Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 330
Filtrar
1.
Neuromolecular Med ; 26(1): 25, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38886284

RESUMEN

This comprehensive review explores the multifaceted role of endothelial progenitor cells (EPCs) in vascular diseases, focusing on their involvement in the pathogenesis and their contributions to enhancing the efficacy of endovascular treatments for intracranial aneurysms (IAs). Initially discovered as CD34+ bone marrow-derived cells implicated in angiogenesis, EPCs have been linked to vascular repair, vasculogenesis, and angiogenic microenvironments. The origin and differentiation of EPCs have been subject to debate, challenging the conventional notion of bone marrow origin. Quantification methods, including CD34+ , CD133+ , and various assays, reveal the influence of factors, like age, gender, and comorbidities on EPC levels. Cellular mechanisms highlight the interplay between bone marrow and angiogenic microenvironments, involving growth factors, matrix metalloproteinases, and signaling pathways, such as phosphatidylinositol-3-kinase (PI3K) and mitogen-activated protein kinase (MAPK). In the context of the pathogenesis of IAs, EPCs play a role in maintaining vascular integrity by replacing injured and dysfunctional endothelial cells. Recent research has also suggested the therapeutic potential of EPCs after coil embolization and flow diversion, and this has led the development of device surface modifications aimed to enhance endothelialization. The comprehensive insights underscore the importance of further research on EPCs as both therapeutic targets and biomarkers in IAs.


Asunto(s)
Células Progenitoras Endoteliales , Aneurisma Intracraneal , Humanos , Aneurisma Intracraneal/terapia , Células Progenitoras Endoteliales/fisiología , Células Progenitoras Endoteliales/trasplante , Procedimientos Endovasculares/métodos , Diferenciación Celular , Animales , Transducción de Señal , Neovascularización Fisiológica , Embolización Terapéutica , Neovascularización Patológica
2.
Restor Neurol Neurosci ; 42(2): 139-149, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38820024

RESUMEN

Background: Hematopoietic stem cells (HSC) are recruited to ischemic areas in the brain and contribute to improved functional outcome in animals. However, little is known regarding the mechanisms of improvement following HSC administration post cerebral ischemia. To better understand how HSC effect post-stroke improvement, we examined the effect of HSC in ameliorating motor impairment and cortical dysfunction following cerebral ischemia. Methods: Baseline motor performance of male adult rats was established on validated motor tests. Animals were assigned to one of three experimental cohorts: control, stroke, stroke + HSC. One, three and five weeks following a unilateral stroke all animals were tested on motor skills after which intracortical microstimulation was used to derive maps of forelimb movement representations within the motor cortex ipsilateral to the ischemic injury. Results: Stroke + HSC animals significantly outperformed stroke animals on single pellet reaching at weeks 3 and 5 (28±3% and 33±3% versus 11±4% and 17±3%, respectively, p < 0.05 at both time points). Control animals scored 44±1% and 47±1%, respectively. Sunflower seed opening task was significantly improved in the stroke + HSC cohort versus the stroke cohort at week five-post stroke (79±4 and 48±5, respectively, p < 0.05). Furthermore, Stroke + HSC animals had significantly larger forelimb motor maps than animals in the stroke cohort. Overall infarct size did not significantly differ between the two stroked cohorts. Conclusion: These data suggest that post stroke treatment of HSC enhances the functional integrity of residual cortical tissue, which in turn supports improved behavioral outcome, despite no observed reduction in infarct size.


Asunto(s)
Isquemia Encefálica , Modelos Animales de Enfermedad , Corteza Motora , Animales , Masculino , Corteza Motora/fisiopatología , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Ratas , Mapeo Encefálico , Células Progenitoras Endoteliales/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Recuperación de la Función/fisiología , Miembro Anterior/fisiopatología , Ratas Sprague-Dawley
3.
Semin Cell Dev Biol ; 155(Pt C): 23-29, 2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-37202277

RESUMEN

The interests in blood endothelial cells arise from their therapeutic potential in vascular repair and regeneration. Our understanding of blood endothelial cells that exist in the circulation has been evolving significantly from the original concept of endothelial progenitor cells. Many studies have uncovered heterogeneities of blood endothelial subtypes where some cells express both endothelial and hematopoietic antigens, and others possess either mature or immature endothelial markers. Due to the lack of definitive cell marker identities, there have been momentums in the field to adopt a technical-oriented labeling system based on the cells' involvement in postnatal neovascularization and cell culture derivatives. Our review streamlines nomenclatures for blood endothelial subtypes and standardizes understanding of their functional differences. Broadly, we will discuss about myeloid angiogenic cells (MACs), endothelial colony-forming cells (ECFCs), blood outgrowth endothelial cells (BOECs) and circulating endothelial cells (CECs). The strategic location of blood endothelial cells confers them essential roles in supporting physiological processes. MACs exert angiogenic effects through paracrine mechanisms, while ECFCs are recruited to sites of vascular injury to participate directly in new vessel formation. BOECs are an in vitro derivative of ECFCs. CECs are shed into the bloodstream from damaged vessels, hence reflective of endothelial dysfunction. With clarity on the functional attributes of blood endothelial subtypes, we present recent advances in their applications in disease modelling, along with serving as biomarkers of vascular tissue homeostasis.


Asunto(s)
Células Progenitoras Endoteliales , Células Progenitoras Endoteliales/fisiología , Técnicas de Cultivo de Célula , Biomarcadores , Neovascularización Fisiológica , Células Cultivadas
4.
Am J Respir Cell Mol Biol ; 68(2): 161-175, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36287629

RESUMEN

Acute respiratory distress syndrome/acute lung injury (ARDS/ALI) involves acute respiratory failure characterized by vascular endothelial and lung alveolar epithelial injury. Endothelial progenitor cells (EPCs) can mediate vasculogenesis. However, the limitations of EPCs, such as low survival and differentiation, are believed to inhibit the effectiveness of autologous cell therapies. This study demonstrated that lysophosphatidic acid (LPA), a bioactive small molecule without immunogenicity, is involved in the survival and antiapoptotic effects in human umbilical cord mesenchymal stem cells. This study aimed to explore whether LPA improves the survival of EPCs, enhancing the cellular therapeutic efficacy in ARDS, and these results will expand the application of LPA in stem cells and regenerative medicine. LPA promoted the colony formation, proliferation, and migration of EPCs and upregulated the expression of vascular endothelial-derived growth factor (VEGF) in EPCs. LPA pretreatment of transplanted EPCs improved the therapeutic effect by increasing EPC numbers in the rat lungs. LPA enhanced EPC proliferation and migration through Lpar1 coupled to Gi/o and Gq/11, respectively. Activation of extracellular signal-related kinase 1/2, or ERK1/2, was related to LPA-induced EPC proliferation but not migration. LPA/Lpar1-mediated Gi/o protein was also shown to be involved in promoting VEGF expression and inhibiting IL-1α expression in EPCs. Low LPA concentrations are present after lung injury; thus, the restoration of LPA may promote endothelial cell homeostasis and lung repair in ARDS. Inhalation of LPA significantly promoted the homing of endogenous EPCs to the lung and reduced lung injury in both rats with LPS-induced ALI and Streptococcus pneumoniae-infected mice. Taken together, these data indicated that LPA/Lpar1-mediated effects in EPCs are involved in maintaining endothelial cell homeostasis and lung tissue repair under physiological conditions.


Asunto(s)
Lesión Pulmonar Aguda , Células Progenitoras Endoteliales , Síndrome de Dificultad Respiratoria , Humanos , Ratas , Ratones , Animales , Células Progenitoras Endoteliales/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Pulmón/metabolismo , Síndrome de Dificultad Respiratoria/terapia , Síndrome de Dificultad Respiratoria/metabolismo , Lesión Pulmonar Aguda/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo
5.
Pharmacol Ther ; 241: 108315, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36436689

RESUMEN

Extensive injury of endothelial cells in blood vasculature, especially in the microcirculatory system, frequently occurs in hosts suffering from sepsis and the accompanied systemic inflammation. Pathological factors, including toxic components derived from invading microbes, oxidative stress associated with tissue ischemia/reperfusion, and vessel active mediators generated during the inflammatory response, are known to play important roles in mediating endothelial injury. Collapse of microcirculation and tissue edema developed from the failure of endothelial barrier function in vital organ systems, including the lung, brain, and kidney, are detrimental, which often predict fatal outcomes. The host body possesses a substantial capacity for maintaining vascular homeostasis and repairing endothelial damage. Bone marrow and vascular wall niches house endothelial progenitor cells (EPCs). In response to septic challenges, EPCs in their niche environment are rapidly activated for proliferation and angiogenic differentiation. In the meantime, release of EPCs from their niches into the blood stream and homing of these vascular precursors to tissue sites of injury are markedly increased. The recruited EPCs actively participate in host defense against endothelial injury and repair of damage in blood vasculature via direct differentiation into endothelial cells for re-endothelialization as well as production of vessel active mediators to exert paracrine and autocrine effects on angiogenesis/vasculogenesis. In recent years, investigations on significance of EPCs in host defense and molecular signaling mechanisms underlying regulation of the EPC response have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches for effective prevention and treatment of sepsis-induced vascular injury as well as vital organ system failure.


Asunto(s)
Células Progenitoras Endoteliales , Sepsis , Humanos , Células Progenitoras Endoteliales/fisiología , Microcirculación , Transducción de Señal , Diferenciación Celular
6.
Respir Res ; 23(1): 290, 2022 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-36274148

RESUMEN

BACKGROUND: Plexiform lesions, which have a dynamic appearance in structure and cellular composition, are the histological hallmark of severe pulmonary arterial hypertension in humans. The pathogenesis of the lesion development remains largely unknown, although it may be related to local inflammation and dysfunction in early progenitor endothelial cells (eEPCs). We tested the hypothesis that eEPCs contribute to the development of plexiform lesions by differentiating into macrophages in the setting of chronic inflammation. METHODS: The eEPC markers CD133 and VEGFR-2, macrophage lineage marker mannose receptor C-type 1 (MRC1), TNFα and nuclear factor erythroid 2-related factor 2 (Nrf2) in plexiform lesions in a broiler model were determined by immunohistochemistry. eEPCs derived from peripheral blood mononuclear cells were exposed to TNFα, and macrophage differentiation and angiogenic capacity of the cells were evaluated by phagocytotic and Matrigel plug assays, respectively. The role of Nrf2 in eEPC-to-macrophage transition as well as in MRC1 expression was also evaluated. Intratracheal installation of TNFα was conducted to determine the effect of local inflammation on the formation of plexiform lesions. RESULTS: Cells composed of the early lesions have a typical eEPC phenotype whereas those in more mature lesions display molecular and morphological characteristics of macrophages. Increased TNFα production in plexiform lesions was observed with lesion progression. In vitro studies showed that chronic TNFα challenge directed eEPCs to macrophage differentiation accompanied by hyperactivation of Nrf2, a stress-responsive transcription factor. Nrf2 activation (Keap1 knockdown) caused a marked downregulation in CD133 but upregulation in MRC1 mRNA. Dual luciferase reporter assay demonstrated that Nrf2 binds to the promoter of MRC1 to trigger its expression. In good agreement with the in vitro observation, TNFα exposure induced macrophage differentiation of eEPCs in Matrigel plugs, resulting in reduced neovascularization of the plugs. Intratracheal installation of TNFα resulted in a significant increase in plexiform lesion density. CONCLUSIONS: This work provides evidence suggesting that macrophage differentiation of eEPCs resulting from chronic inflammatory stimulation contributes to the development of plexiform lesions. Given the key role of Nrf2 in the phenotypic switching of eEPCs to macrophages, targeting this molecular might be beneficial for intervention of plexiform lesions.


Asunto(s)
Células Progenitoras Endoteliales , Hipertensión Pulmonar , Animales , Humanos , Células Progenitoras Endoteliales/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular , Factor de Necrosis Tumoral alfa , Factor 2 Relacionado con NF-E2 , Proteína 1 Asociada A ECH Tipo Kelch , Leucocitos Mononucleares , Pollos , Inflamación , Macrófagos , ARN Mensajero
7.
Nat Rev Rheumatol ; 18(5): 286-300, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35393604

RESUMEN

The observations that traditional cardiovascular disease (CVD) risk factors fail to fully account for the excessive cardiovascular mortality in patients with systemic lupus erythematosus (SLE) compared with the general population have prompted in-depth investigations of non-traditional, SLE-related risk factors that contribute to cardiovascular complications in patients with SLE. Of the various perturbations of vascular physiology, endothelial dysfunction, which is believed to occur in the earliest step of atherosclerosis, has been extensively investigated for its contribution to CVD risk in SLE. Endothelial progenitor cells (EPCs), which play a crucial part in vascular repair, neovascularization and maintenance of endothelial function, are quantitatively and functionally reduced in patients with SLE. Yet, the lack of a unified definition of EPCs, standardization of the quantity and functional assessment of EPCs as well as endothelial function measurement pose challenges to the translation of endothelial function measurements and EPC levels into prognostic markers for CVD in patients with SLE. This Review discusses factors that contribute to CVD in SLE, with particular focus on how endothelial function and EPCs are evaluated currently, and how EPCs are quantitatively and functionally altered in patients with SLE. Potential strategies for the use of endothelial function measurements and EPC quantification as prognostic markers of CVD in patients with SLE, and the limitations of their prognostication potential, are also discussed.


Asunto(s)
Aterosclerosis , Células Progenitoras Endoteliales , Lupus Eritematoso Sistémico , Células Progenitoras Endoteliales/fisiología , Humanos , Lupus Eritematoso Sistémico/epidemiología , Neovascularización Patológica , Factores de Riesgo
8.
Eur J Appl Physiol ; 122(7): 1589-1625, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35305142

RESUMEN

Circulating endothelial progenitor cells (EPCs) contribute to vascular healing and neovascularisation, while exercise is an effective means to mobilise EPCs into the circulation. OBJECTIVES: to systematically examine the acute and chronic effects of different forms of exercise on circulating EPCs in healthy populations. METHODS: Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines were followed. RESULTS: thirty-one articles met the inclusion criteria including 747 participants aged 19 to 76 years. All included trials used flow cytometry for identification of circulating EPCs. Eight and five different EPC phenotypes were identified in the acute and chronic trials, respectively. In the acute trials, moderate intensity continuous (MICON), maximal, prolonged endurance, resistance and high intensity interval training (HIIT) exercise protocols were utilised. Prolonged endurance and resistance exercise had the most profound effect on circulating EPCs followed by maximal exercise. In the chronic trials, MICON exercise, HIIT, HIIT compared to MICON and MICON compared to exergame (exercise modality based on an interactive video game) were identified. MICON exercise had a positive effect on circulating EPCs in older sedentary individuals which was accompanied by improvements in endothelial function and arterial stiffness. Long-stage HIIT (4 min bouts) appears to be an effective means and superior than MICON exercise in mobilising circulating EPCs. In conclusion, both in acute and chronic trials the degree of exercise-induced EPC mobilisation depends upon the exercise regime applied. In future, more research is warranted to examine the dose-response relationship of different exercise forms on circulating EPCs using standardised methodology and EPC phenotype.


Asunto(s)
Células Progenitoras Endoteliales , Entrenamiento de Intervalos de Alta Intensidad , Anciano , Células Progenitoras Endoteliales/fisiología , Ejercicio Físico/fisiología , Terapia por Ejercicio , Citometría de Flujo , Humanos
9.
J Diabetes Res ; 2022: 4067812, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35155684

RESUMEN

Systemic vascular impairment is the most common complication of diabetes. Advanced glycation end products (AGEs) can exacerbate diabetes-related vascular damage by affecting the intima and media through a variety of mechanisms. In the study, we demonstrated that AGEs and their membrane receptor RAGE could induce the differentiation of EPCs into osteoblasts under certain circumstances, thereby promoting accelerated atherosclerosis. Differentiation into osteoblasts was confirmed by positive staining for DiI-acetylated fluorescently labeled low-density lipoprotein and FITC-conjugated Ulex europaeus agglutinin. During differentiation, expression of receptor for AGE (RAGE) was significantly upregulated. This upregulation was attenuated by transfection with RAGE-targeting small interfering (si)RNA. siRNA-mediated knockdown of RAGE expression significantly inhibited the upregulation of AGE-induced calcification-related proteins, such as runt-related transcription factor 2 (RUNX2) and osteoprotegerin (OPG). Additional experiments showed that AGE induction of EPCs significantly induced ERK, p38MAPK, and JNK activation. The AGE-induced upregulation of osteoblast proteins (RUNX2 and OPG) was suppressed by treatment with a p38MAPK inhibitor (SB203580) or JNK inhibitor (SP600125), but not by treatment with an ERK inhibitor (PD98059), which indicated that AGE-induced osteoblast differentiation from EPCs may be mediated by p38MAPK and JNK signaling, but not by ERK signaling. These data suggested that AGEs may bind to RAGE on the EPC membrane to trigger differentiation into osteoblasts. The underlying mechanism appears to involve the p38MAPK and JNK1/2 pathways, but not the ERK1/2 pathway.


Asunto(s)
Antígenos de Neoplasias/farmacología , Células Progenitoras Endoteliales/efectos de los fármacos , Productos Finales de Glicación Avanzada/farmacología , Proteínas Quinasas Activadas por Mitógenos/farmacología , Osteogénesis/genética , Animales , Antígenos de Neoplasias/metabolismo , Médula Ósea , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/fisiología , Productos Finales de Glicación Avanzada/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/fisiología , Ratas , Ratas Sprague-Dawley/metabolismo
10.
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
11.
Thromb Haemost ; 122(3): 456-469, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34214997

RESUMEN

BACKGROUND: Smooth muscle cells (SMCs) are the main driver of neointima formation and restenosis following vascular injury. In animal models, endothelial progenitor cells (EPCs) accelerate endothelial regeneration and reduce neointima formation after arterial injury; however, EPC-capture stents do not reduce target vessel failure compared with conventional stents. Here we examined the influence of EPCs on features of SMCs pivotal for their impact on injury-induced neointima formation including proliferation, migration, and phenotype switch. METHODS AND RESULTS: EPCs, their conditioned medium, and EPC-derived microparticles induced proliferation of SMCs while limiting their apoptosis. In transwell membrane experiments and scratch assays, EPCs stimulated migration of SMCs and accelerated their recovery from scratch-induced injury. Treatment of SMCs with an EPC-derived conditioned medium or microparticles triggered transformation of SMCs toward a synthetic phenotype. However, co-cultivation of EPCs and SMCs enabling direct cell-cell contacts preserved their original phenotype and protected from the transformative effect of SMC cholesterol loading. Adhesion of EPCs to SMCs was stimulated by SMC injury and reduced by blocking CXCR2 and CCR5. Interaction of EPCs with SMCs modulated their secretory products and synergistically increased the release of selected chemokines. Following carotid wire injury in athymic mice, injection of EPCs resulted not only in reduced neointima formation but also in altered cellular composition of the neointima with augmented accumulation of SMCs. CONCLUSION: EPCs stimulate proliferation and migration of SMCs and increase their neointimal accumulation following vascular injury. Furthermore, EPCs context-dependently modify the SMC phenotype with protection from the transformative effect of cholesterol when a direct cell-cell contact is established.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Progenitoras Endoteliales , Neointima , Receptores de Interleucina-8B/metabolismo , Regeneración/fisiología , Lesiones del Sistema Vascular , Adaptación Fisiológica/fisiología , Animales , Apoptosis , Arterias/lesiones , Arterias/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/patología , Células Progenitoras Endoteliales/fisiología , Ratones , Miocitos del Músculo Liso , Neointima/etiología , Neointima/metabolismo , Neointima/patología , Neointima/prevención & control , Receptores CCR5/metabolismo , Transducción de Señal/fisiología , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
12.
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
13.
Mol Med Rep ; 25(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34738620

RESUMEN

Chronic hypertension can lead to kidney damage, known as hypertensive nephropathy or hypertensive nephrosclerosis. Further understanding of the molecular mechanisms via which hypertensive nephropathy develops is essential for effective diagnosis and treatment. The present study investigated the mechanisms by which endothelial progenitor cells (EPCs) repair primary rat kidney cells (PRKs). ELISA, Cell Counting Kit­8 and flow cytometry assays were used to analyze the effects of EPCs or EPC­MVs on the oxidative stress, inflammation, cell proliferation, apoptosis and cycle of PRKs induced by AngII. A PRK injury model was established using angiotensin II (Ang II). After Ang II induction, PRK proliferation was decreased, apoptosis was increased and the cell cycle was blocked at the G1 phase before entering the S phase. It was found that the levels of reactive oxygen species and malondialdehyde were increased, while the levels of glutathione peroxidase and superoxide dismutase were decreased. Moreover, the levels of the inflammatory cytokines IL­1ß, IL­6 and TNF­α were significantly increased. Thus, Ang II damaged PRKs by stimulating oxidative stress and promoting the inflammatory response. However, when PRKs were co­cultured with EPCs, the damage induced by Ang II was significantly reduced. The current study collected the microvesicles (MVs) secreted by EPCs and co­cultured them with Ang II­induced PRKs, and identified that EPC­MVs retained their protective effect on PRKs. In conclusion, EPCs protect PRKs from Ang II­induced damage via secreted MVs.


Asunto(s)
Micropartículas Derivadas de Células/fisiología , Células Progenitoras Endoteliales/metabolismo , Riñón/lesiones , Angiotensina II/efectos adversos , Angiotensina II/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Micropartículas Derivadas de Células/metabolismo , Citocinas/metabolismo , Células Progenitoras Endoteliales/fisiología , Hipertensión Renal/metabolismo , Hipertensión Renal/fisiopatología , Riñón/metabolismo , Masculino , Nefritis/metabolismo , Nefritis/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Cultivo Primario de Células , Ratas , Ratas Endogámicas WKY , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
14.
Mol Med Rep ; 25(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34738629

RESUMEN

Modulating the biological status of endothelial progenitor cells (EPCs), such as function and survival, is essential for therapeutic angiogenesis in ischemic vascular disease environments. This study aimed to explore the role and molecular mechanisms underlying Netrin­1 in the viability and angiogenic function of EPCs. EPCs were isolated from the bone barrow of adult C57/BL6 mice. The apoptosis and various functions of EPCs were analyzed in vitro by manipulating the expression of Netrin­1. The TUNEL assay was performed to detect apoptotic EPCs. Cell migration and tube formation assays were performed to detect EPC function. Trypan blue staining was performed to detect cell viability. Western blot analysis was performed to detect the protein expression levels of Netrin­1, CD146 and apoptotic factors. Quantitative PCR analysis was performed to detect the expression levels of Netrin­1 receptors. The results demonstrated that treatment with exogenous Netrin­1 promoted EPC migration and tube formation, whereas transfection with small interfering (si)RNA targeting Netrin­1 exhibited the opposite effects. Exogenous Netrin­1 protected EPCs from hypoxia­induced apoptosis, whereas the interruption of endogenous Netrin­1 enhancement under hypoxia by Netrin­1­siRNA exacerbated the apoptosis of EPCs. Furthermore, CD146, one of the immunoglobulin receptors activated by Netrin­1, was screened for in the present study. Results demonstrated that CD146 did not participate in Netrin­1­promoted EPC function, but mediated the anti­apoptotic effects of Netrin­1 in EPCs. In conclusion, Netrin­1 enhanced the angiogenic function of EPCs and alleviated hypoxia­induced apoptosis, which was mediated by CD146. This biological function of Netrin­1 may provide a potential therapeutic option to promote EPCs for the treatment of ischemic vascular diseases.


Asunto(s)
Apoptosis/fisiología , Netrina-1/metabolismo , Animales , Antígeno CD146/metabolismo , Antígeno CD146/fisiología , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Células Progenitoras Endoteliales/fisiología , Expresión Génica/genética , Hipoxia/metabolismo , Isquemia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Netrina-1/fisiología , Transducción de Señal/efectos de los fármacos
15.
Physiol Res ; 70(Suppl 1): S13-S20, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34918525

RESUMEN

Heart remodeling occurs as a compensation mechanism for the massive loss of tissue during initial heart failure and the consequent inflammation process. During heart remodeling fibroblasts differentiate to myofibroblasts activate their secretion functions and produce elevated amounts, of extracellular matrix (ECM) proteins, mostly collagen, that form scar tissue and alter the normal degradation of ECM. Scar formation does replace the damaged tissue structurally; however, it impedes the normal contractive function of cardiomyocytes (CMs) and results in long-lasting effects after heart failure. Besides CMs and cardiac fibroblasts, endothelial cells (ECs) and circulating endothelial progenitor cells (cEPCs) contribute to heart repair. This review summarizes the current knowledge of EC-CM crosstalk in cardiac fibrosis (CF), the role of cEPCs in heart regeneration and the contribution of Endothelial-mesenchymal transition (EndoMT).


Asunto(s)
Transdiferenciación Celular , Células Endoteliales/fisiología , Corazón/fisiología , Miocitos Cardíacos/fisiología , Regeneración , Remodelación Ventricular , Células Progenitoras Endoteliales/fisiología , Humanos , Receptor Cross-Talk
16.
Int J Mol Sci ; 22(23)2021 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-34884679

RESUMEN

The mobilization of endothelial progenitor cells (EPCs) into circulation from bone marrow is well known to be present in several clinical settings, including acute coronary syndrome, heart failure, diabetes and peripheral vascular disease. The aim of this review was to explore the current literature focusing on the great opportunity that EPCs can have in terms of regenerative medicine.


Asunto(s)
Células Progenitoras Endoteliales/fisiología , Animales , Enfermedades Cardiovasculares/fisiopatología , Separación Celular , Humanos
17.
BMC Cardiovasc Disord ; 21(1): 524, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34724901

RESUMEN

BACKGROUND: To explore the effects of cardiac exercise rehabilitation on peripheral blood endothelial progenitor cells (EPC) in elderly patients with chronic heart failure. METHODS: 80 elderly patients with chronic heart failure were selected from March 2017 to March 2019 and randomly divided into two groups (N = 40). The control group was treated routinely and walked freely for 30-60 min every day. The patients in the exercise rehabilitation group developed a cardiac exercise rehabilitation plan. Then, cardiac function and peripheral blood B-natriuretic peptide (BNP) levels in the two groups were compared. The cell viability, proliferation, apoptosis, and invasion ability of EPCs were detected. The levels of the PI3K/AKT pathway and eNOS and VEGF were compared. RESULTS: There were no significant differences in all indexes between the two groups before treatment (P > 0.05), and both improved significantly after treatment (P < 0.05). After treatment, LVEF and LVFS in the exercise rehabilitation group were significantly higher than those in the control group (P < 0.05), and LVEDD and LVESD were significantly lower than those in the control group (P < 0.05). The BNP level in the exercise rehabilitation group was significantly lower than that in the control group (P < 0.05). The cell viability, proliferation, invasion ability of EPC, and the levels of PI3K, AKT, eNOS, and VEGF mRNA and protein in the exercise rehabilitation group were significantly higher than those in the control group. Apoptosis rate was significantly lower than those in the control group (P < 0.05). CONCLUSIONS: Visceral exercise rehabilitation can improve cardiac ejection and myocardial function in elderly patients with chronic heart failure, and can promote the vitality, proliferation, and invasion of peripheral blood EPC, and promote the expression of eNOS and VEGF by upregulating the PI3K/AKT pathway to promote angiogenesis and endothelial function.


Asunto(s)
Rehabilitación Cardiaca , Células Progenitoras Endoteliales/fisiología , Insuficiencia Cardíaca/rehabilitación , Péptido Natriurético Encefálico/análisis , Anciano , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Endotelio Vascular/fisiopatología , Terapia por Ejercicio , Femenino , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Óxido Nítrico Sintasa de Tipo III/análisis , Óxido Nítrico Sintasa de Tipo III/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/fisiología , Volumen Sistólico , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Pharmacol Res ; 173: 105920, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34601081

RESUMEN

Vascular damage of hypertension has been the focus of hypertension treatment, and endothelial progenitor cells (EPCs) play an important role in the repair of vascular endothelial damage. Functional damage and decreased number of EPCs are observed in the peripheral circulation of hypertensive patients, but its mechanism is not yet elucidated. Here, we show that the number of EPCs in hypertensive patients is significantly lower than that of normal population, and the cell function decreases with a higher proportion of EPCs at later stages. A decrease in autophagy is responsible for the senescence and damage of EPCs induced by AngII. Moreover, lncRNA-p21 plays a critical regulator role in EPCs' senescence and dysfunction. Furthermore, lncRNA-p21 activates SESN2/AMPK/TSC2 pathway by promoting the transcriptional activity of p53 and enhances autophagy to protect against AngII-induced EPC damage. The data provide evidence that a reversal of decreased autophagy serves as the protective mechanism of EPC injury in hypertensive patients, and lncRNA-p21 is a new therapeutic target for vascular endothelial repair in hypertension.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Progenitoras Endoteliales/patología , Hipertensión , Proteínas Nucleares/metabolismo , ARN Largo no Codificante , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Anciano , Angiotensina II , Animales , Autofagia , Adhesión Celular , Movimiento Celular , Células Progenitoras Endoteliales/fisiología , Femenino , Humanos , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/patología , Masculino , Persona de Mediana Edad , Ratas Endogámicas SHR , Ratas Wistar , beta-Galactosidasa/metabolismo
19.
Plast Reconstr Surg ; 148(6): 936e-945e, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34644264

RESUMEN

BACKGROUND: Endothelial progenitor cells have shown the ability to enhance neovascularization. In this study, the authors tested whether intraosseous delivery of simvastatin could mobilize endothelial progenitor cells and enhance recovery in a hindlimb ischemia model. METHODS: There are eight groups of rats in this study: normal control; type 1 diabetes mellitus control group control without drug intervention; and type 1 diabetes mellitus rats that randomly received intraosseous simvastatin (0, 0.5, or 1 mg) or oral simvastatin administration (0, 20, or 400 mg). All type 1 diabetes mellitus rats had induced hindlimb ischemia. The number of endothelial progenitor cells in peripheral blood, and serum markers, were detected. The recovery of blood flow at 21 days after treatment was used as the main outcome. RESULTS: The authors demonstrated that endothelial progenitor cell mobilization was increased in the simvastatin 0.5- and 1-mg groups compared with the type 1 diabetes mellitus control and simvastatin 0-mg groups at 1, 2, and 3 weeks. Serum vascular endothelial growth factor levels were significantly increased at 2 weeks in the simvastatin 0.5- and 1-mg groups, in addition to the increase of the blood flow and the gastrocnemius weight at 3 weeks. Similar increase can also been seen in simvastatin 400 mg orally but not in simvastatin 20 mg orally. CONCLUSION: These findings demonstrate that a single intraosseous administration of simvastatin mobilized endothelial progenitor cells at a dose one-hundredth of the required daily oral dose in rats, and this potent mobilization of endothelial progenitor cells markedly improved diabetic limb ischemia by means of neovascularization.


Asunto(s)
Isquemia Crónica que Amenaza las Extremidades/tratamiento farmacológico , Diabetes Mellitus Tipo 1/complicaciones , Células Progenitoras Endoteliales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Simvastatina/administración & dosificación , Animales , Isquemia Crónica que Amenaza las Extremidades/etiología , Circulación Colateral/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/inducido químicamente , Células Progenitoras Endoteliales/fisiología , Miembro Posterior/irrigación sanguínea , Humanos , Infusiones Intraóseas , Masculino , Ratas , Estreptozocina/administración & dosificación , Estreptozocina/toxicidad
20.
Theranostics ; 11(17): 8430-8447, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34373751

RESUMEN

Self-assembly of solid organs from single cells would greatly expand applicability of regenerative medicine. Stem/progenitor cells can self-organize into micro-sized organ units, termed organoids, partially modelling tissue function and regeneration. Here we demonstrated 3D self-assembly of adult and induced pluripotent stem cell (iPSC)-derived fibroblasts, keratinocytes and endothelial progenitors into both, planar human skin in vivo and a novel type of spheroid-shaped skin organoids in vitro, under the aegis of human platelet lysate. Methods: Primary endothelial colony forming cells (ECFCs), skin fibroblasts (FBs) and keratinocytes (KCs) were isolated from human tissues and polyclonally propagated under 2D xeno-free conditions. Human tissue-derived iPSCs were differentiated into endothelial cells (hiPSC-ECs), fibroblasts (hiPSC-FBs) and keratinocytes (hiPSC-KCs) according to efficiency-optimized protocols. Cell identity and purity were confirmed by flow cytometry and clonogenicity indicated their stem/progenitor potential. Triple cell type floating spheroids formation was promoted by human platelet-derived growth factors containing culture conditions, using nanoparticle cell labelling for monitoring the organization process. Planar human skin regeneration was assessed in full-thickness wounds of immune-deficient mice upon transplantation of hiPSC-derived single cell suspensions. Results: Organoids displayed a distinct architecture with surface-anchored keratinocytes surrounding a stromal core, and specific signaling patterns in response to inflammatory stimuli. FGF-7 mRNA transfection was required to accelerate keratinocyte long-term fitness. Stratified human skin also self-assembled within two weeks after either adult- or iPSC-derived skin cell-suspension liquid-transplantation, healing deep wounds of mice. Transplant vascularization significantly accelerated in the presence of co-transplanted endothelial progenitors. Mechanistically, extracellular vesicles mediated the multifactorial platelet-derived trophic effects. No tumorigenesis occurred upon xenografting. Conclusion: This illustrates the superordinate progenitor self-organization principle and permits novel rapid 3D skin-related pharmaceutical high-content testing opportunities with floating spheroid skin organoids. Multi-cell transplant self-organization facilitates development of iPSC-based organ regeneration strategies using cell suspension transplantation supported by human platelet factors.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Organoides/metabolismo , Fenómenos Fisiológicos de la Piel/genética , Células Madre/metabolismo , Adulto , Animales , Diferenciación Celular/fisiología , Células Endoteliales/citología , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/fisiología , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Voluntarios Sanos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Queratinocitos/citología , Queratinocitos/fisiología , Masculino , Ratones Endogámicos NOD , Persona de Mediana Edad , Organoides/citología , Regeneración/fisiología , Medicina Regenerativa , Piel/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA