Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 644
Filtrar
1.
Int J Mol Sci ; 25(11)2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38892272

RESUMEN

Endothelial progenitor cells (EPCs) are circulating cells of various origins that possess the capacity for renewing and regenerating the endothelial lining of blood vessels. During physical activity, in response to factors such as hypoxia, changes in osmotic pressure, and mechanical forces, endothelial cells undergo intense physiological stress that results in endothelial damage. Circulating EPCs participate in blood vessel repair and vascular healing mainly through paracrine signalling. Furthermore, physical activity may play an important role in mobilising this important cell population. In this narrative review, we summarise the current knowledge on the biology of EPCs, including their characteristics, assessment, and mobilisation in response to both chronic and acute physical activity in healthy individuals.


Asunto(s)
Células Progenitoras Endoteliales , Ejercicio Físico , Humanos , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/citología , Ejercicio Físico/fisiología , Animales
2.
J Cell Mol Med ; 28(12): e18489, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38899522

RESUMEN

This study explores the impact of senescence on autocrine C-C motif chemokine ligand 5 (CCL5) in human endothelial progenitor cell (EPCs), addressing the poorly understood decline in number and function of EPCs during ageing. We examined the effects of replication-induced senescence on CCL5/CCL5 receptor (CCR5) signalling and angiogenic activity of EPCs in vitro and in vivo. We also explored microRNAs controlling CCL5 secretion in senescent EPCs, its impact on EPC angiogenic activity, and validated our findings in humans. CCL5 secretion and CCR5 levels in senescent EPCs were reduced, leading to attenuated angiogenic activity. CCL5 enhanced EPC proliferation via the CCR5/AKT/P70S6K axis and increased vascular endothelial growth factor (VEGF) secretion. Up-regulation of miR-409 in senescent EPCs resulted in decreased CCL5 secretion, inhibiting the angiogenic activity, though these negative effects were counteracted by the addition of CCL5 and VEGF. In a mouse hind limb ischemia model, CCL5 improved the angiogenic activity of senescent EPCs. Analysis involving 62 healthy donors revealed a negative association between CCL5 levels, age and Framingham Risk Score. These findings propose CCL5 as a potential biomarker for detection of EPC senescence and cardiovascular risk assessment, suggesting its therapeutic potential for age-related cardiovascular disorders.


Asunto(s)
Senescencia Celular , Quimiocina CCL5 , Células Progenitoras Endoteliales , MicroARNs , Neovascularización Fisiológica , Quimiocina CCL5/metabolismo , Quimiocina CCL5/genética , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/citología , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Animales , Neovascularización Fisiológica/genética , Ratones , Proliferación Celular , Masculino , Receptores CCR5/metabolismo , Receptores CCR5/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Regulación hacia Abajo/genética , Isquemia/metabolismo , Isquemia/patología , Isquemia/genética , Transducción de Señal , Angiogénesis
3.
Stem Cell Res Ther ; 15(1): 164, 2024 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-38853275

RESUMEN

BACKGROUND: Transplantation of CD34+ hematopoietic stem and progenitor cells (HSPC) into immunodeficient mice is an established method to generate humanized mice harbouring a human immune system. Different sources and methods for CD34+ isolation have been employed by various research groups, resulting in customized models that are difficult to compare. A more detailed characterization of CD34+ isolates is needed for a better understanding of engraftable hematopoietic and potentially non-hematopoietic cells. Here we have performed a direct comparison of CD34+ isolated from cord blood (CB-CD34+) or fetal liver (FL-CD34+ and FL-CD34+CD14-) and their engraftment into immunocompromised NOD/Shi-scid Il2rgnull (NOG) mice. METHODS: NOG mice were transplanted with either CB-CD34+, FL-CD34+ or FL-CD34+CD14- to generate CB-NOG, FL-NOG and FL-CD14--NOG, respectively. After 15-20 weeks, the mice were sacrificed and human immune cell reconstitution was assessed in blood and several organs. Liver sections were pathologically assessed upon Haematoxylin and Eosin staining. To assess the capability of allogenic tumor rejection in CB- vs. FL-reconstituted mice, animals were subcutaneously engrafted with an HLA-mismatched melanoma cell line. Tumor growth was assessed by calliper measurements and a Luminex-based assay was used to compare the cytokine/chemokine profiles. RESULTS: We show that CB-CD34+ are a uniform population of HSPC that reconstitute NOG mice more rapidly than FL-CD34+ due to faster B cell development. However, upon long-term engraftment, FL-NOG display increased numbers of neutrophils, dendritic cells and macrophages in multiple tissues. In addition to HSPC, FL-CD34+ isolates contain non-hematopoietic CD14+ endothelial cells that enhance the engraftment of the human immune system in FL-NOG mice. We demonstrate that these CD14+CD34+ cells are capable of reconstituting Factor VIII-producing liver sinusoidal endothelial cells (LSEC) in FL-NOG. However, CD14+CD34+ also contribute to hepatic sinusoidal dilatation and immune cell infiltration, which may culminate in a graft-versus-host disease (GVHD) pathology upon long-term engraftment. Finally, using an HLA-A mismatched CDX melanoma model, we show that FL-NOG, but not CB-NOG, can mount a graft-versus-tumor (GVT) response resulting in tumor rejection. CONCLUSION: Our results highlight important phenotypical and functional differences between CB- and FL-NOG and reveal FL-NOG as a potential model to study hepatic sinusoidal dilatation and mechanisms of GVT.


Asunto(s)
Antígenos CD34 , Hígado , Animales , Humanos , Antígenos CD34/metabolismo , Ratones , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos NOD , Trasplante de Células Madre Hematopoyéticas , Ratones SCID , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/trasplante , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Sangre Fetal/citología , Melanoma/patología , Melanoma/inmunología
4.
Int J Mol Sci ; 25(9)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38732080

RESUMEN

Endothelial progenitor cells (EPCs) play a critical role in cardiovascular regeneration. Enhancement of their native properties would be highly beneficial to ensuring the proper functioning of the cardiovascular system. As androgens have a positive effect on the cardiovascular system, we hypothesized that dihydrotestosterone (DHT) could also influence EPC-mediated repair processes. To evaluate this hypothesis, we investigated the effects of DHT on cultured human EPCs' proliferation, viability, morphology, migration, angiogenesis, gene and protein expression, and ability to integrate into cardiac tissue. The results showed that DHT at different concentrations had no cytotoxic effect on EPCs, significantly enhanced the cell proliferation and viability and induces fast, androgen-receptor-dependent formation of capillary-like structures. DHT treatment of EPCs regulated gene expression of androgen receptors and the genes and proteins involved in cell migration and angiogenesis. Importantly, DHT stimulation promoted EPC migration and the cells' ability to adhere and integrate into murine cardiac slices, suggesting it has a role in promoting tissue regeneration. Mass spectrometry analysis further highlighted the impact of DHT on EPCs' functioning. In conclusion, DHT increases the proliferation, migration, and androgen-receptor-dependent angiogenesis of EPCs; enhances the cells' secretion of key factors involved in angiogenesis; and significantly potentiates cellular integration into heart tissue. The data offer support for potential therapeutic applications of DHT in cardiovascular regeneration and repair processes.


Asunto(s)
Movimiento Celular , Proliferación Celular , Dihidrotestosterona , Células Progenitoras Endoteliales , Neovascularización Fisiológica , Receptores Androgénicos , Dihidrotestosterona/farmacología , Humanos , Movimiento Celular/efectos de los fármacos , Receptores Androgénicos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/citología , Animales , Células Cultivadas , Ratones , Supervivencia Celular/efectos de los fármacos , Andrógenos/farmacología , Andrógenos/metabolismo , Masculino
5.
Cell Transplant ; 33: 9636897241253144, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38798036

RESUMEN

This study tested the hypothesis that ITRI Biofilm prevents adhesion of the chest cavity. Combined extracorporeal shock wave (ECSW) + bone marrow-derived autologous endothelial progenitor cell (EPC) therapy was superior to monotherapy for improving heart function (left ventricular ejection fraction [LVEF]) in minipigs with ischemic cardiomyopathy (IC) induced by an ameroid constrictor applied to the mid-left anterior descending artery. The minipigs (n = 30) were equally designed into group 1 (sham-operated control), group 2 (IC), group 3 (IC + EPCs/by directly implanted into the left ventricular [LV] myocardium; 3 [+]/3[-] ITRI Biofilm), group 4 (IC + ECSW; 3 [+]/[3] - ITRI Biofilm), and group 5 (IC + EPCs-ECSW; 3 [+]/[3] - ITRI Biofilm). EPC/ECSW therapy was administered by day 90, and the animals were euthanized, followed by heart harvesting by day 180. In vitro studies demonstrated that cell viability/angiogenesis/cell migratory abilities/mitochondrial concentrations were upregulated in EPCs treated with ECSW compared with those in EPCs only (all Ps < 0.001). The LVEF was highest in group 1/lowest in group 2/significantly higher in group 5 than in groups 3/4 (all Ps < 0.0001) by day 180, but there was no difference in groups 3/4. The adhesion score was remarkably lower in patients who received ITRI Biofilm treatment than in those who did not (all Ps <0.01). The protein expressions of oxidative stress (NOX-1/NOX-2/oxidized protein)/apoptotic (mitochondrial-Bax/caspase3/PARP)/fibrotic (TGF-ß/Smad3)/DNA/mitochondria-damaged (γ-H2AX/cytosolic-cytochrome-C/p-DRP1), and heart failure/pressure-overload (BNP [brain natriuretic peptide]/ß-MHC [beta myosin heavy chain]) biomarkers displayed a contradictory manner of LVEF among the groups (all Ps < 0.0001). The protein expression of endothelial biomarkers (CD31/vWF)/small-vessel density revealed a similar LVEF within the groups (all Ps < 0.0001). ITRI Biofilm treatment prevented chest cavity adhesion and was superior in restoring IC-related LV dysfunction when combined with EPC/ECSW therapy compared with EPC/ECSW therapy alone.


Asunto(s)
Biopelículas , Células Progenitoras Endoteliales , Isquemia Miocárdica , Porcinos Enanos , Animales , Porcinos , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/citología , Isquemia Miocárdica/terapia , Isquemia Miocárdica/complicaciones , Tratamiento con Ondas de Choque Extracorpóreas/métodos , Miocardio/metabolismo , Miocardio/patología , Masculino
6.
Adv Healthc Mater ; 13(14): e2302830, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38366136

RESUMEN

Tissue engineering holds great promise for regenerative medicine, drug discovery, and as an alternative to animal models. However, as soon as the dimensions of engineered tissue exceed the diffusion limit of oxygen and nutriments, a necrotic core forms leading to irreversible damage. To overcome this constraint, the establishment of a functional perfusion network is essential. In this work, digital light processing bioprinting is used to encapsulate endothelial progenitor cells (EPCs) in 3D light-cured hydrogel scaffolds to guide them toward vascular network formation. In these scaffolds, EPCs proliferate and self-organize within a few days into branched tubular structures with predefined geometry, forming capillary-like vascular tubes or trees of diameters in the range of 10 to 100 µm. Presenting a confluent monolayer wall of cells strongly connect by tight junctions around a central lumen-like space, these structures can be microinjected with a fluorescent dye and are stable for several weeks in vitro. These endothelial structures can be recovered and manipulated in an alginate patch without altering their shape or viability. This approach opens new opportunities for future applications, such as stacking with other cell sheets or multicellular constructs to yield bioengineered tissue with higher complexity and functionality.


Asunto(s)
Bioimpresión , Células Progenitoras Endoteliales , Ingeniería de Tejidos , Andamios del Tejido , Humanos , Bioimpresión/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Hidrogeles/química , Capilares/fisiología , Alginatos/química , Impresión Tridimensional
7.
Oxid Med Cell Longev ; 2022: 1282362, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832490

RESUMEN

The low survival rate of endothelial progenitor cells (EPCs) in vivo which are susceptible to adverse microenvironments including inflammation and oxidative stress has become one primary challenge of EPCs transplantation for regenerative therapy. Recent studies reported functional expression of toll-like receptor (TLR) 4 on EPCs and dose-dependent effects of lipopolysaccharide (LPS) on cellular oxidative stress and angiogenic properties. However, the involved mechanism has not yet been elucidated well, and the influence of TLR4 signaling on EPCs survival and function in vivo is unknown. In the present study, we observed the effects of LPS and TLR4/SIRT3 on EPCs mitochondrial permeability and intracellular mitochondrial superoxide. We employed the monocrotaline-induced pulmonary arteriolar injury model to observe the effects of TLR4/SIRT3 on the recruitment and survival of transplanted EPCs. We found the destructive effects of 10 µg/mL LPS on mitochondrial homeostasis, and cellular viability was mediated by TLR4/SIRT3 signals at least partially, and the TLR4 mediates the early-stage recruitment of transplanted EPCs in pulmonary arteriolar inflammation injury; however, SIRT3 has more contribution to the survival of incorporated EPCs and ameliorated arteriolar remodeling in lung vascular tissue. The study provides insights for the critical role of TLR4/SIRT3 in LPS-induced oxidative stress and mitochondrial disorder in EPCs in vitro and in vivo. The TLR4/SIRT3 signaling is important for EPCs resistance against inflammation and oxidative stress and may represent a new manipulating target for developing efficient cell therapy strategy.


Asunto(s)
Células Progenitoras Endoteliales , Sirtuina 3 , Receptor Toll-Like 4 , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Homeostasis , Humanos , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Oxidación-Reducción , Sirtuina 3/metabolismo , Receptor Toll-Like 4/metabolismo
8.
Sci Rep ; 12(1): 1775, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110646

RESUMEN

Human adipose tissue is a rich source of adipose-derived stem cells (ASCs) and vascular endothelial progenitor cells (EPCs). However, no standardized method has been established for the isolation and purification of adipose-derived EPCs (AEPCs). The aim of this study was to establish a method for the isolation and purification of AEPCs. The stromal vascular fraction (SVF) was extracted from human lipoaspirates, and the CD45-CD31+ fraction of the SVF was collected by magnetic-activated cell sorting (MACS). The CD45-CD31+ fraction was cultured for 4.5 days, followed by a second MACS separation to collect the CD31+ fraction. Purified AEPCs were expanded without being overwhelmed by proliferating ASCs, indicating that a high level (> 95%) of AEPC purification is a key factor for their successful isolation and expansion. AEPCs exhibited typical endothelial markers, including CD31, von Willebrand factor, and the isolectin-B4 binding capacity. AEPCs formed colonies, comparable to cultured human umbilical vein endothelial cells (HUVECs). Both AEPCs and HUVECs formed capillary-like networks in the tube formation assay, with no significant difference in network lengths. We are the first to establish a purification and expansion method to isolate these cells. Because adipose tissue is a clinically accessible and abundant tissue, AEPCs may have potential advantages as a therapeutic tool for regenerative medicine.


Asunto(s)
Tejido Adiposo/citología , Biomarcadores/metabolismo , Células Progenitoras Endoteliales/citología , Medicina Regenerativa , Células del Estroma/citología , Tejido Adiposo/metabolismo , Adulto , Células Cultivadas , Células Progenitoras Endoteliales/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células del Estroma/metabolismo
9.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-35163466

RESUMEN

Ischemia-reperfusion injury (IRI) is a major cause of acute kidney injury (AKI) and progression to chronic kidney disease (CKD). However, no effective therapeutic intervention has been established for ischemic AKI. Endothelial progenitor cells (EPCs) have major roles in the maintenance of vascular integrity and the repair of endothelial damage; they also serve as therapeutic agents in various kidney diseases. Thus, we examined whether EPCs have a renoprotective effect in an IRI mouse model. Mice were assigned to sham, EPC, IRI-only, and EPC-treated IRI groups. EPCs originating from human peripheral blood were cultured. The EPCs were administered 5 min before reperfusion, and all mice were killed 72 h after IRI. Blood urea nitrogen, serum creatinine, and tissue injury were significantly increased in IRI mice; EPCs significantly improved the manifestations of IRI. Apoptotic cell death and oxidative stress were significantly reduced in EPC-treated IRI mice. Administration of EPCs decreased the expression levels of NLRP3, cleaved caspase-1, p-NF-κB, and p-p38. Furthermore, the expression levels of F4/80, ICAM-1, RORγt, and IL-17RA were significantly reduced in EPC-treated IRI mice. Finally, the levels of EMT-associated factors (TGF-ß, α-SMA, Snail, and Twist) were significantly reduced in EPC-treated IRI mice. This study shows that inflammasome-mediated inflammation accompanied by immune modulation and fibrosis is a potential target of EPCs as a treatment for IRI-induced AKI and the prevention of progression to CKD.


Asunto(s)
Lesión Renal Aguda/prevención & control , Células Progenitoras Endoteliales/trasplante , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/metabolismo , Animales , Apoptosis/efectos de los fármacos , Nitrógeno de la Urea Sanguínea , Células Cultivadas , Creatinina/sangre , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/inmunología , Células Progenitoras Endoteliales/metabolismo , Humanos , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo
10.
Int J Mol Sci ; 23(1)2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-35008953

RESUMEN

Clinically used heart valve prostheses, despite their progress, are still associated with limitations. Biodegradable poly-ε-caprolactone (PCL) nanofiber scaffolds, as a matrix, were seeded with human endothelial colony-forming cells (ECFCs) and human induced-pluripotent stem cells-derived MSCs (iMSCs) for the generation of tissue-engineered heart valves. Cell adhesion, proliferation, and distribution, as well as the effects of coating PCL nanofibers, were analyzed by fluorescence microscopy and SEM. Mechanical properties of seeded PCL scaffolds were investigated under uniaxial loading. iPSCs were used to differentiate into iMSCs via mesoderm. The obtained iMSCs exhibited a comparable phenotype and surface marker expression to adult human MSCs and were capable of multilineage differentiation. EFCFs and MSCs showed good adhesion and distribution on PCL fibers, forming a closed cell cover. Coating of the fibers resulted in an increased cell number only at an early time point; from day 7 of colonization, there was no difference between cell numbers on coated and uncoated PCL fibers. The mechanical properties of PCL scaffolds under uniaxial loading were compared with native porcine pulmonary valve leaflets. The Young's modulus and mean elongation at Fmax of unseeded PCL scaffolds were comparable to those of native leaflets (p = ns.). Colonization of PCL scaffolds with human ECFCs or iMSCs did not alter these properties (p = ns.). However, the native heart valves exhibited a maximum tensile stress at a force of 1.2 ± 0.5 N, whereas it was lower in the unseeded PCL scaffolds (0.6 ± 0.0 N, p < 0.05). A closed cell layer on PCL tissues did not change the values of Fmax (ECFCs: 0.6 ± 0.1 N; iMSCs: 0.7 ± 0.1 N). Here, a successful two-phase protocol, based on the timed use of differentiation factors for efficient differentiation of human iPSCs into iMSCs, was developed. Furthermore, we demonstrated the successful colonization of a biodegradable PCL nanofiber matrix with human ECFCs and iMSCs suitable for the generation of tissue-engineered heart valves. A closed cell cover was already evident after 14 days for ECFCs and 21 days for MSCs. The PCL tissue did not show major mechanical differences compared to native heart valves, which was not altered by short-term surface colonization with human cells in the absence of an extracellular matrix.


Asunto(s)
Biopolímeros/química , Caproatos/química , Células Progenitoras Endoteliales/citología , Válvulas Cardíacas , Células Madre Pluripotentes Inducidas/citología , Lactonas/química , Células Madre Mesenquimatosas/citología , Ingeniería de Tejidos , Andamios del Tejido , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Cultivadas , Células Progenitoras Endoteliales/metabolismo , Matriz Extracelular , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Nanofibras , Porcinos , Andamios del Tejido/química
11.
PLoS One ; 17(1): e0261498, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35025920

RESUMEN

BACKGROUND: A paracrine mechanism is thought to mediate the proangiogenic capacity of adipose-derived stromal/stem cells (ASCs). However, the precise mechanism by which ASCs promote the formation of blood vessels by endothelial progenitor cells (EPCs) is unclear. METHODS: The EPCs-ASCs cocultures prepared in different ratios were subjected to tube formations assay to verify whether ASCs could directly participate in the tube genesis. The supernatant from cultured ASCs was used to stimulate EPCs to evaluate the effects on the angiogenic property of EPCs, as well as capacity for migration and invasion. A coculture model with transwell chamber were used to explore the regulation of angiogenesis markers expression in EPCs by ASCs. We then mixed ASCs with EPCs and transplanted them with adipose tissue into nude mice to evaluate the effects on angiogenesis in adipose tissue grafts. RESULTS: In the EPCs-ASCs cocultures, the tube formation was significantly decreased as the relative abundance of ASCs increased, while the ASCs was found to migrate and integrated into the agglomerates formed by EPCs. The supernatant from ASCs cultures promoted the migration and invasion of EPCs and the ability to form capillary-like structures. The expression of multiple angiogenesis markers in EPCs were significantly increased when cocultured with ASCs. In vivo, ASCs combined with EPC promoted vascularization in the fat transplant. Immunofluorescence straining of Edu and CD31 indicated that the Edu labeled EPC did not directly participate in the vascularization inside the fat tissue. CONCLUSIONS: ADSC can participate in the tube formation of EPC although it cannot form canonical capillary structures. Meanwhile, Soluble factors secreted by ASCs promotes the angiogenic potential of EPCs. ASCs paracrine signaling appears to promote angiogenesis by increasing the migration and invasion of EPCs and simultaneously upregulating the expression of angiogenesis markers in EPCs. The results of in vivo experiments showed that ASCs combined with EPCs significantly promote the formation of blood vessels in the fat implant. Remarkably, EPCs may promote angiogenesis by paracrine regulation of endogenous endothelial cells (ECs) rather than direct participation in the formation of blood vessels.


Asunto(s)
Células Progenitoras Endoteliales/trasplante , Supervivencia de Injerto/fisiología , Neovascularización Fisiológica/fisiología , Células del Estroma/trasplante , Tejido Adiposo/citología , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Animales , Técnicas de Cultivo de Célula , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Ratones , Ratones Desnudos , Comunicación Paracrina/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Conejos , Células del Estroma/citología , Células del Estroma/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Dev Biol ; 482: 44-54, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34915023

RESUMEN

Development of the mammalian lymphatic vasculature is a stepwise process requiring the specification of lymphatic endothelial cell progenitors in the embryonic veins, and their subsequent budding to give rise to most of the mature lymphatic vasculature. In mice, formation of the lymphatic vascular network starts inside the cardinal vein at around E9.5 when a subpopulation of venous endothelial cells gets committed into the lymphatic lineage by their acquisition of Prox1 expression. Identification of critical genes regulating lymphatic development facilitated the detailed cellular and molecular characterization of some of the cellular and molecular mechanisms regulating the early steps leading to the formation of the mammalian lymphatic vasculature. A better understanding of basic aspects of early lymphatic development, and the availability of novel tools and animal models has been instrumental in the identification of important novel functional roles of this vasculature network.


Asunto(s)
Células Endoteliales/citología , Células Progenitoras Endoteliales/citología , Linfangiogénesis/genética , Linfangiogénesis/fisiología , Vasos Linfáticos/embriología , Animales , Embrión de Mamíferos/embriología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Food Funct ; 13(1): 76-90, 2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-34882161

RESUMEN

Blood outgrowth endothelial cells (BOECs) have received growing attention in relation to cardiovascular disease (CVD). However, the effect of diet intervention, a primary strategy for CVD prevention, on BOECs is not reported. This study aims to investigate the effect of following a healthy dietary pattern (HDP) with or without wolfberry consumption, healthy food with potential cardiovascular benefits, on the number and function of BOECs in middle-aged and older adults. Twenty-four subjects consumed either an HDP only (n = 9) or an HDP supplemented with 15 g day-1 wolfberries (n = 15) for 16 weeks. At pre- and post-intervention, vascular health biomarkers and composite CVD risk indicators were assessed. BOECs were derived from peripheral blood mononuclear cells and their angiogenic and migration activities were measured. Isolated BOECs have typical endothelial cobblestone morphology, express von Willebrand factor and KDR. Consuming an HDP improved the BOEC colony's growth rate, which was demonstrated by significant time effects in the colony's culture time between passages 1 and 2 (P = 0.038). Both interventions increased BOECs' tube formation capacity. Moreover, HDP intervention contributed to a time effect on BOEC migration activity (P = 0.040 for t1/2gap). Correlation analysis revealed that BOEC colony number was positively associated with blood pressure, atherogenic index, vascular age, and Framingham risk score. In conclusion, adherence to an HDP improved BOECs' function in middle-aged and older populations, while additional wolfberry consumption did not provide an enhanced effect. Our results provide mechanistic dissection on the beneficial effects on BOECs of dietary pattern modification.


Asunto(s)
Dieta Saludable , Células Progenitoras Endoteliales , Frutas , Factores de Riesgo de Enfermedad Cardiaca , Lycium , Presión Sanguínea/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/fisiología , Femenino , Humanos , Lípidos/sangre , Masculino , Persona de Mediana Edad
14.
Exp Cell Res ; 411(1): 112985, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34942190

RESUMEN

Deep venous thrombosis (DVT) endangers human health. Endothelial progenitor cells (EPCs) were proven to promote thrombolysis and miR-204-5p was discovered to be low-expressed in DVT patients. This study concentrated on exploring whether miR-204-5p had a regulatory effect on EPCs and DVT. Concretely, the expression of miR-204-5p in DVT patients' blood was detected by qRT-PCR. The target of miR-204-5p was predicted by bioinformatics and verified by dual-luciferase reporter assay. After rat EPCs were isolated, identified, and transfected with miR-204-5p agomiR, antagomiR, or SPRED1 plasmids, the viability, migration, invasion, and tube formation of EPCs were detected by MTT, wound healing, Transwell, and tube formation assays, respectively. MiR-204-5p, SPRED1, p-PI3K, PI3K, p-AKT, AKT, VEGFA, and Ang1 expressions in EPCs were measured by qRT-PCR or Western blot. EPCs transfected with miR-204-5p overexpression lentivirus plasmid were injected into the DVT rat model. The histopathology of the thrombus and the homing of EPCs to thrombus in the DVT rats were observed by hematoxylin-eosin staining and confocal microscopy, respectively. We found that miR-204-5p was low-expressed in DVT patients and SPRED1 was a target gene of miR-204-5p. MiR-204-5p agomiR promoted the viability, migration, invasion, and tube formation of EPCs, the levels of VEGFA and Ang1 and the activation of PI3K/AKT pathway in EPCs, while miR-204-5p antagomiR and SPRED1 worked oppositely. SPRED1 reversed the effect of miR-204-5p agomiR on EPCs. Up-regulated miR-204-5p inhibited thrombosis and promoted EPCs homing to thrombus in DVT rats. Collectively, up-regulated miR-204-5p enhanced the angiogenesis of EPCs and thrombolysis in DVT rats by targeting SPRED1.


Asunto(s)
Células Progenitoras Endoteliales/fisiología , Regulación de la Expresión Génica , MicroARNs/genética , Neovascularización Fisiológica , Proteínas Represoras/antagonistas & inhibidores , Terapia Trombolítica/métodos , Trombosis de la Vena/terapia , Adulto , Animales , Apoptosis , Biomarcadores/metabolismo , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Células Cultivadas , Células Progenitoras Endoteliales/citología , Femenino , Humanos , Masculino , Pronóstico , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Activación Transcripcional , Regulación hacia Arriba , Trombosis de la Vena/metabolismo , Trombosis de la Vena/patología
15.
J Diabetes Res ; 2021: 9962877, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34722777

RESUMEN

Endothelial progenitor cells (EPCs) are involved in the neovascularization in traumatic and ischemic sites, but EPCs are "detained" in bone marrow under diabetic conditions, which results in reduction of the number of EPCs and their biological activity in peripheral blood. Based on our previous study to mobilize autologous bone marrow EPCs by administering AMD3100+G-CSF to realize the optimal effect, our present study is aimed at exploring the effects of transplanting EPCs locally in a wound model of diabetic mice. First, we prepared and identified EPCs, and the biological functions and molecular characteristics were compared between EPCs from DB/+ and DB/DB mice. Then, we performed full-thickness skin resection in DB/DB mice and tested the effect of local transplantation of EPCs on skin wound healing. The wound healing process was recorded using digital photographs. The animals were sacrificed on postoperative days 7, 14, and 17 for histological and molecular analysis. Our results showed that DB/+ EPCs were biologically more active than those of DB/DB EPCs. When compared with the control group, local transplantation of EPCs accelerated wound healing in DB/DB mice by promoting wound granulation tissue formation, angiogenesis, and collagen fiber deposition, but there was no significant difference in wound healing between DB/+ EPCs and DB/DB EPCs transplanted into the wound. Furthermore, local transplantation of EPCs promoted the expression of SDF-1, CXCR4, and VEGF. We speculated that EPC transplantation may promote wound healing through the SDF-1/CXCR4 axis. This point is worth exploring further. Present data are of considerable significance because they raise the possibility of promoting wound healing by isolating autologous EPCs from the patient, which provides a new approach for the clinical treatment of diabetic wounds in the future.


Asunto(s)
Movimiento Celular , Diabetes Mellitus/metabolismo , Células Progenitoras Endoteliales/trasplante , Neovascularización Fisiológica , Piel/lesiones , Cicatrización de Heridas , Animales , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Ratones , Piel/metabolismo , Piel/patología , Trasplante Homólogo
16.
Eur Rev Med Pharmacol Sci ; 25(19): 6101-6108, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34661270

RESUMEN

OBJECTIVE: The purpose of this study was to explore the relationship between the number of endothelial progenitor cells (EPCs) and coronary heart disease (CHD). PATIENTS AND METHODS: A total of 24 patients with CHD were chosen from Lanzhou City and Xianyang City, and then, 24 healthy controls who matched the CHD group in gender, age and address were chosen as control group. C-reactive protein (CRP) and c-reaction protein (hs-CRP) were detected. The levels of interleukin-8 (IL-8), vascular endothelial growth factor (VEGF), homocysteine (Hcy), hypoxia-inducible factor-1 (HIF-1α) and stromal cell-derived factor 1 (SDF-1α) were detected. RESULTS: The number of EPCs in control groups was both increased compared with CHD group (p<0.05). The number of EPCs in Xianyang control group was increased compared with Lanzhou control group (p<0.05). Compared with the control group, the levels of TC, LDL and CRP in the CHD group were higher (p<0.05). Compared with Lanzhou control group, Hcy level was decreased in Lanzhou CHD group (p<0.05). Compared with Xianyang control group, the levels of IL-8 and VEGF were increased, but the levels of HIF-1α and Hcy were decreased in the Xianyang CHD group (p<0.05). The expressions of IL-8, VEGF, Hcy and HIF-1α were increased in Lanzhou control group than the Xianyang control group (p<0.05). In Lanzhou CHD group, Spearman correlation analysis showed that the number of EPCs was negatively related to hs-CRP content (r=-0.631, p<0.05). CONCLUSIONS: The decrease of EPCs caused by high altitude may increase the expressions of various cytokines, leading to the occurrence of CHD.


Asunto(s)
Altitud , Enfermedad Coronaria/etiología , Citocinas/metabolismo , Células Progenitoras Endoteliales/citología , Adulto , Anciano , Proteína C-Reactiva/metabolismo , Estudios de Casos y Controles , Enfermedad Coronaria/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo
17.
Bioengineered ; 12(1): 8031-8042, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34670479

RESUMEN

The present study aimed to prepare a kind of controlled-releasing insulin-like growth factor 1 (IGF-1)/spider silk protein nanofibrous membrane using a electrostatic spinning method and evaluated its effect on the cell viability of endothelial progenitor cells (EPCs). Recombinant spidroin named as GMCDRSSP-IgF-1 was electro-spun into nanofibrous membrane which can be degraded by protease and be capable of sustained-release of IGF-1. The membrane can be degraded after being treated with thrombin. The release assay results showed that IGF-1 concentration could be maintained at 20 ng/ml for a long time with treatment of Tobacco Etch Virus (TEV) protease. The viability of EPCs on GMCDRSSP-IgF-1 nanofibrous membrane was significantly increased with the presence of TEV protease. The controlled and sustained release of IGF-1 from the nanofibrous membrane could promote the adhesion and viability of EPCs. In summary, the nanofibrous membrane that exhibits controlled degradation and sustained release of IGF-1 was prepared with electrostatic spinning from genetically modified recombinant spider silk protein. The nanofibrous membrane exhibited good blood compatibility and cytocompatibility. With the presence of TEV protease, the sustained-release of IGF-1 significantly promoted the adhesion and viability of EPCs. The new nanofibrous membrane can be potentially used as a scaffold for EPCs culture in vitro and future in vivo studies.


Asunto(s)
Células Progenitoras Endoteliales/citología , Fibroínas/genética , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/farmacología , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cisteína Endopeptidasas/metabolismo , Preparaciones de Acción Retardada , Fibroínas/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Proteínas Recombinantes/farmacología , Electricidad Estática , Ingeniería de Tejidos , Andamios del Tejido/química
18.
J Cell Mol Med ; 25(21): 10088-10100, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34592781

RESUMEN

It was shown that endothelial progenitor cells (EPCs) have bidirectional differentiation potential and thus perform different biological functions. The purpose of this study was to investigate the effects of slight up-regulation of the Kir2.1 channel on EPC transdifferentiation and the potential mechanism on cell function and transformed cell type. First, we found that the slight up-regulation of Kir2.1 expression promoted the expression of the stem cell stemness factors ZFX and NS and inhibited the expression of senescence-associated ß-galactosidase. Further studies showed the slightly increased expression of Kir2.1 could also improve the expression of pericyte molecular markers NG2, PDGFRß and Desmin. Moreover, adenovirus-mediated Kir2.1 overexpression had an enhanced contractile response to norepinephrine of EPCs. These results suggest that the up-regulated expression of the Kir2.1 channel promotes EPC transdifferentiation into a pericyte phenotype. Furthermore, the mechanism of EPC transdifferentiation to mesenchymal cells (pericytes) was found to be closely related to the channel functional activity of Kir2.1 and revealed that this channel could promote EPC EndoMT by activating the Akt/mTOR/Snail signalling pathway. Overall, this study suggested that in the early stage of inflammatory response, regulating the Kir2.1 channel expression affects the biological function of EPCs, thereby determining the maturation and stability of neovascularization.


Asunto(s)
Diferenciación Celular , Células Progenitoras Endoteliales/metabolismo , Pericitos/metabolismo , Canales de Potasio de Rectificación Interna/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Biomarcadores , Autorrenovación de las Células , Células Cultivadas , Senescencia Celular , Desmina/metabolismo , Células Progenitoras Endoteliales/citología , Modelos Biológicos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Pericitos/citología , Canales de Potasio de Rectificación Interna/metabolismo , Ratas , Transducción de Señal
19.
J Diabetes Res ; 2021: 6626627, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34557552

RESUMEN

BACKGROUND: Circulating endothelial progenitor cells (EPCs) play important roles in vascular repair. However, the mechanisms of high-glucose- (HG-) induced cord blood EPC senescence and the role of B2 receptor (B2R) remain unknown. METHODS: Cord blood samples from 26 patients with gestational diabetes mellitus (GDM) and samples from 26 healthy controls were collected. B2R expression on circulating CD34+ cells of cord blood mononuclear cells (CBMCs) was detected using flow cytometry. The plasma concentrations of 8-isoprostaglandin F2α (8-iso-PGF2α) and nitric oxide (NO) were measured. EPCs were treated with HG (40 mM) alone or with bradykinin (BK) (1 nM). The B2R and eNOS small interfering RNAs (siRNAs) and the PI3K antagonist LY294002 were added to block B2R, eNOS, and PI3K separately. To determine the number of senescent cells, senescence-associated ß-galactosidase (SA-ß-gal) staining was performed. The level of mitochondrial reactive oxygen species (ROS) in EPCs was assessed by Mito-Sox staining. Cell viability was evaluated by Cell Counting Kit-8 (CCK-8) assays. Mitochondrial DNA (mtDNA) copy number and the relative length of telomeres were detected by real time-PCR. The distribution of human telomerase reverse transcriptase (hTERT) in the nucleus, cytosol, and mitochondria of EPCs was detected by immunofluorescence. The expression of B2R, p16, p21, p53, P-Ser473AKT, T-AKT, eNOS, and hTERT was demonstrated by Western blot. RESULTS: B2R expression on circulating CD34+ cells of CBMCs was significantly reduced in patients with GDM compared to healthy controls. Furthermore, B2R expression on circulating CD34+ cells of CBMCs was inversely correlated with plasma 8-iso-PGF2α concentrations and positively correlated with plasma NO levels. BK treatment decreased EPC senescence and ROS generation. Furthermore, BK treatment of HG-exposed cells led to elevated P-Ser473AKT and eNOS protein expression compared with HG treatment alone. BK reduced hTERT translocation in HG-induced senescent EPCs. B2R siRNA, eNOS siRNA, and antagonist of the PI3K signalling pathway blocked the protective effects of BK. CONCLUSION: BK, acting through PI3K-AKT-eNOS signalling pathways, reduced hTERT translocation, increased the relative length of telomeres while reducing mtDNA copy number, and finally protected against EPC senescence induced by HG.


Asunto(s)
Bradiquinina/farmacología , Senescencia Celular/efectos de los fármacos , Células Progenitoras Endoteliales/efectos de los fármacos , Receptor de Bradiquinina B2/metabolismo , Estudios de Casos y Controles , Células Cultivadas , ADN Mitocondrial/genética , Diabetes Gestacional , Dinoprost/análogos & derivados , Dinoprost/sangre , Células Progenitoras Endoteliales/citología , Femenino , Sangre Fetal , Dosificación de Gen , Glucosa/farmacología , Humanos , Recién Nacido , Óxido Nítrico/sangre , Óxido Nítrico Sintasa de Tipo III/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Telomerasa , Telómero
20.
BMB Rep ; 54(9): 470-475, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34488932

RESUMEN

Low-dose metronomic chemotherapy has been introduced as a less toxic and effective strategy to inhibit tumor angiogenesis, but its anti-angiogenic mechanism on endothelial progenitor cells (EPCs) has not been fully elucidated. Here, we investigated the functional role of regulated in development and DNA damage response 1 (REDD1), an endogenous inhibitor of mTORC1, in low-dose doxorubicin (DOX)-mediated dysregulation of EPC functions. DOX treatment induced REDD1 expression in bone marrow mononuclear cells (BMMNCs) and subsequently reduced mTORC1-dependent translation of endothelial growth factor (VEGF) receptor (Vegfr)-2 mRNA, but not that of the mRNA transcripts for Vegfr-1, epidermal growth factor receptor, and insulin-like growth factor-1 receptor. This selective event was a risk factor for the inhibition of BMMNC differentiation into EPCs and their angiogenic responses to VEGF-A, but was not observed in Redd1-deficient BMMNCs. Low-dose metronomic DOX treatment reduced the mobilization of circulating EPCs in B16 melanoma-bearing wild-type but not Redd1-deficient mice. However, REDD1 overexpression inhibited the differentiation and mobilization of EPCs in both wild-type and Redd1-deficient mice. These data suggest that REDD1 is crucial for metronomic DOX-mediated EPC dysfunction through the translational repression of Vegfr-2 transcript, providing REDD1 as a potential therapeutic target for the inhibition of tumor angiogenesis and tumor progression. [BMB Reports 2021; 54(9): 470-475].


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Doxorrubicina/farmacología , Factores de Transcripción/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Ratones , Ratones Noqueados , Neovascularización Patológica , Óxido Nítrico/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA