Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 649
Filter
1.
Int J Mol Sci ; 25(18)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39337516

ABSTRACT

Adult stem cell therapy via intramyocardial injection of autologous CD34+ stem cells has been shown to improve exercise capacity and reduce angina frequency and mortality in patients with refractory angina (RA). However, the cost of such therapy is a limitation to its adoption in clinical practice. Our goal was to determine whether the less costly, less invasive, and widely accessible, FDA-approved alternative treatment for RA patients, known as enhanced external counterpulsation (EECP), mobilizes endogenous CD34+ stem cells and whether such mobilization is associated with the clinical benefits seen with intramyocardial injection. We monitored changes in circulating levels of CD34+/CD133+ and CD34+/KDR+ cells in RA patients undergoing EECP therapy and in a comparator cohort of RA patients undergoing an exercise regimen known as cardiac rehabilitation. Changes in exercise capacity in both cohorts were monitored by measuring treadmill times (TT), double product (DP) scores, and Canadian Cardiovascular Society (CCS) angina scores between pre- and post-treatment treadmill stress tests. Circulating levels of CD34+/CD133+ cells increased in patients undergoing EECP and were significant (ß = -2.38, p = 0.012) predictors of improved exercise capacity in these patients. CD34+/CD133+ cells isolated from RA patients could differentiate into endothelial cells, and their numbers increased during EECP therapy. Our results support the hypothesis that mobilized CD34+/CD133+ cells repair vascular damage and increase collateral circulation in RA patients. They further support clinical interventions that can mobilize adult CD34+ stem cells as therapy for patients with RA and other vascular diseases.


Subject(s)
AC133 Antigen , Angina Pectoris , Antigens, CD34 , Counterpulsation , Endothelial Progenitor Cells , Humans , AC133 Antigen/metabolism , Antigens, CD34/metabolism , Female , Male , Angina Pectoris/therapy , Angina Pectoris/blood , Angina Pectoris/metabolism , Middle Aged , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Aged , Counterpulsation/methods , Hematopoietic Stem Cell Mobilization/methods
2.
Front Endocrinol (Lausanne) ; 15: 1396794, 2024.
Article in English | MEDLINE | ID: mdl-39076517

ABSTRACT

Diabetes mellitus (DM) is a metabolic disease characterized by hyperglycemia, leading to various vascular complications. Accumulating evidence indicates that endothelial colony-forming cells (ECFCs) have attractive prospects for repairing and restoring blood vessels. Thus, ECFCs may be a novel therapeutic option for diabetic patients with vascular complications who require revascularization therapy. However, it has been reported that the function of ECFCs is impaired in DM, which poses challenges for the autologous transplantation of ECFCs. In this review, we summarize the molecular mechanisms that may be responsible for ECFC dysfunction and discuss potential strategies for improving the therapeutic efficacy of ECFCs derived from patients with DM. Finally, we discuss barriers to the use of ECFCs in human studies in light of the fact that there are no published reports using these cells in humans.


Subject(s)
Diabetic Angiopathies , Humans , Diabetic Angiopathies/therapy , Animals , Endothelial Progenitor Cells/transplantation , Endothelial Progenitor Cells/cytology , Endothelial Cells/transplantation , Endothelial Cells/cytology , Stem Cell Transplantation/methods
3.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892272

ABSTRACT

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.


Subject(s)
Endothelial Progenitor Cells , Exercise , Humans , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Exercise/physiology , Animals
4.
J Cell Mol Med ; 28(12): e18489, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38899522

ABSTRACT

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.


Subject(s)
Cellular Senescence , Chemokine CCL5 , Endothelial Progenitor Cells , MicroRNAs , Neovascularization, Physiologic , Animals , Humans , Male , Mice , Angiogenesis , Cell Proliferation , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , Down-Regulation/genetics , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Ischemia/metabolism , Ischemia/pathology , Ischemia/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Physiologic/genetics , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics
5.
Stem Cell Res Ther ; 15(1): 164, 2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38853275

ABSTRACT

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.


Subject(s)
Antigens, CD34 , Liver , Animals , Humans , Antigens, CD34/metabolism , Mice , Liver/metabolism , Liver/pathology , Mice, Inbred NOD , Hematopoietic Stem Cell Transplantation , Mice, SCID , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/transplantation , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Fetal Blood/cytology , Melanoma/pathology , Melanoma/immunology
6.
Angiogenesis ; 27(3): 545-560, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38733496

ABSTRACT

Regenerative capabilities of the endothelium rely on vessel-resident progenitors termed endothelial colony forming cells (ECFCs). This study aimed to investigate if these progenitors are impacted by conditions (i.e., obesity or atherosclerosis) characterized by increased serum levels of oxidized low-density lipoprotein (oxLDL), a known inducer of Endothelial-to-Mesenchymal Transition (EndMT). Our investigation focused on understanding the effects of EndMT on the self-renewal capabilities of progenitors and the associated molecular alterations. In the presence of oxLDL, ECFCs displayed classical features of EndMT, through reduced endothelial gene and protein expression, function as well as increased mesenchymal genes, contractility, and motility. Additionally, ECFCs displayed a dramatic loss in self-renewal capacity in the presence of oxLDL. RNA-sequencing analysis of ECFCs exposed to oxLDL validated gene expression changes suggesting EndMT and identified SOX9 as one of the highly differentially expressed genes. ATAC sequencing analysis identified SOX9 binding sites associated with regions of dynamic chromosome accessibility resulting from oxLDL exposure, further pointing to its importance. EndMT phenotype and gene expression changes induced by oxLDL in vitro or high fat diet (HFD) in vivo were reversed by the silencing of SOX9 in ECFCs or the endothelial-specific conditional knockout of Sox9 in murine models. Overall, our findings support that EndMT affects vessel-resident endothelial progenitor's self-renewal. SOX9 activation is an early transcriptional event that drives the mesenchymal transition of endothelial progenitor cells. The identification of the molecular network driving EndMT in vessel-resident endothelial progenitors presents a new avenue in understanding and preventing a range of condition where this process is involved.


Subject(s)
Lipoproteins, LDL , SOX9 Transcription Factor , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Animals , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Mice , Humans , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Epithelial-Mesenchymal Transition , Mice, Inbred C57BL , Male , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Cell Self Renewal , Endothelial Cells/metabolism
7.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732080

ABSTRACT

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.


Subject(s)
Cell Movement , Dihydrotestosterone , Endothelial Progenitor Cells , Fetal Blood , Receptors, Androgen , Fetal Blood/cytology , Dihydrotestosterone/pharmacology , Humans , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Cell Proliferation , Cell Survival , Gene Expression , Vascular Endothelial Growth Factor Receptor-2/genetics , Membrane Proteins/genetics , Matrix Metalloproteinase 9/genetics , Basigin/genetics , Animals , Mice , Heart Ventricles/cytology , Cell Movement/drug effects
8.
Cell Transplant ; 33: 9636897241253144, 2024.
Article in English | MEDLINE | ID: mdl-38798036

ABSTRACT

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.


Subject(s)
Biofilms , Endothelial Progenitor Cells , Myocardial Ischemia , Swine, Miniature , Animals , Swine , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Myocardial Ischemia/therapy , Myocardial Ischemia/complications , Extracorporeal Shockwave Therapy/methods , Myocardium/metabolism , Myocardium/pathology , Male
9.
Dev Cell ; 59(14): 1842-1859.e12, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38663400

ABSTRACT

Placental ischemia, resulting from inadequate remodeling of uterine spiral arteries, is a factor in the development of preeclampsia. However, the effect of endothelial progenitor cells that play a role in the vascular injury-repair program is largely unexplored during remodeling. Here, we observe that preeclampsia-afflicted uterine spiral arteries transition to a synthetic phenotype in vascular smooth muscle cells and characterize the regulatory axis in endothelial progenitor cells during remodeling in human decidua basalis. Excessive sEng, secreted by AMP-activated protein kinase (AMPK)-deficient endothelial progenitor cells through the inhibition of HO-1, damages residual endothelium and leads to the accumulation of extracellular matrix produced by vascular smooth muscle cells during remodeling, which is further confirmed by animal models. Collectively, our findings suggest that the impaired functionality of endothelial progenitor cells contributes to the narrowing of remodeled uterine spiral arteries, leading to reduced utero-placental perfusion. This mechanism holds promise in elucidating the pathogenesis of preeclampsia.


Subject(s)
Endothelial Progenitor Cells , Placenta , Placental Circulation , Pre-Eclampsia , Uterus , Vascular Remodeling , Female , Pregnancy , Humans , Animals , Pre-Eclampsia/pathology , Pre-Eclampsia/metabolism , Placenta/blood supply , Placenta/metabolism , Uterus/blood supply , Uterus/metabolism , Vascular Remodeling/physiology , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/cytology , Mice , Uterine Artery/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology
10.
Adv Healthc Mater ; 13(14): e2302830, 2024 06.
Article in English | MEDLINE | ID: mdl-38366136

ABSTRACT

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.


Subject(s)
Bioprinting , Endothelial Progenitor Cells , Tissue Engineering , Tissue Scaffolds , Humans , Bioprinting/methods , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Hydrogels/chemistry , Capillaries/physiology , Alginates/chemistry , Printing, Three-Dimensional
11.
Oxid Med Cell Longev ; 2022: 1282362, 2022.
Article in English | MEDLINE | ID: mdl-35832490

ABSTRACT

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.


Subject(s)
Endothelial Progenitor Cells , Sirtuin 3 , Toll-Like Receptor 4 , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Homeostasis , Humans , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Oxidation-Reduction , Sirtuin 3/metabolism , Toll-Like Receptor 4/metabolism
12.
Int J Mol Sci ; 23(3)2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35163466

ABSTRACT

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.


Subject(s)
Acute Kidney Injury/prevention & control , Endothelial Progenitor Cells/transplantation , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/prevention & control , Acute Kidney Injury/metabolism , Animals , Apoptosis/drug effects , Blood Urea Nitrogen , Cells, Cultured , Creatinine/blood , Disease Models, Animal , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/immunology , Endothelial Progenitor Cells/metabolism , Humans , Male , Mice , Oxidative Stress/drug effects , Reperfusion Injury/immunology , Reperfusion Injury/metabolism
13.
Sci Rep ; 12(1): 1775, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35110646

ABSTRACT

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.


Subject(s)
Adipose Tissue/cytology , Biomarkers/metabolism , Endothelial Progenitor Cells/cytology , Regenerative Medicine , Stromal Cells/cytology , Adipose Tissue/metabolism , Adult , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Female , Humans , Male , Middle Aged , Stromal Cells/metabolism
14.
PLoS One ; 17(1): e0261498, 2022.
Article in English | MEDLINE | ID: mdl-35025920

ABSTRACT

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.


Subject(s)
Endothelial Progenitor Cells/transplantation , Graft Survival/physiology , Neovascularization, Physiologic/physiology , Stromal Cells/transplantation , Adipose Tissue/cytology , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Cell Culture Techniques , Cell Movement , Cells, Cultured , Coculture Techniques , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Mice , Mice, Nude , Paracrine Communication/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rabbits , Stromal Cells/cytology , Stromal Cells/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
Int J Mol Sci ; 23(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35008953

ABSTRACT

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.


Subject(s)
Biopolymers/chemistry , Caproates/chemistry , Endothelial Progenitor Cells/cytology , Heart Valves , Induced Pluripotent Stem Cells/cytology , Lactones/chemistry , Mesenchymal Stem Cells/cytology , Tissue Engineering , Tissue Scaffolds , Animals , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Extracellular Matrix , Humans , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Nanofibers , Swine , Tissue Scaffolds/chemistry
16.
Food Funct ; 13(1): 76-90, 2022 Jan 04.
Article in English | MEDLINE | ID: mdl-34882161

ABSTRACT

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.


Subject(s)
Diet, Healthy , Endothelial Progenitor Cells , Fruit , Heart Disease Risk Factors , Lycium , Blood Pressure/physiology , Cell Movement/physiology , Cells, Cultured , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/physiology , Female , Humans , Lipids/blood , Male , Middle Aged
17.
Exp Cell Res ; 411(1): 112985, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34942190

ABSTRACT

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.


Subject(s)
Endothelial Progenitor Cells/physiology , Gene Expression Regulation , MicroRNAs/genetics , Neovascularization, Physiologic , Repressor Proteins/antagonists & inhibitors , Thrombolytic Therapy/methods , Venous Thrombosis/therapy , Adult , Animals , Apoptosis , Biomarkers/metabolism , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Progenitor Cells/cytology , Female , Humans , Male , Prognosis , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcriptional Activation , Up-Regulation , Venous Thrombosis/metabolism , Venous Thrombosis/pathology
18.
Dev Biol ; 482: 44-54, 2022 02.
Article in English | MEDLINE | ID: mdl-34915023

ABSTRACT

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.


Subject(s)
Endothelial Cells/cytology , Endothelial Progenitor Cells/cytology , Lymphangiogenesis/genetics , Lymphangiogenesis/physiology , Lymphatic Vessels/embryology , Animals , Embryo, Mammalian/embryology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
19.
J Diabetes Res ; 2021: 9962877, 2021.
Article in English | MEDLINE | ID: mdl-34722777

ABSTRACT

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.


Subject(s)
Cell Movement , Diabetes Mellitus/metabolism , Endothelial Progenitor Cells/transplantation , Neovascularization, Physiologic , Skin/injuries , Wound Healing , Animals , Disease Models, Animal , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Mice , Skin/metabolism , Skin/pathology , Transplantation, Homologous
20.
Eur Rev Med Pharmacol Sci ; 25(19): 6101-6108, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34661270

ABSTRACT

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.


Subject(s)
Altitude , Coronary Disease/etiology , Cytokines/metabolism , Endothelial Progenitor Cells/cytology , Adult , Aged , C-Reactive Protein/metabolism , Case-Control Studies , Coronary Disease/epidemiology , Female , Humans , Male , Middle Aged , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL