and hematopoietic lineage markers. Among the DMSO-activated cell lineage markers, the expression of Mixl1 and Flk1 was dramatically upregulated at both the transcript and protein levels, and the populations of Mixl1+, Flk1+ and Mixl1+/Flk1+ cells also increased significantly. DMSO modulated cell cycle molecules and induced cell apoptosis, resulting in significant cell death during EB formation of P19 cells. An inhibitor of Flk1, SU5416 significantly blocked expressions of TGF-ß superfamily members, mesodermal cell lineage markers and cell cycle molecules but it did not affect Wnt molecules. These results demonstrate that Mixl1 and Flk1 play roles as key downstream or interacting effectors of Wnt/TGF-ß signaling pathway during DMSO-induced mesodermal specification in P19 cells.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Mesodermo/citología , Factor de Crecimiento Transformador beta/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Wnt/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular Tumoral , Dimetilsulfóxido/farmacología , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Células Madre Embrionarias/citología , Citometría de Flujo , Humanos , Inmunohistoquímica , Mesodermo/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Biochem Biophys Res Commun ; 468(1-2): 372-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26498525

RESUMEN

A number of researchers have been reporting a wide range of in vitro and in vivo studies of cell engraftment to enhance angiogenesis using stem cells. Despite these efforts, studies involving three-dimensional (3D) culture method that mimics in vivo environment have not reached its peak yet. In this study, we investigated the change and effects on cellular angiogenic growth factors through sphere formation of adipose stem cell (ASC) which is engineered by poly-2-hydroxyethyl methacrylate (Poly-HEMA). First of all, we successfully induced sphere formation of ASC (sph-ASC) on Poly-HEMA coated plates. sph-ASC represented significantly higher expression levels of anti-apoptotic and hypoxic factors compared to monolayer adherent ASC (adh-ASC). Interestingly, sph-ASC showed higher mRNA levels of the following genes; CD31, CD144, vWF, IGF-2, MCP-1, PDGF-A, VEGF-A, VEGF-C, and FGF-2. In addition, mRNA expressions of angiogenic growth factor receptors such as Flk1, FGFR1, FGFR2, and Tie2 were elevated in sph-ASC. In protein level, Cytokine/Chemokines antibody array revealed a significant increase of FGF-2 in sph-ASC (3.17-fold) compared to adh-ASC. To investigate the effects of FGF-2 on sph-ASC, Matrigel angiogenic invasion assay showed significant reduced level of FGF-2 in FGF-2 siRNA transfected sph-ASC (2.27-fold) compared to negative control siRNA transfected sph-ASC. These findings suggest that Poly-HEMA coated plates induce sphere formation of ASC which has significantly higher expression of FGF-2, and plays a critical role as a major regulating growth factor of in vitro angiogenesis.


Asunto(s)
Tejido Adiposo/citología , Materiales Biocompatibles Revestidos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neovascularización Fisiológica , Polihidroxietil Metacrilato/metabolismo , Esferoides Celulares/citología , Células Madre/citología , Animales , Movimiento Celular , Células Cultivadas , Humanos , Ratones Endogámicos C57BL , Esferoides Celulares/metabolismo , Células Madre/metabolismo , Ingeniería de Tejidos
10.
Stem Cells ; 32(4): 998-1011, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24715692

RESUMEN

Redox balance has been suggested as an important determinant of "stemness" in embryonic stem cells (ESCs). In this study, we demonstrate that peroxiredoxin (Prx) plays a pivotal role in maintenance of ESC stemness during neurogenesis through suppression of reactive oxygen species (ROS)-sensitive signaling. During neurogenesis, Prx I and Oct4 are expressed in a mutually dependent manner and their expression is abruptly downregulated by an excess of ROS. Thus, in Prx I(-/-) or Prx II(-/-) ESCs, rapid loss of stemness can occur due to spontaneous ROS overload, leading to their active commitment into neurons; however, stemness is restored by the addition of an antioxidant or an inhibitor of c-Jun N-terminal kinase (JNK). In addition, Prx I and Prx II appear to have a tight association with the mechanism underlying the protection of ESC stemness in developing teratomas. These results suggest that Prx functions as a protector of ESC stemness by opposing ROS/JNK cascades during neurogenesis. Therefore, our findings have important implications for understanding of maintenance of ESC stemness through involvement of antioxidant enzymes and may lead to development of an alternative stem cell-based therapeutic strategy for production of high-quality neurons in large quantity.


Asunto(s)
Células Madre Embrionarias/enzimología , MAP Quinasa Quinasa 4/metabolismo , Neurogénesis/fisiología , Peroxirredoxinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Células Madre Embrionarias/citología , MAP Quinasa Quinasa 4/genética , Ratones , Ratones Noqueados , Peroxirredoxinas/genética
11.
Circ J ; 79(4): 880-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25737017

RESUMEN

BACKGROUND: Aberrant expression of microRNAs is associated with neointimal hyperplasia (NIH) in type 2 diabetes. We prospectively compared the effects of pioglitazone on coronary NIH and changes in microRNAs according to NIH status in type 2 diabetic patients during 9-month follow-up. METHODS AND RESULTS: Type 2 diabetic patients were randomly assigned to the pioglitazone (n=36) or control groups (n=36) after coronary stenting. Primary endpoint was the comparison of changes in neointimal volume on OCT and in the level of circulating microRNA-17,-24,-92a,-126 and -145 during 9-month follow-up. Secondary endpoint was the comparison of changes in brachial artery flow-mediated dilation and inflammatory markers such as IL-6, TNF-α, hsCRP, adiponectin, sICAM-1, and sVCAM-1 between the 2 groups. Neointimal volume was significantly lower in the pioglitazone group (25.02±17.78 mm(3)vs. 55.10±30.01 mm(3), P<0.001) with significant increases in circulating microRNA-24 (0.264±0.084 vs. 0.006±0.030, P<0.001) during follow-up. FMD was significantly greater in the pioglitazone than control group at 9 months (0.47±0.14 mm vs. 0.28±0.18 mm, P<0.05, respectively). Decreases in inflammatory markers such as IL-6, TNF-α, and sVCAM-1 were significantly greater in the pioglitazone than the control group during the follow-up. CONCLUSIONS: Pioglitazone significantly decreased NIH with increases in circulating microRNA-24 at 9-month follow-up. The decrease in microRNA-24 could be used as a potential predictor of increases in NIH in type 2 diabetic patients.


Asunto(s)
Vasos Coronarios , Diabetes Mellitus Tipo 2 , Angiopatías Diabéticas , Hipoglucemiantes/administración & dosificación , MicroARNs/sangre , Tiazolidinedionas/administración & dosificación , Tomografía de Coherencia Óptica , Anciano , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Angiopatías Diabéticas/sangre , Angiopatías Diabéticas/tratamiento farmacológico , Angiopatías Diabéticas/patología , Femenino , Humanos , Hiperplasia/sangre , Hiperplasia/tratamiento farmacológico , Hiperplasia/patología , Masculino , Persona de Mediana Edad , Neointima/sangre , Neointima/tratamiento farmacológico , Neointima/patología , Pioglitazona
12.
Biochem Biophys Res Commun ; 449(4): 405-11, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24858689

RESUMEN

Proangiogenic cell therapy using autologous progenitors is a promising strategy for treating ischemic disease. Considering that neovascularization is a harmonized cellular process that involves both endothelial cells and vascular smooth muscle cells, peripheral blood-originating endothelial colony-forming cells (ECFCs) and smooth muscle progenitor cells (SMPCs), which are similar to mature endothelial cells and vascular smooth muscle cells, could be attractive cellular candidates to achieve therapeutic neovascularization. We successfully induced populations of two different vascular progenitor cells (ECFCs and SMPCs) from adult peripheral blood. Both progenitor cell types expressed endothelial-specific or smooth muscle-specific genes and markers, respectively. In a protein array focused on angiogenic cytokines, SMPCs demonstrated significantly higher expression of bFGF, EGF, TIMP2, ENA78, and TIMP1 compared to ECFCs. Conditioned medium from SMPCs and co-culture with SMPCs revealed that SMPCs promoted cell proliferation, migration, and the in vitro angiogenesis of ECFCs. Finally, co-transplantation of ECFCs and SMPCs induced robust in vivo neovascularization, as well as improved blood perfusion and tissue repair, in a mouse ischemic hindlimb model. Taken together, we have provided the first evidence of a cell therapy strategy for therapeutic neovascularization using two different types of autologous progenitors (ECFCs and SMPCs) derived from adult peripheral blood.


Asunto(s)
Células Endoteliales/citología , Miocitos del Músculo Liso/citología , Neovascularización Fisiológica , Células Madre/citología , Adulto , Animales , Supervivencia Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/terapia , Ratones , Modelos Animales , Miocitos del Músculo Liso/metabolismo , Células Madre/metabolismo
13.
Cells ; 12(16)2023 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-37626839

RESUMEN

The generation of mature and vascularized human pluripotent stem cell-derived cardiac organoids (hPSC-COs) is necessary to ensure the validity of drug screening and disease modeling. This study investigates the effects of cellular aggregate (CA) stemness and self-organization on the generation of mature and vascularized hPSC-COs and elucidates the mechanisms underlying cardiac organoid (CO) maturation and vascularization. COs derived from 2-day-old CAs with high stemness (H-COs) and COs derived from 5-day-old CAs with low stemness (L-COs) were generated in a self-organized microenvironment via Wnt signaling induction. This study finds that H-COs exhibit ventricular, structural, metabolic, and functional cardiomyocyte maturation and vessel networks consisting of endothelial cells, smooth muscle cells, pericytes, and basement membranes compared to L-COs. Transcriptional profiling shows the upregulation of genes associated with cardiac maturation and vessel formation in H-COs compared with the genes in L-COs. Through experiments with LIMK inhibitors, the activation of ROCK-LIMK-pCofilin via ECM-integrin interactions leads to cardiomyocyte maturation and vessel formation in H-COs. Furthermore, the LIMK/Cofilin signaling pathway induces TGFß/NODAL and PDGF pathway activation for the maturation and vascularization of H-COs. The study demonstrates for the first time that LIMK/Cofilin axis activation plays an important role in the generation of mature and vascularized COs.


Asunto(s)
Células Endoteliales , Organoides , Humanos , Miocitos Cardíacos , Vía de Señalización Wnt , Factores Despolimerizantes de la Actina , Matriz Extracelular , Neovascularización Patológica , Integrinas
14.
J Cell Physiol ; 227(11): 3678-92, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22378194

RESUMEN

To identify potential downstream targets of Nanog, a key transcription factor in the maintenance of pluripotency of embryonic stem (ES) and embryonal carcinoma (EC) cells, global gene expression profiles in Nanog small interfering RNA (siRNA)-transfected P19 EC stem cells were performed using cDNA, 60-mer, and 30-mer microarray platforms. The putative Nanog target genes identified by Nanog silencing were verified using reverse transcription-polymerase chain reaction after Nanog overexpression. Downregulation of Nanog in P19 cells resulted in reduction of pluripotency markers, such as Fgf4, Klf2, Mtf2, Oct-4, Rex1, Sox1, Yes, and Zfp143, whereas overexpression of Nanog in P19 cells reversely upregulated their expression. However, expressions of pluripotency markers Cripto, germ cell nuclear factor, Sox2, and Zfp57 as well as leukemia inhibitory factor (LIF)/Stat3 pathway molecules LIF, IL6st, and Stat3 were not affected after 48 h transfection with Nanog siRNA or construct. Nanog silencing also downregulated expression of molecules involved in the p53- and cell cycle-signaling pathway (Atf3, Jdp2, Cul3, Hist1hic, and Bcl6), whereas expression of E2f1, Tob1, Lyn, and Smarcc1 was upregulated by Nanog silencing. Expressions of cyclins D1, D2, D3, and E1 as well as cyclin-dependent kinase (Cdk) 1 and Cdk6 were downregulated by Nanog silencing in P19 cells, whereas Nanog overexpression reversely increased their expressions. Taken together, examination of global transcriptional changes after Nanog silencing followed by verification by Nanog overexpression has revealed new molecules involved in the maintenance of self-renewal and in the regulation of the p53- and cell cycle-pathway of P19 cells.


Asunto(s)
Proteínas de Ciclo Celular , Células Madre de Carcinoma Embrionario , Células Madre Embrionarias , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Células Madre de Carcinoma Embrionario/citología , Células Madre de Carcinoma Embrionario/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/genética , Proteínas de Homeodominio/genética , Humanos , Proteína Homeótica Nanog , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , ARN Interferente Pequeño , Transducción de Señal , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo
15.
Circ J ; 76(5): 1213-21, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22354195

RESUMEN

BACKGROUND: The correlations between circulating angiogenic cell mobilizations and improvement of microvascular integrity were investigated in patients (n=110) with acute myocardial infarction (AMI) during an 8-month follow up. METHODS AND RESULTS: Coronary flow reserve (CFR) was measured at baseline and at 8 months by using an intracoronary Doppler wire. Serial changes in the absolute numbers of circulating angiogenic cells such as CD34+, CXCR4+, CD117+, CD133+ and C-met+ were measured at baseline, day 1, day 5 and at 8 months. The absolute numbers of circulating angiogenic cells at day 1 were significantly higher than those at baseline. A positive correlation was found between the numbers of circulating angiogenic cells of CD34+, CXCR4+, CD117+ and CD133+ cells at day 1 and the CFR changes from baseline. The cut-off value of CFR changes at 8 months by a receiver operating characteristic curve between a circulating CD34+ cell at day 1 and changes of CFR at 8 months was 0. Late-loss showed the positive correlation with the absolute number of C-met+ cells and the negative correlation with the absolute number of CXCR4+ cells after AMI. The negative correlation was found between changes in high-sensitive C-reactive protein and soluble intercellular adhesion molecule-1 and changes in CFR at 8 months. CONCLUSIONS: The recovery of microvascular integrity after acute ischemic injury was expedited by the increases in circulating angiogenic cell mobilization together with the greater decreases in inflammatory cytokines. The improvement in CFR could be predicted by the measurement of circulating angiogenic cells after AMI.


Asunto(s)
Circulación Coronaria , Movilización de Célula Madre Hematopoyética , Infarto del Miocardio/sangre , Infarto del Miocardio/fisiopatología , Células Madre/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/sangre , Citocinas/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Estudios Prospectivos , Receptores CXCR4/sangre , Células Madre/patología , Factores de Tiempo
16.
Biosens Bioelectron ; 198: 113828, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-34847362

RESUMEN

An exosome species containing CD63 as a marker of melanoma was isolated from bulk exosome population and used as a sample for detecting malignant melanoma. A calcium binding protein (CBP) was produced and then used to raise monoclonal antibody. The antibody was sensitive to a conformational change of CBP caused by Ca2+ binding. Immuno-magnetic beads were prepared by immobilizing the conformation-sensitive binder and subsequent binding of CBP conjugated with the capture antibody specific to CD63. These immuno-beads were used to isolate CD63-positive exosome from a bulk exosome sample (normal or melanoma) based on the 'calcium switch-on/off' mechanism through magnetic separation. After recovery, the subpopulation sample was analyzed by immunoassays for cavelion1 (Cav1), CD81, and CD9 as sub-subpopulation markers. Normalized signals of Cav1 and/or CD81 over CD9 were higher in melanoma samples than in normal samples, depending on clinical stages (I, II, and IV) of patients. This was in contrast to assay results for the bulk exosome population that showed a completely mixed state of melanoma and normal samples. These results showed that an exosome subpopulation sample prepared using a 'Ca2+-dependent switch' technology might be useful for diagnosing malignant melanoma at an early stage to increase 5-year survival rates.


Asunto(s)
Técnicas Biosensibles , Exosomas , Melanoma , Biomarcadores de Tumor , Calcio , Humanos , Melanoma/diagnóstico
17.
Phytother Res ; 25(2): 239-49, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20641058

RESUMEN

The effects of red ginseng extract on circulating angiogenic cell mobilization and improvement of microvascular integrity were evaluated in ST-elevation acute myocardial infarction (AMI) patients during 8-month follow-up. AMI patients (n = 50) were randomly assigned to the red ginseng group (3 g/day, n = 25) or the placebo group (n = 25) after coronary stenting. Coronary flow reserve (CFR) was measured at baseline and at 8 months with an intracoronary Doppler wire. Serial changes in the absolute numbers of circulating angiogenic cells such as CD34(+) , CXCR4(+) , CD117(+) , CD133(+) and C-met(+) were measured at baseline, 1 day, 5 days and at 8 months. CFR were similar between the two groups at baseline, and CFR was significantly higher in the red ginseng group than in the placebo group (2.80 ± 0.91 and 2.56 ± 0.77, p < 0.05, respectively) after 8 months of red ginseng administration. The absolute numbers of circulating CD34(+) , CXCR4(+) and CD117(+) cells were significantly higher in the red ginseng group at 1 and 5 days after stenting. Significant positive correlations were found between the numbers of circulating angiogenic cells at day 1 and the changes from baseline in CFR for CD34(+) , CXCR4(+) , CD117(+) and C-met(+) cells. Red ginseng extract increased CD34(+) , CXCR4(+) and CD117(+) circulating angiogenic cell mobilization and decreased inflammation in AMI patients, thereby improving CFR during the 8-month follow-up.


Asunto(s)
Corazón/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Panax/química , Fitoterapia , Extractos Vegetales/uso terapéutico , Anciano , Velocidad del Flujo Sanguíneo , Ecocardiografía Doppler , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica/efectos de los fármacos , Extractos Vegetales/farmacología , Estudios Prospectivos , Método Simple Ciego
18.
Biofabrication ; 13(4)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34404035

RESUMEN

A novel tissue engineering strategy using 3D bio-print technology has become a promising therapeutic method for acute myocardial infarction (AMI) in an animal model. However, the application of 3D bio-printed tissue remains limited due to poor graft survival. Therefore, it is a scientific priority to enhance graft survival by precisely adjusting the 3D environment of encapsulated cells. In this study, novel transplantable 3D cardiac mesh (cMesh) tissue with a porous mesh structure was presented using human cardiomyocytes, human cardiac fibroblasts, and gelatin-methacryloyl-collagen hydrogel. Cardiomyocytes and cardiac fibroblasts were well spreaded. The cardiomyocytes were connected with a gap junction channel in bio-printed cMesh and a 3D cardiac patch with an aggregated structure. Porous cMesh demonstrated structural advantages by increased phosphorylation of mTOR, AKT, and ERK signals associated with cell survival. Transplanted cMesh in rats with AMI improved long-term graft survival, vessel formation, and stabilization, reduced fibrosis, increased left ventricle thickness, and enhanced cardiac function. Our results suggest that porous cMesh provides structural advantages and a positive therapeutic effect in an AMI animal model.


Asunto(s)
Infarto del Miocardio , Mallas Quirúrgicas , Animales , Gelatina , Hidrogeles , Infarto del Miocardio/terapia , Miocitos Cardíacos , Impresión Tridimensional , Ratas , Ingeniería de Tejidos
19.
Cells ; 10(10)2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34685725

RESUMEN

Mature cardiomyocytes (CMs) obtained from human pluripotent stem cells (hPSCs) have been required for more accurate in vitro modeling of adult-onset cardiac disease and drug discovery. Here, we found that FGF4 and ascorbic acid (AA) induce differentiation of BG01 human embryonic stem cell-cardiogenic mesoderm cells (hESC-CMCs) into mature and ventricular CMs. Co-treatment of BG01 hESC-CMCs with FGF4+AA synergistically induced differentiation into mature and ventricular CMs. FGF4+AA-treated BG01 hESC-CMs robustly released acute myocardial infarction (AMI) biomarkers (cTnI, CK-MB, and myoglobin) into culture medium in response to hypoxic injury. Hypoxia-responsive genes and potential cardiac biomarkers proved in the diagnosis and prognosis of coronary artery diseases were induced in FGF4+AA-treated BG01 hESC-CMs in response to hypoxia based on transcriptome analyses. This study demonstrates that it is feasible to model hypoxic stress in vitro using hESC-CMs matured by soluble factors.


Asunto(s)
Ácido Ascórbico/farmacología , Diferenciación Celular , Factor 4 de Crecimiento de Fibroblastos/farmacología , Células Madre Embrionarias Humanas/patología , Modelos Biológicos , Miocitos Cardíacos/patología , Estrés Fisiológico , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular , Medios de Cultivo/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ventrículos Cardíacos/patología , Células Madre Embrionarias Humanas/efectos de los fármacos , Humanos , Infarto del Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Transcriptoma/genética
20.
Biomaterials ; 278: 121133, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34571434

RESUMEN

The generation of mature ventricular cardiomyocytes (CMs) resembling adult CMs from human pluripotent stem cells (hPSCs) is necessary for disease modeling and drug discovery. To investigate the effect of self-organizing capacity on the generation of mature cardiac organoids (COs), we generated cardiac mesoderm cell-derived COs (CMC-COs) and CM-derived COs (CM-COs) and evaluated COs. CMC-COs exhibited more organized sarcomere structures and mitochondria, well-arranged t-tubule structures, and evenly distributed intercalated discs. Increased expressions of ventricular CM, cardiac metabolic, t-tubule formation, K+ ion channel, and junctional markers were confirmed in CMC-COs. Mature ventricular-like function such as faster motion vector speed, decreased beats per min, increased peak-to-peak duration, and prolonged APD50 and APD90 were observed in CMC-COs. Transcriptional profiling revealed that extracellular matrix-integrin, focal adhesion, and LEFTY-PITX2 signaling pathways are upregulated in CMC-COs. LEFTY knockdown affected ECM-integrin-FA signaling pathways in CMC-COs. Here, we found that high self-organizing capacity of CMCs is critical for the generation of mature and ventricular COs. We also demonstrated that LEFTY-PITX2 signaling plays key roles for CM maturation and specification into ventricular-like CM subtype in CMC-COs. CMC-COs are an attractive resource for disease modeling and drug discovery.


Asunto(s)
Proteínas de Homeodominio , Células Madre Pluripotentes Inducidas , Factores de Determinación Derecha-Izquierda , Miocitos Cardíacos , Células Madre Pluripotentes , Factores de Transcripción , Diferenciación Celular , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Determinación Derecha-Izquierda/metabolismo , Mesodermo , Organoides , Transducción de Señal , Factores de Transcripción/metabolismo , Proteína del Homeodomínio PITX2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA