Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.515
Filter
1.
Sci Rep ; 14(1): 18469, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122748

ABSTRACT

Endothelial progenitor cells (EPCs) play a crucial role in maintaining vascular health and aiding in the repair of damaged blood vessels. However, the specific impact of EPCs-derived exosomes on vascular endothelial cell injury caused by lipopolysaccharide (LPS) remains inadequately understood. This study aims to explore the potential benefits of EPC-exosomes in mitigating LPS-induced vascular injury and to elucidate the underlying mechanism. Initially, EPCs were isolated from mouse peripheral blood, and their identity was confirmed through flow cytometry and immunocytochemistry. Subsequently, the exosomes derived from EPCs were identified using transmission electron microscopy (TEM) and western blot analysis. A sepsis model was induced by subjecting brain microvascular endothelial cells (BMECs) to LPS-induced injury. Both EPC and their exosomes demonstrated a significant increase in BMECs proliferation, reduced apoptosis, decreased levels of pro-inflammatory factors (TNF-α, IL-6, and caspase-3), and enhanced sprouting and angiogenesis of BMECs. Notable, the Exosomes demonstrated a more pronounced impact on these parameters. Furthermore, both EPCs and Exosomes exhibited significantly increased levels of miR-126a-5p, with the Exosomes showing a more substantial enhancement. These findings suggest that supplementing exosomal miR-126a-5p from EPCs can provide protective effects on BMECs, offering a potential therapeutic option for treating sepsis-induced microvascular endothelial cell injury.


Subject(s)
Brain , Endothelial Cells , Endothelial Progenitor Cells , Exosomes , Lipopolysaccharides , MicroRNAs , Exosomes/metabolism , Animals , Endothelial Progenitor Cells/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Lipopolysaccharides/toxicity , Mice , Brain/metabolism , Brain/pathology , Endothelial Cells/metabolism , Apoptosis , Cell Proliferation , Microvessels/metabolism , Male , Sepsis/metabolism , Mice, Inbred C57BL
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.
J Cell Mol Med ; 28(13): e18523, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38957039

ABSTRACT

This research explores the role of microRNA in senescence of human endothelial progenitor cells (EPCs) induced by replication. Hsa-miR-134-5p was found up-regulated in senescent EPCs where overexpression improved angiogenic activity. Hsa-miR-134-5p, which targeted transforming growth factor ß-activated kinase 1-binding protein 1 (TAB1) gene, down-regulated TAB1 protein, and inhibited phosphorylation of p38 mitogen-activated protein kinase (p38) in hsa-miR-134-5p-overexpressed senescent EPCs. Treatment with siRNA specific to TAB1 (TAB1si) down-regulated TAB1 protein and subsequently inhibited p38 activation in senescent EPCs. Treatment with TAB1si and p38 inhibitor, respectively, showed angiogenic improvement. In parallel, transforming growth factor Beta 1 (TGF-ß1) was down-regulated in hsa-miR-134-5p-overexpressed senescent EPCs and addition of TGF-ß1 suppressed the angiogenic improvement. Analysis of peripheral blood mononuclear cells (PBMCs) disclosed expression levels of hsa-miR-134-5p altered in adult life, reaching a peak before 65 years, and then falling in advanced age. Calculation of the Framingham risk score showed the score inversely correlates with the hsa-miR-134-5p expression level. In summary, hsa-miR-134-5p is involved in the regulation of senescence-related change of angiogenic activity via TAB1-p38 signalling and via TGF-ß1 reduction. Hsa-miR-134-5p has a potential cellular rejuvenation effect in human senescent EPCs. Detection of human PBMC-derived hsa-miR-134-5p predicts cardiovascular risk.


Subject(s)
Adaptor Proteins, Signal Transducing , Cardiovascular Diseases , Cellular Senescence , Endothelial Progenitor Cells , Leukocytes, Mononuclear , MicroRNAs , p38 Mitogen-Activated Protein Kinases , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Endothelial Progenitor Cells/metabolism , Cellular Senescence/genetics , Leukocytes, Mononuclear/metabolism , Middle Aged , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Male , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , Female , Aged , Neovascularization, Physiologic/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Adult , Risk Factors
4.
Circ Res ; 135(4): e94-e113, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-38957991

ABSTRACT

BACKGROUND: Cerebral vascular malformations (CCMs) are primarily found within the brain, where they result in increased risk for stroke, seizures, and focal neurological deficits. The unique feature of the brain vasculature is the blood-brain barrier formed by the brain neurovascular unit. Recent studies suggest that loss of CCM genes causes disruptions of blood-brain barrier integrity as the inciting events for CCM development. CCM lesions are proposed to be initially derived from a single clonal expansion of a subset of angiogenic venous capillary endothelial cells (ECs) and respective resident endothelial progenitor cells (EPCs). However, the critical signaling events in the subclass of brain ECs/EPCs for CCM lesion initiation and progression are unclear. METHODS: Brain EC-specific CCM3-deficient (Pdcd10BECKO) mice were generated by crossing Pdcd10fl/fl mice with Mfsd2a-CreERT2 mice. Single-cell RNA-sequencing analyses were performed by the chromium single-cell platform (10× genomics). Cell clusters were annotated into EC subtypes based on visual inspection and GO analyses. Cerebral vessels were visualized by 2-photon in vivo imaging and tissue immunofluorescence analyses. Regulation of mTOR (mechanistic target of rapamycin) signaling by CCM3 and Cav1 (caveolin-1) was performed by cell biology and biochemical approaches. RESULTS: Single-cell RNA-sequencing analyses from P10 Pdcd10BECKO mice harboring visible CCM lesions identified upregulated CCM lesion signature and mitotic EC clusters but decreased blood-brain barrier-associated EC clusters. However, a unique EPC cluster with high expression levels of stem cell markers enriched with mTOR signaling was identified from early stages of the P6 Pdcd10BECKO brain. Indeed, mTOR signaling was upregulated in both mouse and human CCM lesions. Genetic deficiency of Raptor (regulatory-associated protein of mTOR), but not of Rictor (rapamycin-insensitive companion of mTOR), prevented CCM lesion formation in the Pdcd10BECKO model. Importantly, the mTORC1 (mTOR complex 1) pharmacological inhibitor rapamycin suppressed EPC proliferation and ameliorated CCM pathogenesis in Pdcd10BECKO mice. Mechanistic studies suggested that Cav1/caveolae increased in CCM3-depleted EPC-mediated intracellular trafficking and complex formation of the mTORC1 signaling proteins. CONCLUSIONS: CCM3 is critical for maintaining blood-brain barrier integrity and CCM3 loss-induced mTORC1 signaling in brain EPCs initiates and facilitates CCM pathogenesis.


Subject(s)
Endothelial Progenitor Cells , Hemangioma, Cavernous, Central Nervous System , Mechanistic Target of Rapamycin Complex 1 , Signal Transduction , Animals , Hemangioma, Cavernous, Central Nervous System/metabolism , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/pathology , Mice , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Brain/metabolism , Brain/pathology , Brain/blood supply , Mice, Knockout , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Mice, Inbred C57BL , Membrane Proteins/metabolism , Membrane Proteins/genetics
5.
Biomaterials ; 311: 122699, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38981153

ABSTRACT

The treatment of osteoporotic bone defects poses a challenge due to the degradation of the skeletal vascular system and the disruption of local bone metabolism within the osteoporotic microenvironment. However, it is feasible to modulate the disrupted local bone metabolism imbalance through enhanced vascularization, a theory termed "vascularization-bone metabolic balance". This study developed a 3D-printed polycaprolactone (PCL) scaffold modified with EPLQLKM and SVVYGLR peptides (PCL-SE). The EPLQLKM peptide attracts bone marrow-derived mesenchymal stem cells (BMSCs), while the SVVYGLR peptide enhances endothelial progenitor cells (EPCs) vascular differentiation, thus regulating bone metabolism and fostering bone regeneration through the paracrine effects of EPCs. Further mechanistic research demonstrated that PCL-SE promoted the vascularization of EPCs, activating the Notch signaling pathway in BMSCs, leading to the upregulation of osteogenesis-related genes and the downregulation of osteoclast-related genes, thereby restoring bone metabolic balance. Furthermore, PCL-SE facilitated the differentiation of EPCs into "H"-type vessels and the recruitment of BMSCs to synergistically enhance osteogenesis, resulting in the regeneration of normal microvessels and bone tissues in cases of femoral condylar bone defects in osteoporotic SD rats. This study suggests that PCL-SE supports in-situ vascularization, remodels bone metabolic translational balance, and offers a promising therapeutic regimen for osteoporotic bone defects.


Subject(s)
Bone Regeneration , Homeostasis , Mesenchymal Stem Cells , Neovascularization, Physiologic , Osteogenesis , Osteoporosis , Printing, Three-Dimensional , Rats, Sprague-Dawley , Tissue Scaffolds , Animals , Bone Regeneration/drug effects , Osteoporosis/metabolism , Osteoporosis/therapy , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Tissue Scaffolds/chemistry , Osteogenesis/drug effects , Neovascularization, Physiologic/drug effects , Polyesters/chemistry , Cell Differentiation/drug effects , Female , Rats , Endothelial Progenitor Cells/metabolism , Bone and Bones/metabolism
6.
Nagoya J Med Sci ; 86(2): 292-303, 2024 May.
Article in English | MEDLINE | ID: mdl-38962405

ABSTRACT

Endothelial dysfunction is important in the pathology of pulmonary hypertension, and circulating endothelial progenitor cells (EPCs) have been studied to evaluate endothelial dysfunction. In patients with chronic thromboembolic pulmonary hypertension (CTEPH), riociguat reportedly increases the number of circulating EPCs. However, the relationship between EPC numbers at baseline and changes in clinical parameters after riociguat administration has not been fully elucidated. Here, we evaluated 27 treatment-naïve patients with CTEPH and analyzed the relationships between EPC number at diagnosis and clinical variables (age, hemodynamics, atrial blood gas parameters, brain natriuretic peptide, and exercise tolerance) before and after riociguat initiation. EPCs were defined as CD45dim CD34+ CD133+ cells and measured by flow cytometry. A low number of circulating EPCs at diagnosis was significantly correlated with increased reductions in mean pulmonary arterial pressure (mPAP) (correlation coefficient = 0.535, P = 0.004) and right atrial pressure (correlation coefficient = 0.618, P = 0.001) upon riociguat treatment. We then divided the study population into two groups according to the mPAP change: a weak-response group (a decrease in mPAP of 4 mmHg or less) and a strong-response group (a decrease in mPAP of more than 4 mmHg). The number of EPCs at diagnosis was significantly lower in the strong-response group than in the weak-response group (P = 0.022), but there were no significant differences in other clinical variables or in medication profiles. In conclusion, circulating EPC numbers could be a potential predictor of the therapeutic effect of riociguat in CTEPH patients.


Subject(s)
Endothelial Progenitor Cells , Hypertension, Pulmonary , Pyrazoles , Pyrimidines , Humans , Pyrimidines/therapeutic use , Pyrimidines/pharmacology , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Male , Female , Middle Aged , Hypertension, Pulmonary/drug therapy , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Aged , Chronic Disease , Pulmonary Embolism/drug therapy , Pulmonary Embolism/blood , Treatment Outcome
7.
Stem Cell Res Ther ; 15(1): 225, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075518

ABSTRACT

BACKGROUND: This study explores the potential role of Thioredoxin-interacting protein (TXNIP) silencing in endothelial colony-forming cells (ECFCs) within the scope of age-related comorbidities and impaired vascular repair. We aim to elucidate the effects of TXNIP silencing on vasculogenic properties, paracrine secretion, and neutrophil recruitment under conditions of metabolic stress. METHODS: ECFCs, isolated from human blood cord, were transfected with TXNIP siRNA and exposed to a high glucose and ß-hydroxybutyrate (BHB) medium to simulate metabolic stress. We evaluated the effects of TXNIP silencing on ECFCs' functional and secretory responses under these conditions. Assessments included analyses of gene and protein expression profiles, vasculogenic properties, cytokine secretion and neutrophil recruitment both in vitro and in vivo. The in vivo effects were examined using a murine model of hindlimb ischemia to observe the physiological relevance of TXNIP modulation under metabolic disorders. RESULTS: TXNIP silencing did not mitigate the adverse effects on cell recruitment, vasculogenic properties, or senescence induced by metabolic stress in ECFCs. However, it significantly reduced IL-8 secretion and consequent neutrophil recruitment under these conditions. In a mouse model of hindlimb ischemia, endothelial deletion of TXNIP reduced MIP-2 secretion and prevented increased neutrophil recruitment induced by age-related comorbidities. CONCLUSIONS: Our findings suggest that targeting TXNIP in ECFCs may alleviate ischemic complications exacerbated by metabolic stress, offering potential clinical benefits for patients suffering from age-related comorbidities.


Subject(s)
Carrier Proteins , Interleukin-8 , Neutrophil Infiltration , Stress, Physiological , Animals , Interleukin-8/metabolism , Interleukin-8/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Humans , Mice , Neutrophil Infiltration/drug effects , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/drug effects , Ischemia/metabolism , Ischemia/pathology , RNA, Small Interfering/metabolism , Thioredoxins/metabolism , Thioredoxins/genetics , Hindlimb/blood supply , Mice, Inbred C57BL , Glucose/metabolism
10.
Biomed Pharmacother ; 177: 117022, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38917756

ABSTRACT

BACKGROUND: The transplantation of endothelial progenitor cells (EPCs) has been shown to reduce neointimal hyperplasia following arterial injury. However, the efficacy of this approach is hampered by limited homing of EPCs to the injury site. Additionally, the in vivo recruitment and metabolic activity of transplanted EPCs have not been continuously monitored. METHODS: EPCs were labeled with indocyanine green (ICG)-conjugated superparamagnetic iron oxide nanoparticles (SPIONs) and subjected to external magnetic field targeting to enhance their delivery to a carotid balloon injury (BI) model in Sprague-Dawley rats. Magnetic particle imaging (MPI)/ fluorescence imaging (FLI) multimodal in vivo imaging, 3D MPI/CT imaging and MPI/FLI ex vivo imaging was performed after injury. Carotid arteries were collected and analyzed for pathology and immunofluorescence staining. The paracrine effects were analyzed by enzyme-linked immunosorbent assay. RESULTS: The application of a magnetic field significantly enhanced the localization and retention of SPIONs@PEG-ICG-EPCs at the site of arterial injury, as evidenced by both in vivo continuous monitoring and ex vivo by observation. This targeted delivery approach effectively inhibited neointimal hyperplasia and increased the presence of CD31-positive cells at the injury site. Moreover, serum levels of SDF-1α, VEGF, IGF-1, and TGF-ß1 were significantly elevated, indicating enhanced paracrine activity. CONCLUSIONS: Our findings demonstrate that external magnetic field-directed delivery of SPIONs@PEG-ICG-EPCs to areas of arterial injury can significantly enhance their therapeutic efficacy. This enhancement is likely mediated through increased paracrine signaling. These results underscore the potential of magnetically guided SPIONs@PEG-ICG-EPCs delivery as a promising strategy for treating arterial injuries.


Subject(s)
Carotid Artery Injuries , Endothelial Progenitor Cells , Hyperplasia , Magnetic Fields , Magnetic Iron Oxide Nanoparticles , Neointima , Rats, Sprague-Dawley , Animals , Endothelial Progenitor Cells/metabolism , Magnetic Iron Oxide Nanoparticles/chemistry , Neointima/pathology , Carotid Artery Injuries/pathology , Male , Rats
11.
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
12.
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
13.
Neuromolecular Med ; 26(1): 25, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886284

ABSTRACT

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.


Subject(s)
Endothelial Progenitor Cells , Intracranial Aneurysm , Humans , Intracranial Aneurysm/therapy , Endothelial Progenitor Cells/physiology , Endothelial Progenitor Cells/transplantation , Endovascular Procedures/methods , Cell Differentiation , Animals , Signal Transduction , Neovascularization, Physiologic , Embolization, Therapeutic , Neovascularization, Pathologic
14.
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
15.
Mol Med ; 30(1): 84, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867168

ABSTRACT

BACKGROUND: Deep vein thrombosis (DVT) is a common vascular surgical disease caused by the coagulation of blood in the deep veins, and predominantly occur in the lower limbs. Endothelial progenitor cells (EPCs) are multi-functional stem cells, which are precursors of vascular endothelial cells. EPCs have gradually evolved into a promising treatment strategy for promoting deep vein thrombus dissolution and recanalization through the stimulation of various physical and chemical factors. METHODS: In this study, we utilized a mouse DVT model and performed several experiments including qRT-PCR, Western blot, tube formation, wound healing, Transwell assay, immunofluorescence, flow cytometry analysis, and immunoprecipitation to investigate the role of HOXD9 in the function of EPCs cells. The therapeutic effect of EPCs overexpressing HOXD9 on the DVT model and its mechanism were also explored. RESULTS: Overexpression of HOXD9 significantly enhanced the angiogenesis and migration abilities of EPCs, while inhibiting cell apoptosis. Additionally, results indicated that HOXD9 specifically targeted the HRD1 promoter region and regulated the downstream PINK1-mediated mitophagy. Interestingly, intravenous injection of EPCs overexpressing HOXD9 into mice promoted thrombus dissolution and recanalization, significantly decreasing venous thrombosis. CONCLUSIONS: The findings of this study reveal that HOXD9 plays a pivotal role in stimulating vascular formation in endothelial progenitor cells, indicating its potential as a therapeutic target for DVT management.


Subject(s)
Disease Models, Animal , Endothelial Progenitor Cells , Homeodomain Proteins , Mitophagy , Neovascularization, Physiologic , Venous Thrombosis , Animals , Endothelial Progenitor Cells/metabolism , Mice , Venous Thrombosis/metabolism , Venous Thrombosis/genetics , Venous Thrombosis/therapy , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mitophagy/genetics , Neovascularization, Physiologic/genetics , Cell Movement , Male , Apoptosis , Humans , Angiogenesis
16.
Med Sci Monit ; 30: e945471, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864115

ABSTRACT

The Editors of Medical Science Monitor wish to inform you that the above manuscript has been retracted from publication due to concerns with the credibility and originality of the study, the manuscript content, and the Figure images. Reference: Rongfeng Zhang, Jianwei Liu, Shengpeng Yu, Dong Sun, Xiaohua Wang, Jingshu Fu, Jie Shen, Zhao Xie. Osteoprotegerin (OPG) Promotes Recruitment of Endothelial Progenitor Cells (EPCs) via CXCR4 Signaling Pathway to Improve Bone Defect Repair. Med Sci Monit, 2019; 25: 5572-5579. DOI: 10.12659/MSM.916838.


Subject(s)
Endothelial Progenitor Cells , Osteoprotegerin , Receptors, CXCR4 , Signal Transduction , Endothelial Progenitor Cells/metabolism , Receptors, CXCR4/metabolism , Osteoprotegerin/metabolism , Animals , Bone Regeneration/drug effects , Humans , Bone and Bones/metabolism , Osteogenesis/drug effects , Male , Mice , Wound Healing/drug effects
17.
J Cardiovasc Pharmacol ; 84(2): 220-226, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38922584

ABSTRACT

ABSTRACT: Sodium-glucose cotransporter-2 (SGLT-2) inhibitors have been shown to reduce the risk of cardiovascular mortality and hospitalizations in patients with heart failure (HF) with preserved or reduced ejection fraction (HFpEF or HFrEF). The mechanism for this benefit is not clear. Endothelial progenitor cells (EPCs) are bone marrow-derived cells able to differentiate into functional endothelial cells and participate in endothelial repair. The aim of this study was to evaluate the effect of SGLT-2 inhibitors on the level and function of EPCs in patients with HF. We enrolled 20 patients with symptomatic HF, 12 with HFrEF and 8 with HFpEF (aged 73.3 ± 10.2 years, 95% men). Blood samples were drawn at 2 time points: baseline and ≥3 months after initiation of SGLT-2 inhibitor therapy. Circulating EPC levels were evaluated by expression of vascular endothelial growth factor receptor-2 (VEGFR-2), CD34, and CD133 by flow cytometry. EPC colony forming units (CFUs) were quantified after 7 days in culture. The proportion of cells that coexpressed VEGFR-2 and CD34 or VEGFR-2 and CD133 was higher following 3 months of SGLT-2 inhibitors [0.26% (interquartile range, IQR 0.10-0.33) versus 0.55% (IQR 0.28-0.91), P = 0.002; 0.12% (IQR 0.07-0.15) versus 0.24% (IQR 0.15-0.39), P = 0.001, respectively]. EPC CFUs were also increased following SGLT-2 inhibitor treatment [23 (IQR 3.7-37.8) versus 79.4 (IQR 25.1-110.25) colonies/10 6 cells, P = 0.0039]. In patients with symptomatic HF, both HFpEF and HFrEF, treatment with SGLT-2 inhibitors is associated with an increase in the level and function of circulating EPCs. This augmentation in EPCs may be a contributing mechanism to the clinical benefit of SGLT-2 inhibitors in patients with HF.


Subject(s)
Endothelial Progenitor Cells , Heart Failure , Sodium-Glucose Transporter 2 Inhibitors , Stroke Volume , Vascular Endothelial Growth Factor Receptor-2 , Humans , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Male , Heart Failure/physiopathology , Heart Failure/drug therapy , Heart Failure/metabolism , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Aged , Female , Middle Aged , Treatment Outcome , Aged, 80 and over , Cells, Cultured , Stroke Volume/drug effects , Time Factors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Biomarkers/blood , Antigens, CD34/metabolism , Antigens, CD34/blood , AC133 Antigen/metabolism , Ventricular Function, Left/drug effects , Sodium-Glucose Transporter 2/metabolism
18.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167323, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38925483

ABSTRACT

BACKGROUND: Peripheral artery disease (PAD) is an ischemic disease with a rising incidence worldwide. The lncRNA H19 (H19) is enriched in endothelial progenitor cells (EPCs), and transplantation of pyroptosis-resistant H19-overexpressed EPCs (oe-H19-EPCs) may promote vasculogenesis and blood flow recovery in PAD, especially with critical limb ischemia (CLI). METHODS: EPCs isolated from human peripheral blood was characterized using immunofluorescence and flow cytometry. Cell proliferation was determined with CCK8 and EdU assays. Cell migration was assessed by Transwell and wound healing assays. The angiogenic potential was evaluated using tube formation assay. The pyroptosis pathway-related protein in EPCs was detected by western blot. The binding sites of H19 and FADD on miR-107 were analyzed using Luciferase assays. In vivo, oe-H19-EPCs were transplanted into a mouse ischemic limb model, and blood flow was detected by laser Doppler imaging. The transcriptional landscape behind the therapeutic effects of oe-H19-EPCs on ischemic limbs were examined with whole transcriptome sequencing. RESULTS: Overexpression of H19 in EPCs led to an increase in proliferation, migration, and tube formation abilities. These effects were mediated through pyroptosis pathway, which is regulated by the H19/miR-107/FADD axis. Transplantation of oe-H19-EPCs in a mouse ischemic limb model promoted vasculogenesis and blood flow recovery. Whole transcriptome sequencing indicated significant activation of vasculogenesis pathway in the ischemic limbs following treatment with oe-H19-EPCs. CONCLUSIONS: Overexpression of H19 increases FADD level by competitively binding to miR-107, leading to enhanced proliferation, migration, vasculogenesis, and inhibition of pyroptosis in EPCs. These effects ultimately promote the recovery of blood flow in CLI.


Subject(s)
Endothelial Progenitor Cells , Fas-Associated Death Domain Protein , Ischemia , MicroRNAs , Pyroptosis , RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Pyroptosis/genetics , Endothelial Progenitor Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Ischemia/metabolism , Ischemia/pathology , Ischemia/genetics , Humans , Animals , Mice , Fas-Associated Death Domain Protein/metabolism , Fas-Associated Death Domain Protein/genetics , Male , Lower Extremity/blood supply , Lower Extremity/pathology , Cell Movement/genetics , Cell Proliferation , Neovascularization, Physiologic/genetics , Mice, Inbred C57BL , Peripheral Arterial Disease/metabolism , Peripheral Arterial Disease/pathology , Peripheral Arterial Disease/genetics , Disease Models, Animal
19.
Stem Cells Transl Med ; 13(7): 625-636, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38733609

ABSTRACT

The management of diabetes mellitus and its resultant end organ dysfunction represents a major challenge to global health-care systems. Diabetic cardiac and kidney disease commonly co-occur and are significant contributors to the morbidity and mortality of patients with diabetes, carrying a poor prognosis. The tight link of these parallel end organ manifestations suggests a deeper common underlying pathology. Here, we outline the mechanistic link between diabetic cardiac and kidney disease, providing evidence for the role of endothelial dysfunction in both processes and the potential for cellular therapy to correct these disorders. Specifically, we review the preclinical and clinical evidence for endothelial progenitor cell therapy in cardiac, kidney, and cardio-renal disease applications. Finally, we outline novel approaches to endothelial progenitor cell therapy through cell enhancement and the use of extracellular vesicles, discussing published and future work.


Subject(s)
Endothelial Progenitor Cells , Humans , Endothelial Progenitor Cells/metabolism , Animals , Diabetic Nephropathies/therapy , Stem Cell Transplantation/methods , Extracellular Vesicles/metabolism
20.
Brain Inj ; 38(10): 835-847, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-38716911

ABSTRACT

BACKGROUND: Hyperlipidemia is a risk factor for stroke, and worsens neurological outcome after stroke. Endothelial progenitor cells (EPCs), which become dysfunctional in cerebral ischemia, hold capacity to promote revascularization. OBJECTIVE: We investigated the role of dyslipidemia in impairment of EPC-mediated angiogenesis in cerebral ischemic mice. METHODS AND RESULTS: The high fat diet (HFD)-fed mice following by ischemic stroke exhibited increased infarct volumes and neurological severity scores, and poorer angiogenesis. Bone marrow-EPCs treated with palmitic acid (PA) showed impaired functions and inhibited activity of AMP-activated protein kinase (AMPK). Notably, AMPK deficiency aggravated EPC dysfunction, further decreased mitochondrial membrane potential, and increased reactive oxygen species level in EPCs with PA treatment. Furthermore, the expression of fatty acid oxidation (FAO)-related genes was remarkably reduced, and carnitine palmitoyltransferase 1A (CPT1A) protein expression was downregulated in AMPK-deficient EPCs. AMPK deficiency aggravated neurological severity scores and angiogenesis in ischemic brain of HFD-fed mice, accompanied by suppressed protein level of CPT1A. EPC transplantation corrected impaired neurological severity scores and angiogenesis in AMPK-deficient mice. CONCLUSION: Our findings suggest that AMPK deficiency aggravates poor angiogenesis in ischemic brain by mediating FAO and oxidative stress thereby inducing EPC dysfunction in hyperlipidemic mice.


Subject(s)
Endothelial Progenitor Cells , Fatty Acids , Hyperlipidemias , Ischemic Stroke , Mice, Inbred C57BL , Animals , Endothelial Progenitor Cells/metabolism , Hyperlipidemias/metabolism , Hyperlipidemias/complications , Mice , Male , Fatty Acids/metabolism , Ischemic Stroke/metabolism , Diet, High-Fat/adverse effects , AMP-Activated Protein Kinases/metabolism , Disease Models, Animal , Oxidation-Reduction , Carnitine O-Palmitoyltransferase/metabolism , Neovascularization, Physiologic/physiology , Mice, Knockout , Angiogenesis
SELECTION OF CITATIONS
SEARCH DETAIL