Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 330
Filter
Add more filters

Publication year range
1.
Semin Cell Dev Biol ; 155(Pt C): 23-29, 2024 03 01.
Article in English | MEDLINE | ID: mdl-37202277

ABSTRACT

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.


Subject(s)
Endothelial Progenitor Cells , Endothelial Progenitor Cells/physiology , Cell Culture Techniques , Biomarkers , Neovascularization, Physiologic , Cells, Cultured
2.
EMBO J ; 39(8): e104270, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32149421

ABSTRACT

Hematopoietic stem cells (HSCs) develop from the hemogenic endothelium in cluster structures that protrude into the embryonic aortic lumen. Although much is known about the molecular characteristics of the developing hematopoietic cells, we lack a complete understanding of their origin and the three-dimensional organization of the niche. Here, we use advanced live imaging techniques of organotypic slice cultures, clonal analysis, and mathematical modeling to show the two-step process of intra-aortic hematopoietic cluster (IACH) formation. First, a hemogenic progenitor buds up from the endothelium and undergoes division forming the monoclonal core of the IAHC. Next, surrounding hemogenic cells are recruited into the IAHC, increasing their size and heterogeneity. We identified the Notch ligand Dll4 as a negative regulator of the recruitment phase of IAHC. Blocking of Dll4 promotes the entrance of new hemogenic Gfi1+ cells into the IAHC and increases the number of cells that acquire HSC activity. Mathematical modeling based on our data provides estimation of the cluster lifetime and the average recruitment time of hemogenic cells to the cluster under physiologic and Dll4-inhibited conditions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Aorta/embryology , Calcium-Binding Proteins/genetics , Cell Division , Endothelial Progenitor Cells/physiology , Female , Hemangioblasts/physiology , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Models, Theoretical
3.
Am J Respir Cell Mol Biol ; 68(2): 161-175, 2023 02.
Article in English | MEDLINE | ID: mdl-36287629

ABSTRACT

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.


Subject(s)
Acute Lung Injury , Endothelial Progenitor Cells , Respiratory Distress Syndrome , Humans , Rats , Mice , Animals , Endothelial Progenitor Cells/physiology , Vascular Endothelial Growth Factor A/metabolism , Lung/metabolism , Respiratory Distress Syndrome/therapy , Respiratory Distress Syndrome/metabolism , Acute Lung Injury/metabolism , Receptors, Lysophosphatidic Acid/metabolism
4.
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
5.
Respir Res ; 23(1): 290, 2022 Oct 23.
Article in English | MEDLINE | ID: mdl-36274148

ABSTRACT

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.


Subject(s)
Endothelial Progenitor Cells , Hypertension, Pulmonary , Animals , Humans , Endothelial Progenitor Cells/physiology , Vascular Endothelial Growth Factor Receptor-2 , Tumor Necrosis Factor-alpha , NF-E2-Related Factor 2 , Kelch-Like ECH-Associated Protein 1 , Leukocytes, Mononuclear , Chickens , Inflammation , Macrophages , RNA, Messenger
6.
Eur J Appl Physiol ; 122(7): 1589-1625, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35305142

ABSTRACT

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.


Subject(s)
Endothelial Progenitor Cells , High-Intensity Interval Training , Aged , Endothelial Progenitor Cells/physiology , Exercise/physiology , Exercise Therapy , Flow Cytometry , Humans
7.
Genes Dev ; 28(19): 2175-87, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25274728

ABSTRACT

The mammalian lymphatic vasculature is important for returning fluids from the extracellular tissue milieu back to the blood circulation. We showed previously that Prox1 dosage is important for the development of the mammalian lymphatic vasculature. The lack of Prox1 activity results in the complete absence of lymphatic endothelial cells (LECs). In Prox1 heterozygous embryos, the number of LECs is reduced because of a decrease in the progenitor pool in the cardinal vein. This reduction is caused by some progenitor cells being unable to maintain Prox1 expression. In this study, we identified Vegfr3, the cognate receptor of the lymphangiogenic growth factor Vegfc, as a dosage-dependent, direct in vivo target of Prox1. Using various mouse models, we also determined that Vegfr3 regulates Prox1 by establishing a feedback loop necessary to maintain the identity of LEC progenitors and that Vegfc-mediated activation of Vegfr3 signaling is necessary to maintain Prox1 expression in LEC progenitors. We propose that this feedback loop is the main sensing mechanism controlling the number of LEC progenitors and, as a consequence, the number of budding LECs that will form the embryonic lymphatic vasculature.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/physiology , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/physiology , Homeodomain Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Cell Count , Embryo, Nonmammalian , Gene Expression Regulation , Homeodomain Proteins/genetics , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Mice , Signal Transduction , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics
8.
Am J Respir Cell Mol Biol ; 65(5): 473-488, 2021 11.
Article in English | MEDLINE | ID: mdl-34293272

ABSTRACT

Compromised alveolar development and pulmonary vascular remodeling are hallmarks of pediatric lung diseases such as bronchopulmonary dysplasia (BPD) and alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Although advances in surfactant therapy, corticosteroids, and antiinflammatory drugs have improved clinical management of preterm infants, those who suffer with severe vascular complications still lack viable treatment options. Paucity of the alveolar capillary network in ACDMPV causes respiratory distress and leads to mortality in a vast majority of infants with ACDMPV. The discovery of endothelial progenitor cells (EPCs) in 1997 brought forth the paradigm of postnatal vasculogenesis and hope for promoting vascularization in fragile patient populations, such as those with BPD and ACDMPV. The identification of diverse EPC populations, both hematopoietic and nonhematopoietic in origin, provided a need to identify progenitor cell-selective markers that are linked to progenitor properties needed to develop cell-based therapies. Focusing on the future potential of EPCs for regenerative medicine, this review will discuss various aspects of EPC biology, beginning with the identification of hematopoietic, nonhematopoietic, and tissue-resident EPC populations. We will review knowledge related to cell surface markers, signature gene expression, and key transcriptional regulators and will explore the translational potential of EPCs for cell-based therapy for BPD and ACDMPV. The ability to produce pulmonary EPCs from patient-derived induced pluripotent stem cells in vitro holds promise for restoring vascular growth and function in the lungs of patients with pediatric pulmonary disorders.


Subject(s)
Bronchopulmonary Dysplasia/pathology , Endothelial Progenitor Cells/physiology , Lung Diseases/therapy , Persistent Fetal Circulation Syndrome/pathology , Animals , Bronchopulmonary Dysplasia/therapy , Cell Differentiation , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/transplantation , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells , Infant, Premature , Lung/blood supply , Lung/embryology , Lung/metabolism , Lung Diseases/pathology , Persistent Fetal Circulation Syndrome/therapy
9.
Cell Immunol ; 364: 104360, 2021 06.
Article in English | MEDLINE | ID: mdl-33866285

ABSTRACT

Ig-GAD2, an antigen-specific immune modulator, requires bone marrow (BM) cell transfer in order to restore beta (ß)-cell formation and induce recovery from established type 1 diabetes (T1D). The BM cells provide endothelial precursor cells (EPCs) that give rise to islet resident endothelial cells (ECs). This study shows that, during development of T1D, the immune attack causes collateral damage to the islet vascular network. The EPC-derived ECs repair and restore islet blood vessel integrity. In addition, ß-cell genetic tracing indicates that the newly formed ß-cells originate from residual ß-cells that escaped the immune attack and, unexpectedly, from ß-cell precursors. This indicates that the rejuvenated islet microenvironment invigorates formation of new ß-cells not only from residual ß-cells but also from precursor cells. This is twofold significant from the perspective of precursor cells as a safe reserve for restoration of ß-cell mass and its promise for therapy of T1D long after diagnosis.


Subject(s)
Bone Marrow Cells/physiology , Diabetes Mellitus, Type 1/therapy , Endothelial Progenitor Cells/physiology , Immunologic Factors/therapeutic use , Insulin-Secreting Cells/physiology , Recombinant Fusion Proteins/therapeutic use , Animals , Cell Differentiation , Cell Self Renewal , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Glutamate Decarboxylase/genetics , Humans , Immunoglobulins/genetics , Immunologic Factors/genetics , Mice , Mice, Inbred NOD , Recombinant Fusion Proteins/genetics , Regeneration , Regional Blood Flow
10.
Microvasc Res ; 134: 104107, 2021 03.
Article in English | MEDLINE | ID: mdl-33212112

ABSTRACT

In regenerative medicine, autologous endothelial colony forming cells (ECFCs) bear the greatest potential to be used for surface endothelialization of tissue engineered constructs, as they are easily attainable and possess a high proliferation rate. The aim of this study was to develop a standardized pre-conditioning protocol under dynamic conditions simulating the physiology of human circulation to improve the formation of a flow resistant monolayer of ECFCs and to enhance the antithrombogenicity of the endothelial cells. The main focus of the study was to consequently compare the cellular behavior under a steady laminar flow against a pulsatile flow. Mononuclear cells were isolated out of peripheral blood (PB) buffy coats and plated on uncoated tissue culture flasks in anticipation of guidelines for Advanced Therapy Medicinal Products. ECFCs were identified by typical surface markers such as CD31, CD146 and VE-Cadherin. To explore the effects of dynamic cultivation, ECFCs and human umbilical vein endothelial cells were comparatively cultured under either laminar or pulsatile (1 Hz) flow conditions with different grades of shear stress (5 dyn/cm2versus 20 dyn/cm2). High shear stress of 20 dyn/cm2 led to a significant upregulation of the antithrombotic gene marker thrombomodulin in both cell types, but only ECFCs orientated and elongated significantly after shear stress application forming a confluent endothelial cell layer. The work therefore documents a suitable protocol to pre-condition PB-derived ECFCs for sustainable endothelialization of blood contacting surfaces and provides essential knowledge for future cultivations in bioreactor systems.


Subject(s)
Endothelial Progenitor Cells/physiology , Human Umbilical Vein Endothelial Cells/physiology , Mechanotransduction, Cellular , Pulsatile Flow , Tissue Engineering , Antigens, CD/metabolism , Bioreactors , CD146 Antigen/metabolism , Cadherins/metabolism , Cell Culture Techniques/instrumentation , Cell Shape , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Female , Glucose/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Middle Aged , Neovascularization, Physiologic , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Stress, Mechanical , Thrombomodulin/genetics , Thrombomodulin/metabolism
11.
Pharmacol Res ; 173: 105920, 2021 11.
Article in English | MEDLINE | ID: mdl-34601081

ABSTRACT

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.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endothelial Progenitor Cells/pathology , Hypertension , Nuclear Proteins/metabolism , RNA, Long Noncoding , Tuberous Sclerosis Complex 2 Protein/metabolism , Aged , Angiotensin II , Animals , Autophagy , Cell Adhesion , Cell Movement , Endothelial Progenitor Cells/physiology , Female , Humans , Hypertension/genetics , Hypertension/metabolism , Hypertension/pathology , Male , Middle Aged , Rats, Inbred SHR , Rats, Wistar , beta-Galactosidase/metabolism
12.
BMC Cardiovasc Disord ; 21(1): 524, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34724901

ABSTRACT

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.


Subject(s)
Cardiac Rehabilitation , Endothelial Progenitor Cells/physiology , Heart Failure/rehabilitation , Natriuretic Peptide, Brain/analysis , Aged , Class I Phosphatidylinositol 3-Kinases/metabolism , Endothelium, Vascular/physiopathology , Exercise Therapy , Female , Heart Failure/blood , Heart Failure/physiopathology , Humans , Male , Nitric Oxide Synthase Type III/analysis , Nitric Oxide Synthase Type III/physiology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Signal Transduction/physiology , Stroke Volume , Vascular Endothelial Growth Factor A/metabolism
13.
Exp Cell Res ; 388(1): 111839, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31935381

ABSTRACT

Endothelial progenitor cell (EPC) recruitment and angiogenesis play crucial roles in aneurysm neck endothelialization, but the mechanisms of EPC recruitment and angiogenesis are still unclear. Recent studies have shown that long noncoding RNAs (lncRNAs) can regulate the function and differentiation of cells in various ways. LncRNA TUG1 is involved in liver cancer and glioma-mediated angiogenesis. The aim of this study was to investigate the role of lncRNA TUG1 in regulating EPC migration and differentiation. Overexpression and knockdown of lncRNA TUG1 with lentivirus, scratch assays, Transwell assays and tube formation assays using EPCs isolated from rat bone marrow showed that lncRNA TUG1 overexpression promoted EPC migration, invasion and differentiation. Moreover, ELISAs showed that lncRNA TUG1 overexpression increased VEGF expression. Bioinformatics prediction, luciferase assays, Western blots and RIP assays indicated that lncRNA TUG1 functions as a ceRNA (competing endogenous RNA) for miR-6321 and that miR-6321 inhibits EPC migration and differentiation through its target, ATF2. As a potential therapeutic target, lncRNA TUG1 may play a vital role in the pathogenesis of aneurysms.


Subject(s)
Cell Differentiation , Cell Movement , Endothelial Progenitor Cells/metabolism , RNA, Long Noncoding/genetics , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Animals , Cells, Cultured , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/physiology , Male , RNA, Long Noncoding/metabolism , Rats , Rats, Sprague-Dawley
14.
Med Sci Monit ; 27: e928153, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33770068

ABSTRACT

BACKGROUND The primary cause of death in patients with diabetes mellitus (DM) is diabetic macroangiopathy, a complication that related to the function and number of endothelial progenitor cells (EPCs). Salvianic acid A (SAA) is a water-soluble active ingredient of Salvia miltiorrhiza, a traditional Chinese medicine used to treat cardiovascular diseases. The purpose of this study was to explore the effects of SAA on the function of rat EPCs cultured in vitro in a high-glucose environment. MATERIAL AND METHODS Bone marrow-derived EPCs from 40 Sprague-Dawley rats were identified by fluorescence staining. Cell viability, apoptosis, tube formation, lactated dehydrogenase (LDH) release, and nitric oxide (NO) production were detected by 3-[4,5-dimethylthylthiazol-2-yl]-2,5 diphenyltetrazolium bromide assay, flow cytometry, tube formation, LDH, and 3-amino,4-aminomethyl-2',7'-difluorescein, and diacetate assays, respectively. The expression levels of proteins were examined by western blotting. RESULTS Cultured EPCs showed a cobblestone morphology and positive expression of Dil-ac-LDL and FITC-UEA-1. High glucose impaired cell viability. Different concentrations of SAA had no significant effect on EPC viability. SAA reduced the apoptosis rate and LDH release, but promoted tube formation, viability, and NO production in high-glucose-treated EPCs. The ratios of p-AKT/AKT and p-eNOS/eNOS in high-glucose-treated EPCs were elevated by SAA. Phosphoinositide 3-kinase inhibitor LY294002 blocked the rescue effects of SAA on high-glucose-treated EPCs. CONCLUSIONS SAA protected EPCs against high-glucose-induced dysfunction via the AKT/eNOS pathway.


Subject(s)
Diabetes Mellitus/drug therapy , Endothelial Cells/physiology , Endothelial Progenitor Cells/physiology , Lactates/metabolism , Medicine, Chinese Traditional , Nitric Oxide Synthase Type III/metabolism , Animals , Cell Survival , Cells, Cultured , Glucose/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Salvia , Signal Transduction
15.
Int J Mol Sci ; 22(23)2021 Nov 28.
Article in English | MEDLINE | ID: mdl-34884679

ABSTRACT

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.


Subject(s)
Endothelial Progenitor Cells/physiology , Animals , Cardiovascular Diseases/physiopathology , Cell Separation , Humans
16.
J Cell Mol Med ; 24(18): 10589-10603, 2020 09.
Article in English | MEDLINE | ID: mdl-32761803

ABSTRACT

Low-energy shock wave (LESW) has been recognized as a promising non-invasive intervention to prevent the organs or tissues against ischaemia reperfusion injury (IRI), whereas its effect on kidney injury is rarely explored. To investigate the protective role of pretreatment with LESW on renal IRI in rats, animals were randomly divided into Sham, LESW, IRI and LESW + IRI groups. At 4, 12, 24 hours and 3 and 7 days after reperfusion, serum samples and renal tissues were harvested for performing the analysis of renal function, histopathology, immunohistochemistry, flow cytometry and Western blot, as well as enzyme-linked immunosorbent assay. Moreover, circulating endothelial progenitor cells (EPCs) were isolated, labelled with fluorescent dye and injected by tail vein. The fluorescent signals of EPCs were detected using fluorescence microscope and in vivo imaging system to track the distribution of injected circulating EPCs. Results showed that pretreatment with LESW could significantly reduce kidney injury biomarkers, tubular damage, and cell apoptosis, and promote cell proliferation and vascularization in IRI kidneys. The renoprotective role of LESW pretreatment would be attributed to the remarkably increased EPCs in the treated kidneys, part of which were recruited from circulation through SDF-1/CXCR7 pathway. In conclusion, pretreatment with LESW could increase the recruitment of circulating EPCs to attenuate and repair renal IRI.


Subject(s)
Endothelial Progenitor Cells/physiology , Extracorporeal Shockwave Therapy , Kidney/blood supply , Reperfusion Injury/prevention & control , Animals , Apoptosis , Cell Movement , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Chemokine CXCL12/physiology , Extracorporeal Shockwave Therapy/methods , Fluorescent Dyes/pharmacokinetics , In Situ Nick-End Labeling , Kidney/pathology , Kidney/physiology , Male , Microscopy, Fluorescence , Microvessels/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/biosynthesis , Receptors, CXCR/genetics , Receptors, CXCR/physiology , Regeneration , Reperfusion Injury/blood , Reperfusion Injury/pathology , Signal Transduction , Time Factors
17.
Am J Pathol ; 189(11): 2269-2292, 2019 11.
Article in English | MEDLINE | ID: mdl-31421071

ABSTRACT

Lymphatic metastasis is a high-impact prognostic factor for mortality of breast cancer (BC) patients, and it directly depends on tumor-associated lymphatic vessels. We previously reported that lipopolysaccharide-induced inflammatory lymphangiogenesis is strongly promoted by myeloid-derived lymphatic endothelial cell progenitors (M-LECPs) derived from the bone marrow (BM). As BC recruits massive numbers of provascular myeloid cells, we hypothesized that M-LECPs, within this recruited population, are specifically programmed to promote tumor lymphatics that increase lymph node metastasis. In support of this hypothesis, high levels of M-LECPs were found in peripheral blood and tumor tissues of BC patients. Moreover, the density of M-LECPs and lymphatic vessels positive for myeloid marker proteins strongly correlated with patient node status. It was also established that tumor M-LECPs coexpress lymphatic-specific, stem/progenitor and M2-type macrophage markers that indicate their BM hematopoietic-myeloid origin and distinguish them from mature lymphatic endothelial cells, tumor-infiltrating lymphoid cells, and tissue-resident macrophages. Using four orthotopic BC models, we show that mouse M-LECPs are similarly recruited to tumors and integrate into preexisting lymphatics. Finally, we demonstrate that adoptive transfer of in vitro differentiated M-LECPs, but not naïve or nondifferentiated BM cells, significantly increased metastatic burden in ipsilateral lymph nodes. These data support a causative role of BC-induced lymphatic progenitors in tumor lymphangiogenesis and suggest molecular targets for their inhibition.


Subject(s)
Breast Neoplasms/pathology , Endothelial Progenitor Cells/physiology , Endothelium, Lymphatic/pathology , Myeloid Cells/physiology , Animals , Bone Marrow Cells/physiology , Cell Line, Tumor , Female , Humans , Lymphangiogenesis/physiology , Lymphatic Metastasis , Lymphatic Vessels/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID
18.
Am J Physiol Regul Integr Comp Physiol ; 318(6): R1091-R1102, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32349514

ABSTRACT

In women who conceived with or without assisted reproduction, we evaluated endothelial function by EndoPAT [reactive hyperemia index (RHI)], circulating numbers of endothelial cells (CEC) and endothelial progenitor cells (EPC), and their function before during and after pregnancy. In vitro fertilization (IVF) pregnancies were stratified by method of conception and corpus luteum (CL) number-controlled ovarian stimulation (>1 CL) or programmed (0 CL) cycles and spontaneous singleton pregnancies (1 CL). We observed 1) comparable gestational decline of RHI in the three participant groups secondary to gestational rise of baseline preocclusion pulse-wave amplitude (PWA) incorporated into the RHI calculation by EndoPAT software; 2) progressive rise in "normalized" RHI throughout pregnancy (calculated by substituting prepregnancy baseline preocclusion PWA into the RHI equation), greater in spontaneous conception vs. IVF cohorts; 3) similar gestational increase of maximum PWA and time to maximum PWA after the ischemia stimulus among the three participant groups; 4) modest gestational increase of ischemia response (reactive hyperemia) in the spontaneous conception group and no change or significant decline, respectively, in women who conceived using programmed or controlled ovarian stimulation cycles; 5) enhanced basal nitric oxide production by early (primitive) outgrowth EPC during pregnancy in women who conceived spontaneously, but not through IVF; and 6) gestational increase in CEC in all three participant cohorts, more pronounced in women who conceived by IVF using programmed cycles. On balance, the evidence supported enhanced endothelial function during pregnancy in spontaneous conceptions but less so in IVF pregnancies using either controlled ovarian stimulation or programmed cycles.


Subject(s)
Endothelial Progenitor Cells/physiology , Endothelium, Vascular/physiology , Fertilization in Vitro , Adult , Female , Humans , Middle Aged , Pregnancy , Young Adult
19.
Reproduction ; 159(4): 465-478, 2020 04.
Article in English | MEDLINE | ID: mdl-31990676

ABSTRACT

Angiogenesis is fundamental to the expansion of the placental vasculature during pregnancy. Integrins are associated with vascular formation; and osteopontin is a candidate ligand for integrins to promote angiogenesis. Endothelial progenitor cells (EPCs) are released from bone marrow into the blood and incorporate into newly vascularized tissue where they differentiate into mature endothelium. Results of studies in women suggest that EPCs may play an important role in maintaining placental vascular integrity during pregnancy, although little is known about how EPCs are recruited to these tissues. Our goal was to determine the αv integrin mediated effects of osteopontin on EPC adhesion and incorporation into angiogenic vascular networks. EPCs were isolated from 6 h old piglets. RT-PCR revealed that EPCs initially had a monocyte-like phenotype in culture that became more endothelial-like with cell passage. Immunofluorescence microscopy confirmed that the EPCs express platelet endothelial cell adhesion molecule, vascular endothelial cadherin, and von Willebrand factor. When EPCs were cultured on OPN-coated slides, the αv integrin subunit was observed in focal adhesions at the basal surface of EPCs. Silencing of αv integrin reduced EPC binding to OPN and focal adhesion assembly. In vitro siRNA knockdown in EPCs,demonstrated that OPN stimulates EPC incorporation into human umbilical vein endothelial cell (HUVEC) networks via αv-containing integrins. Finally, in situ hybridization and immunohistochemistry localized osteopontin near placental blood vessels. In summary, OPN binds the αv integrin subunit on EPCs to support EPC adhesion and increase EPC incorporation into angiogenic vascular networks.


Subject(s)
Endothelial Progenitor Cells/physiology , Integrin alphaV/metabolism , Neovascularization, Physiologic , Osteopontin/metabolism , Animals , Cell Separation , Female , Focal Adhesions/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Placenta/metabolism , Pregnancy , Swine
20.
FASEB J ; 33(8): 9142-9153, 2019 08.
Article in English | MEDLINE | ID: mdl-31084577

ABSTRACT

Endothelial colony-forming cells (ECFCs), a proliferative subpopulation of endothelial progenitor cells, are involved in angiogenesis and endothelial repair. In this study, we investigated endothelial barrier characteristics of ECFCs, whether vitamin D supports cell-cell adhesion and barrier integrity, and how it affects ECFC mobilization and actin dynamics. Although ECFC barrier was disrupted under inflammatory conditions, this effect was rescued by vitamin D treatment, leading to higher stability of an ECFC monolayer. Furthermore, vitamin D enhanced ECFC mobilization toward directional migration. In addition, immunocytochemistry, quantitative real-time PCR, and immunoblotting analysis showed that vitamin D increased endothelial interconnections through vascular endothelial cadherin (VE-cadherin) junctions and by impacting cell dynamics through cofilin and VE-cadherin phosphorylation. Our results suggest that vitamin D treatment efficiently counteracts inflammation in an ECFC monolayer, resulting in higher ECFC barrier integrity. This study provides evidence of a new beneficial effect of vitamin D for ECFC homeostasis.-Schröder-Heurich, B., von Hardenberg, S., Brodowski, L., Kipke, B., Meyer, N., Borns, K., von Kaisenberg, C. S., Brinkmann, H., Claus, P., von Versen-Höynck, F. Vitamin D improves endothelial barrier integrity and counteracts inflammatory effects on endothelial progenitor cells.


Subject(s)
Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/physiology , Inflammation/drug therapy , Vitamin D/pharmacology , Adherens Junctions/drug effects , Antigens, CD/genetics , Antigens, CD/physiology , Cadherins/genetics , Cadherins/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Coculture Techniques , Colony-Forming Units Assay , Endothelial Progenitor Cells/pathology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/pathology , Inflammation/physiopathology , Tumor Necrosis Factor-alpha/metabolism , Vitamin D/physiology
SELECTION OF CITATIONS
SEARCH DETAIL