Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 435
Filter
1.
J Am Heart Assoc ; 13(9): e031972, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38639380

ABSTRACT

BACKGROUND: Coronary microvascular dysfunction (CMD) represents an early functional characteristic of coronary vascular aging. Klotho (α-klotho) is a circulating protein inversely linked to physiological aging. We examined low klotho as a potential marker for vascular aging in patients with CMD and no coronary artery disease. METHODS AND RESULTS: Patients undergoing nonurgent angiogram for chest pain who had no coronary artery disease underwent invasive coronary microvascular and endothelial function testing. CMD was defined by ≤50% increase in coronary blood flow (percentage change in coronary blood flow) in response to intracoronary acetylcholine or coronary flow reserve ≤2. Fresh arterial whole blood was used to analyze circulating endothelial progenitor cells with flow cytometry. Stored arterial plasma was used for klotho analysis by ELISA. Participants with CMD (n=62) were compared with those without CMD (n=36). Those with CMD were age 55±10 years (versus 51±11 years; P=0.07) and 73% women (versus 81%; P=0.38). Traditional risk factors for coronary artery disease were similar between groups. Patients with CMD had less klotho (0.88±1.50 versus 1.75±2.38 ng/mL; P=0.03), and the odds of low klotho in CMD were significant in a logistic regression model after adjusting for traditional cardiovascular risk factors (odds ratio [OR], 0.80 [95% CI, 0.636-0.996]; P=0.05). Higher klotho was associated with higher numbers of endothelial progenitor cells with vascular regenerative potential (CD34+ and CD34+CD133+KDR+). Among a subgroup of patients with atherosclerotic cardiovascular disease risk <5% (n=58), CMD remained associated with lower klotho (OR, 0.80 [95% CI, 0.636-0.996]; P=0.047). CONCLUSIONS: Klotho may be a biomarker for CMD and may be a therapeutic target for groups of patients without significant traditional cardiovascular risk.


Subject(s)
Biomarkers , Coronary Circulation , Glucuronidase , Klotho Proteins , Humans , Female , Male , Glucuronidase/blood , Middle Aged , Biomarkers/blood , Coronary Circulation/physiology , Coronary Vessels/physiopathology , Coronary Vessels/diagnostic imaging , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Adult , Coronary Angiography , Microcirculation , Coronary Artery Disease/blood , Coronary Artery Disease/physiopathology , Coronary Artery Disease/diagnosis , Aged , Flow Cytometry , Enzyme-Linked Immunosorbent Assay
2.
Oncogene ; 43(13): 944-961, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38351345

ABSTRACT

Metastasis causes most cancer-related deaths, and the role and mechanism of periostin (POSTN) in the metastasis of hepatocellular carcinoma (HCC) remain undiscovered. In this study, DEN and HTVi HCC models were performed in hepatic-specific Postn ablation and Postn knock-in mouse to reveal the role of POSTN in HCC metastasis. Furthermore, POSTN was positively correlated with circulating EPCs level and promoted EPC mobilization and tumour infiltration. POSTN also mediated the crosstalk between HCC and EPCs, which promoted metastasis ability and upregulated CD36 expression in HCC through indirect crosstalk. Chemokine arrays further revealed that hepatic-derived POSTN induced elevated CCL2 expression and secretion in EPCs, and CCL2 promoted prometastatic traits in HCC. Mechanistic studies showed that POSTN upregulated CCL2 expression in EPCs via the αvß3/ILK/NF-κB pathway. CCL2 further induced CD36 expression via the CCR2/STAT3 pathway by directly binding to the promoter region of CD36. Finally, CD36 was verified to have a prometastatic role in vitro and to be correlated with POSTN expression, metastasis and recurrence in HCC in clinical samples. Our findings revealed that crosstalk between HCC and EPCs is mediated by periostin/CCL2/CD36 signalling which promotes HCC metastasis and emphasizes a potential therapeutic strategy for preventing HCC metastasis.


Subject(s)
CD36 Antigens , Carcinoma, Hepatocellular , Chemokine CCL2 , Endothelial Progenitor Cells , Liver Neoplasms , Periostin , Animals , Mice , Carcinoma, Hepatocellular/pathology , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Liver Neoplasms/pathology , Signal Transduction/genetics , Tumor Microenvironment/genetics , Chemokine CCL2/metabolism , CD36 Antigens/metabolism
3.
Mol Oral Microbiol ; 39(2): 47-61, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37188376

ABSTRACT

We found that GroEL in Porphyromonas gingivalis accelerated tumor growth and increased mortality in tumor-bearing mice; GroEL promoted proangiogenic function, which may be the reason for promoting tumor growth. To understand the regulatory mechanisms by which GroEL increases the proangiogenic function of endothelial progenitor cells (EPCs), we explored in this study. In EPCs, MTT assay, wound-healing assay, and tube formation assay were performed to analyze its activity. Western blot and immunoprecipitation were used to study the protein expression along with next-generation sequencing for miRNA expression. Finally, a murine tumorigenesis animal model was used to confirm the results of in vitro. The results indicated that thrombomodulin (TM) direct interacts with PI3 K/Akt to inhibit the activation of signaling pathways. When the expression of TM is decreased by GroEL stimulation, molecules in the PI3 K/Akt signaling axis are released and activated, resulting in increased migration and tube formation of EPCs. In addition, GroEL inhibits TM mRNA expression by activating miR-1248, miR-1291, and miR-5701. Losing the functions of miR-1248, miR-1291, and miR-5701 can effectively alleviate the GroEL-induced decrease in TM protein levels and inhibit the proangiogenic abilities of EPCs. These results were also confirmed in animal experiments. In conclusion, the intracellular domain of the TM of EPCs plays a negative regulatory role in the proangiogenic capabilities of EPCs, mainly through direct interaction between TM and PI3 K/Akt to inhibit the activation of signaling pathways. The effects of GroEL on tumor growth can be reduced by inhibiting the proangiogenic properties of EPCs through the inhibition of the expression of specific miRNAs.


Subject(s)
Endothelial Progenitor Cells , MicroRNAs , Neoplasms , Mice , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Porphyromonas gingivalis/genetics , Proto-Oncogene Proteins c-akt/metabolism , Thrombomodulin/genetics , Thrombomodulin/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Physiologic/physiology
4.
J Transl Med ; 21(1): 881, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38057857

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) has impacted approximately 390 million people worldwide and the morbidity is increasing every year. However, due to the poor treatment efficacy of COPD, exploring novel treatment has become the hotpot of study on COPD. Endothelial progenitor cells (EPCs) aging is a possible molecular way for COPD development. We aimed to explore the effector whether intravenous administration of EPCs has therapeutic effects in COPD mice. METHODS: COPD mice model was induced by cigarette smoke exposure and EPCs were injected intravenously to investigate their effects on COPD mice. At day 127, heart, liver, spleen, lung and kidney tissues of mice were harvested. The histological effects of EPCs intervention on multiple organs of COPD mice were detected by morphology assay. Quantitative real-time PCR and Western blotting were used to detect the effect of EPCs intervention on the expression of multi-organ senescence-related indicators. And we explored the effect of EPCs systematically intervening on senescence-related USP7/p300 pathway. RESULTS: Compared with COPD group, senescence-associated ß-galactosidase activity was decreased, protein and mRNA expression of p16 was down-regulated, while protein and mRNA expression of cyclin D1 and TERT were up-regulated of multiple organs, including lung, heart, liver, spleen and kidney in COPD mice after EPCs system intervention. But the morphological alterations of the tissues described above in COPD mice failed to be reversed. Mechanistically, EPCs systemic administration inhibited the expression of mRNA and protein of USP7 and p300 in multiple organs of COPD mice, exerting therapeutic effects. CONCLUSIONS: EPCs administration significantly inhibited the senescence of multiple organs in COPD mice via down-regulating USP7/p300 pathway, which presents a possibility of EPCs therapy for COPD.


Subject(s)
Endothelial Progenitor Cells , Pulmonary Disease, Chronic Obstructive , Signal Transduction , Animals , Humans , Mice , Cellular Senescence , Endothelial Progenitor Cells/pathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/therapy , Pulmonary Disease, Chronic Obstructive/metabolism , RNA, Messenger/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Down-Regulation
5.
Nat Aging ; 3(11): 1401-1414, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37946040

ABSTRACT

The stem cell theory of aging dictates that a decline in the number and/or function of stem cells causes tissue degeneration and aging; however, it still lacks unequivocal experimental support. Here, using lineage tracing and single-cell transcriptomics, we identify a population of CD133+ bone marrow-derived endothelial-like cells (ELCs) as potential endothelial progenitor cells, which contribute to tubular structures in vitro and neovascularization in vivo. We demonstrate that supplementation with wild-type and young ELCs respectively restores neovascularization and extends lifespan in progeric and naturally aged mice. Mechanistically, we identify an upregulation of farnesyl diphosphate synthase (FDPS) in aged CD133+ ELCs-a key enzyme in isoprenoid biosynthesis. Overexpression of FDPS compromises the neovascularization capacity of CD133+ ELCs, whereas FDPS inhibition by pamidronate enhances neovascularization, improves health measures and extends lifespan in aged mice. These findings highlight stem cell-based strategies for the treatment of progeria and age-related pathologies.


Subject(s)
Endothelial Progenitor Cells , Mice , Animals , Endothelial Progenitor Cells/pathology , Longevity , Neovascularization, Pathologic/pathology , Stem Cells/pathology
6.
J Int Adv Otol ; 19(2): 76-80, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36975077

ABSTRACT

BACKGROUND: This study aimed to investigate the correlation between lipoprotein-associated phospholipase A2, endothelial progenitor cells, and sudden sensorineural hearing loss. METHODS: The number of endothelial progenitor cells and lipoprotein-associated phospholipase A2 levels collected from peripheral blood samples were measured and compared between sudden sensorineural hearing loss group and control group. RESULTS: The number of endothelial progenitor cells was reduced in sudden sensorineural hearing loss group compared to control group (38.88 ± 10.73 in sudden sensorineural hearing loss group vs. 77.14 ± 8.56 in control group, P <.01). The lipoprotein-associated phospholipase A2 level was markedly increased in sudden sensorineural hearing loss group compared to control group (244.94 ± 59.547 in sudden sensorineural hearing loss group vs. 189.00 ± 50.987 in control group, P <.05). CONCLUSION: The number of endothelial progenitor cells was decreased and lipoprotein-associated phospholipase A2 levels were increased in sudden sensorineural hearing loss patients. Changes in the number of endothelial progenitor cells and lipoprotein-associated phospholipase A2 levels may be involved in the pathogenesis of sudden sensorineural hearing loss.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase , Endothelial Progenitor Cells , Hearing Loss, Sensorineural , Hearing Loss, Sudden , Humans , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Endothelial Progenitor Cells/pathology
7.
Int J Mol Sci ; 24(2)2023 Jan 08.
Article in English | MEDLINE | ID: mdl-36674749

ABSTRACT

Moyamoya arteriopathy (MMA) is a rare cerebrovascular disorder that causes recurrent ischemic and hemorrhagic strokes, leading young patients to severe neurological deficits. The pathogenesis of MMA is still unknown. The disease onset in a wide number of pediatric cases raises the question of the role of genetic factors in the disease's pathogenesis. In these patients, MMA's clinical course, or progression, is largely unclear. By performing a comprehensive molecular and cellular profile in the plasma and CSF, respectively, of MMA pediatric patients, our study is aimed at assessing the levels of circulating endothelial progenitor cells (cEPC) and the release of selected proteins at an early disease stage to clarify MMA pathogenesis and progression. We employed cytofluorimetric methods and immunoassays in pediatric MMA patients and matched control subjects by age and sex. We detected increased levels of cEPC in peripheral blood and an upregulation of angiogenic markers in CSF (i.e., angiopoietin-2 and VEGF-A). This finding is probably associated with deregulated angiogenesis, as stated by the moderate severity of collateral vessel network development (Suzuki III-IV). The absence of significant modulation of neurofilament light in CSF led us to rule out the presence of substantial neuronal injury in MMA children. Despite the limited cohort of pediatric patients, we found some peculiar cellular and molecular characteristics in their blood and CSF samples. Our findings may be confirmed by wider and perspective studies to identify predictive or prognostic circulating biomarkers and potential therapeutic targets for personalized care of MMA pediatric patients.


Subject(s)
Endothelial Progenitor Cells , Hemorrhagic Stroke , Moyamoya Disease , Humans , Child , Endothelial Progenitor Cells/pathology , Moyamoya Disease/pathology
8.
Acta Pharmacol Sin ; 44(5): 999-1013, 2023 May.
Article in English | MEDLINE | ID: mdl-36347996

ABSTRACT

Non-healing diabetic wounds (DW) are a serious clinical problem that remained poorly understood. We recently found that topical application of growth differentiation factor 11 (GDF11) accelerated skin wound healing in both Type 1 DM (T1DM) and genetically engineered Type 2 diabetic db/db (T2DM) mice. In the present study, we elucidated the cellular and molecular mechanisms underlying the action of GDF11 on healing of small skin wound. Single round-shape full-thickness wound of 5-mm diameter with muscle and bone exposed was made on mouse dorsum using a sterile punch biopsy 7 days following the onset of DM. Recombinant human GDF11 (rGDF11, 50 ng/mL, 10 µL) was topically applied onto the wound area twice a day until epidermal closure (maximum 14 days). Digital images of wound were obtained once a day from D0 to D14 post-wounding. We showed that topical application of GDF11 accelerated the healing of full-thickness skin wounds in both type 1 and type 2 diabetic mice, even after GDF8 (a muscle growth factor) had been silenced. At the cellular level, GDF11 significantly facilitated neovascularization to enhance regeneration of skin tissues by stimulating mobilization, migration and homing of endothelial progenitor cells (EPCs) to the wounded area. At the molecular level, GDF11 greatly increased HIF-1ɑ expression to enhance the activities of VEGF and SDF-1ɑ, thereby neovascularization. We found that endogenous GDF11 level was robustly decreased in skin tissue of diabetic wounds. The specific antibody against GDF11 or silence of GDF11 by siRNA in healthy mice mimicked the non-healing property of diabetic wound. Thus, we demonstrate that GDF11 promotes diabetic wound healing via stimulating endothelial progenitor cells mobilization and neovascularization mediated by HIF-1ɑ-VEGF/SDF-1ɑ pathway. Our results support the potential of GDF11 as a therapeutic agent for non-healing DW.


Subject(s)
Diabetes Mellitus, Experimental , Endothelial Progenitor Cells , Growth Differentiation Factors , Wound Healing , Animals , Humans , Mice , Bone Morphogenetic Proteins/metabolism , Chemokine CXCL12/drug effects , Chemokine CXCL12/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Growth Differentiation Factors/therapeutic use , Growth Differentiation Factors/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/drug effects , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
9.
Adv Clin Exp Med ; 31(11): 1215-1229, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36047895

ABSTRACT

BACKGROUND: Diabetes mellitus (DM) often causes stenosis and occlusion of hindlimb blood vessels, which are also the main cause for hindlimb ischemia in elderly people. OBJECTIVES: To investigate the therapeutic effect of endothelial progenitor cell (EPC) transplantation on diabetic hindlimb ischemia. MATERIAL AND METHODS: Endothelial progenitor cells were separated, labeled with PKH-26 and transplanted into rat models (107 cells/100 g). Dichlorodihydrofluorescein diacetate (DCFH-DA) was used to detect any oxidative stress. Streptozotocin (STZ) was injected to establish a diabetic rat model and hindlimb ischemia model was established via operation. Western blotting was used to detect total ß-catenin (T-ß-catenin) and non-phospho-ß-catenin (NP-ß-catenin) levels. The malondialdehyde (MDA), superoxide dismutase (SOD), Wnt3a, Wnt5a and Wnt7a levels were detected using enzyme-linked immunosorbent assay (ELISA). Oxidative stress was measured using DCFH-DA and dihydroethidium (DHE). The endothelial biomarker CD31 was observed to highlight vessels, and PKH-26 to trace migration/adhesion of EPCs. RESULTS: Endothelial progenitor cells were successfully isolated and identified, and diabetic hindlimb ischemic rat models were created. Tempol remarkably improved blood flow in diabetic hindlimb ischemic rats compared to DM+EPCs rats at 14 days (p < 0.001) and 28 days post-operation (p < 0.001). High oxidative stress was observed in diabetic hindlimb ischemic rats. Tempol significantly inhibited oxidative stress levels in diabetic hindlimb ischemic rats. Furthermore, Tempol significantly promoted angiogenesis in diabetic hindlimb ischemic rats compared to DM+EPCs rats. The ß-catenin inhibitor, XAV (DM+EPCs+Tempol+XAV group), significantly suppressed blood flow recovery and angiogenesis in diabetic hindlimb ischemic rats when compared to the DM+EPCs+Tempol group at 14 days (p = 0.026) and 28 days (p < 0.001). The XAV remarkably reduced T-ß-catenin (p < 0.001) and N-ß-catenin (p = 0.030) levels in Tempol-treated diabetic hindlimb ischemic rats, as compared to the DM+EPCs+Tempol group. The Wnt5a participated in the pathology of diabetic hindlimb ischemia. CONCLUSIONS: There are high oxidative stress levels in both EPCs in high-glucose environments and diabetic hindlimb ischemia, which can lead to limited blood flow recovery. The high oxidative stress caused the inhibition of Wnt/ß-catenin signaling pathway, leading to limited blood flow recovery in diabetic hindlimb ischemia. At the same time, Wnt5a participated in the EPC-mediated blood flow recovery.


Subject(s)
Diabetes Mellitus , Endothelial Progenitor Cells , Animals , Rats , beta Catenin/metabolism , Diabetes Mellitus/metabolism , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Hindlimb/blood supply , Ischemia , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/physiology , Oxidative Stress , Wnt Signaling Pathway
10.
Int J Mol Sci ; 23(13)2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35806336

ABSTRACT

Oxidized low-density lipoprotein (ox-LDL) is the most harmful form of cholesterol associated with vascular atherosclerosis and hepatic injury, mainly due to inflammatory cell infiltration and subsequent severe tissue injury. Lox-1 is the central ox-LDL receptor expressed in endothelial and immune cells, its activation regulating inflammatory cytokines and chemotactic factor secretion. Recently, a Lox-1 truncated protein isoform lacking the ox-LDL binding domain named LOXIN has been described. We have previously shown that LOXIN overexpression blocked Lox-1-mediated ox-LDL internalization in human endothelial progenitor cells in vitro. However, the functional role of LOXIN in targeting inflammation or tissue injury in vivo remains unknown. In this study, we investigate whether LOXIN modulated the expression of Lox-1 and reduced the inflammatory response in a high-fat-diet mice model. Results indicate that human LOXIN blocks Lox-1 mediated uptake of ox-LDL in H4-II-E-C3 cells. Furthermore, in vivo experiments showed that overexpression of LOXIN reduced both fatty streak lesions in the aorta and inflammation and fibrosis in the liver. These findings were associated with the down-regulation of Lox-1 in endothelial cells. Then, LOXIN prevents hepatic and aortic tissue damage in vivo associated with reduced Lox-1 expression in endothelial cells. We encourage future research to understand better the underlying molecular mechanisms and potential therapeutic use of LOXIN.


Subject(s)
Atherosclerosis , Endothelial Progenitor Cells , Phthalazines , Animals , Aorta/metabolism , Aorta/pathology , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Diet, High-Fat/adverse effects , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipoproteins, LDL/metabolism , Liver/metabolism , Mice , Phthalazines/pharmacology , Scavenger Receptors, Class E/genetics , Scavenger Receptors, Class E/metabolism
11.
Arq Neuropsiquiatr ; 80(5): 469-474, 2022 05.
Article in English | MEDLINE | ID: mdl-35613207

ABSTRACT

BACKGROUND: At present, the etiology and pathogenesis of Moyamoya disease (MMD) are not completely clear. Patients are usually diagnosed after cerebrovascular events. Therefore, it is of great clinical significance to explore the predictive factors of MMD. OBJECTIVE: This study aimed to investigate the serum level of CoQ10B, the amount of endothelial progenitor cells (EPCs), and mitochondrial function of EPCs in MMD patients. METHODS: Forty-one MMD patients and 20 healthy controls were recruited in this study. Patients with MMD were divided into two groups: Ischemic type (n=23) and hemorrhagic type (n=18). Blood samples were collected from the antecubital vein and analyzed by CoQ10B ELISA and flow cytometry. Measures of mitochondrial function of EPCs include oxygen consumption rate (OCR), mitochondrial membrane potential, Ca2+ concentration, adenosine triphosphatases activity and ROS level. RESULTS: The serum CoQ10B level in MMD patients was significantly lower than that in healthy controls (p<0.001). The relative number of EPCs in MMD patients was significantly higher than that in healthy controls (p<0.001). Moreover, the OCR, mitochondrial membrane potential and ATPase activity were decreased and the Ca2+ and reactive oxygen species levels were increased in MMD patients (p<0.001). CONCLUSIONS: Our results showed obviously decreased serum CoQ10B level and increased EPCs number in patients with MMD compared with healthy patients, and the mitochondria function of EPCs in MMD patients was abnormal.


Subject(s)
Endothelial Progenitor Cells , Moyamoya Disease , Calcium , Disease Progression , Endothelial Progenitor Cells/pathology , Humans , Moyamoya Disease/pathology
12.
J Am Soc Nephrol ; 33(3): 565-582, 2022 03.
Article in English | MEDLINE | ID: mdl-35091451

ABSTRACT

BACKGROUND: Endothelial cell injury is a common nidus of renal injury in patients and consistent with the high prevalence of AKI reported during the coronavirus disease 2019 pandemic. This cell type expresses integrin α5 (ITGA5), which is essential to the Tie2 signaling pathway. The microRNA miR-218-5p is upregulated in endothelial progenitor cells (EPCs) after hypoxia, but microRNA regulation of Tie2 in the EPC lineage is unclear. METHODS: We isolated human kidney-derived EPCs (hkEPCs) and surveyed microRNA target transcripts. A preclinical model of ischemic kidney injury was used to evaluate the effect of hkEPCs on capillary repair. We used a genetic knockout model to evaluate the effect of deleting endogenous expression of miR-218 specifically in angioblasts. RESULTS: After ischemic in vitro preconditioning, miR-218-5p was elevated in hkEPCs. We found miR-218-5p bound to ITGA5 mRNA transcript and decreased ITGA5 protein expression. Phosphorylation of 42/44 MAPK decreased by 73.6% in hkEPCs treated with miR-218-5p. Cells supplemented with miR-218-5p downregulated ITGA5 synthesis and decreased 42/44 MAPK phosphorylation. In a CD309-Cre/miR-218-2-LoxP mammalian model (a conditional knockout mouse model designed to delete pre-miR-218-2 exclusively in CD309+ cells), homozygotes at e18.5 contained avascular glomeruli, whereas heterozygote adults showed susceptibility to kidney injury. Isolated EPCs from the mouse kidney contained high amounts of ITGA5 and showed decreased migratory capacity in three-dimensional cell culture. CONCLUSIONS: These results demonstrate the critical regulatory role of miR-218-5p in kidney EPC migration, a finding that may inform efforts to treat microvascular kidney injury via therapeutic cell delivery.


Subject(s)
Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Integrin alpha5/metabolism , MicroRNAs/physiology , Acute Kidney Injury/pathology , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, TIE-2/physiology , Signal Transduction/physiology
13.
J Endocrinol Invest ; 45(6): 1173-1180, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35089541

ABSTRACT

PURPOSE: Osteocalcin (OC), an osteoblast-derived regulator of metabolic processes, and circulating early endothelial progenitor cells (EPC, CD34 - /CD133 + /KDR +) expressing OC (OC +) are potential candidates linking bone metabolism and the vasculature and might be involved in vascular atherosclerotic calcification. This study aimed at assessing the association of circulating levels of different OC forms and of EPCs count with disease severity in patients with documented coronary atherosclerosis (CAD). METHODS: Patients (n = 59) undergoing coronary angiography were divided, according to stenosis severity, into (1) early coronary atherosclerosis (ECA) (n = 22), and (2) late coronary atherosclerosis (LCA) (n = 37). Total OC (TOC), carboxylated OC (cOC), undercarboxylated OC (unOC) were quantified by ELISA. EPC OC + count was assessed by flow cytometry. RESULTS: EPC OC + counts showed significant differences between ECA and LCA groups. unOC and unOC/TOC ratio were inversely correlated with EPC OC + count. A significant decrease in TOC and unOC plasma levels was associated with higher cardiovascular risk factors (CVRFs) number. EPC OC + count was correlated with LDL-C, total cholesterol, and triglycerides, with a greater significance in the LCA group. No association between the different forms of circulating OC (TOC, ucOC, cOC) and severity of CAD was found. CONCLUSION: This study showed a significant association between EPCs (CD34 - /CD133 + /KDR + /OC +), CAD severity and CVRFs, suggesting an active role for EPC OC + in the development of CAD. An inverse correlation between TOC, ucOC, and number of CVRFs was observed, suggesting that OC, regardless of its carboxylation status, may be developed as a further cardiovascular risk biomarker.


Subject(s)
Coronary Artery Disease , Endothelial Progenitor Cells , Osteocalcin , Antigens, CD34 , Biomarkers/blood , Biomarkers/metabolism , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Female , Humans , Male , Osteocalcin/blood , Osteocalcin/metabolism , Severity of Illness Index
14.
Arch Physiol Biochem ; 128(4): 1071-1080, 2022 Aug.
Article in English | MEDLINE | ID: mdl-32374186

ABSTRACT

BACKGROUND: We speculated impacts of BM-MSCs and UC-EPCs on reversal of hepatic injury induced by carbon tetrachloride (CCl4). Fifty adult rats were divided into five groups: control group, CCl4A group, CCl4B group, CCl4/BM-MSCs group and CCl4/UC-EPCs group. Blood samples were driven to measure concentration of albumin and ALT. Quantitative expression of HGF, TGF-ß, MMP-2, and VEGF were assessed by PCR. Histological and immunohistochemistry examination of the liver tissue were performed. RESULTS: There was elevating albumin (p < .05) and reducing ALT (p < .05) concentrations in groups treated with BM-MSCs and UC-EPCs compared to untreated CCL4A&B groups. UC-EPCs treated group have significantly higher MMP-2 and VEGF (p < .01) genes expression than BM-MSCs treated group. Furthermore, UC-EPCs were more valuable than BMMSCs in increasing gene expression of HGF (p < .05) and immunohistochemistry of α-SMA and Ki-67 (p < .01). BM-MSCs have significantly lower TGF-ß (p < .00) compared to UC-EPCs. CONCLUSION: This study highlighted on liver regeneration role of both UC-EPCs and BM-MSCs in liver fibrosis.


Subject(s)
Endothelial Progenitor Cells , Mesenchymal Stem Cells , Albumins , Animals , Carbon Tetrachloride/toxicity , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/therapy , Matrix Metalloproteinase 2/metabolism , Mesenchymal Stem Cells/pathology , Rats , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
15.
Arterioscler Thromb Vasc Biol ; 42(1): e27-e43, 2022 01.
Article in English | MEDLINE | ID: mdl-34670408

ABSTRACT

OBJECTIVE: Capillary malformation (CM) occurs sporadically and is associated with Sturge-Weber syndrome. The somatic mosaic mutation in GNAQ (c.548G>A, p.R183Q) is enriched in endothelial cells (ECs) in skin CM and Sturge-Weber syndrome brain CM. Our goal was to investigate how the mutant Gαq (G-protein αq subunit) alters EC signaling and disrupts capillary morphogenesis. Approach and Results: We used lentiviral constructs to express p.R183Q or wild-type GNAQ in normal human endothelial colony forming cells (EC-R183Q and EC-WT, respectively). EC-R183Q constitutively activated PLC (phospholipase C) ß3, a downstream effector of Gαq. Activated PLCß3 was also detected in human CM tissue sections. Bulk RNA sequencing analyses of mutant versus wild-type EC indicated constitutive activation of PKC (protein kinase C), NF-κB (nuclear factor kappa B) and calcineurin signaling in EC-R183Q. Increased expression of downstream targets in these pathways, ANGPT2 (angiopoietin-2) and DSCR (Down syndrome critical region protein) 1.4 were confirmed by quantitative PCR and immunostaining of human CM tissue sections. The Gαq inhibitor YM-254890 as well as siRNA targeted to PLCß3 reduced mRNA expression levels of these targets in EC-R183Q while the pan-PKC inhibitor AEB071 reduced ANGPT2 but not DSCR1.4. EC-R183Q formed enlarged blood vessels in mice, reminiscent of those found in human CM. shRNA knockdown of ANGPT2 in EC-R183Q normalized the enlarged vessels to sizes comparable those formed by EC-WT. CONCLUSIONS: Gαq-R183Q, when expressed in ECs, establishes constitutively active PLCß3 signaling that leads to increased ANGPT2 and a proangiogenic, proinflammatory phenotype. EC-R183Q are sufficient to form enlarged CM-like vessels in mice, and suppression of ANGPT2 prevents the enlargement. Our study provides the first evidence that endothelial Gαq-R183Q is causative for CM and identifies ANGPT2 as a contributor to CM vascular phenotype.


Subject(s)
Angiopoietin-2/metabolism , Capillaries/metabolism , Endothelial Progenitor Cells/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Neovascularization, Pathologic , Sturge-Weber Syndrome/metabolism , Adolescent , Adult , Aged , Angiopoietin-2/genetics , Animals , Capillaries/abnormalities , Cells, Cultured , Child , Child, Preschool , Endothelial Progenitor Cells/pathology , Endothelial Progenitor Cells/transplantation , Female , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Infant , Infant, Newborn , Male , Mice, Nude , Mutation , Phenotype , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Protein Kinase C/metabolism , Signal Transduction , Sturge-Weber Syndrome/genetics , Sturge-Weber Syndrome/pathology , Up-Regulation
16.
Thromb Haemost ; 122(3): 456-469, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34214997

ABSTRACT

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


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , Endothelial Progenitor Cells , Neointima , Receptors, Interleukin-8B/metabolism , Regeneration/physiology , Vascular System Injuries , Adaptation, Physiological/physiology , Animals , Apoptosis , Arteries/injuries , Arteries/metabolism , Cells, Cultured , Disease Models, Animal , Endothelial Progenitor Cells/pathology , Endothelial Progenitor Cells/physiology , Mice , Myocytes, Smooth Muscle , Neointima/etiology , Neointima/metabolism , Neointima/pathology , Neointima/prevention & control , Receptors, CCR5/metabolism , Signal Transduction/physiology , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
17.
Front Immunol ; 12: 737990, 2021.
Article in English | MEDLINE | ID: mdl-34659230

ABSTRACT

Angiogenesis is a critical process in the formation of new capillaries and a key participant in rheumatoid arthritis (RA) pathogenesis. The adipokine apelin (APLN) plays critical roles in several cellular functions, including angiogenesis. We report that APLN treatment of RA synovial fibroblasts (RASFs) increased angiopoietin-1 (Ang1) expression. Ang1 antibody abolished endothelial progenitor cell (EPC) tube formation and migration in conditioned medium from APLN-treated RASFs. We also found significantly higher levels of APLN and Ang1 expression in synovial fluid from RA patients compared with those with osteoarthritis. APLN facilitated Ang1-dependent EPC angiogenesis by inhibiting miR-525-5p synthesis via phospholipase C gamma (PLCγ) and protein kinase C alpha (PKCα) signaling. Importantly, infection with APLN shRNA mitigated EPC angiogenesis, articular swelling, and cartilage erosion in ankle joints of mice with collagen-induced arthritis. APLN is therefore a novel therapeutic target for RA.


Subject(s)
Angiopoietin-1/metabolism , Apelin/metabolism , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Endothelial Progenitor Cells/metabolism , Fibroblasts/metabolism , MicroRNAs/metabolism , Neovascularization, Pathologic , Synoviocytes/metabolism , 3T3 Cells , Angiopoietin-1/genetics , Animals , Apelin/genetics , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Chick Embryo , Endothelial Progenitor Cells/pathology , Fibroblasts/pathology , Humans , Mice , MicroRNAs/genetics , RNA Interference , Signal Transduction , Synoviocytes/pathology
18.
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
19.
Sci Rep ; 11(1): 18431, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34531450

ABSTRACT

The aim of this study was to analyse the association between human immunodeficiency virus (HIV) related clinical and analytical parameters and the presence of subclinical atherosclerosis as well as endothelial dysfunction. This was a prospective cohort study of HIV-positive patients who underwent intima media thickness (IMT) determination and coronary artery calcium scoring to determine subclinical atherosclerosis. To detect endothelial dysfunction, the breath holding index, flow-mediated dilation and the concentration of endothelial progenitor cells (EPCs) were measured. Patients with an IMT ≥ 0.9 mm had an average of 559.3 ± 283.34 CD4/µl, and those with an IMT < 0.9 mm had an average of 715.4 ± 389.92 CD4/µl (p = 0.04). Patients with a low calcium score had a significantly higher average CD4 cell value and lower zenith viral load (VL) than those with a higher score (707.7 ± 377.5 CD4/µl vs 477.23 ± 235.7 CD4/µl (p = 0.01) and 7 × 104 ± 5 × 104 copies/ml vs 23.4 × 104 ± 19 × 104 copies/ml (p = 0.02)). The number of early EPCs in patients with a CD4 nadir < 350/µl was lower than that in those with a CD4 nadir ≥ 350 (p = 0.03). In HIV-positive patients, low CD4 cell levels and high VL were associated with risk of developing subclinical atherosclerosis. HIV patients with CD4 cell nadir < 350/µl may have fewer early EPCs.


Subject(s)
Atherosclerosis/diagnosis , Endothelium, Vascular/pathology , HIV Infections/complications , Adult , Aged , Atherosclerosis/complications , Breath Holding , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Endothelial Progenitor Cells/pathology , Endothelium, Vascular/physiopathology , Female , Humans , Male , Middle Aged , Vasodilation
20.
Am J Physiol Heart Circ Physiol ; 321(5): H920-H932, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34533398

ABSTRACT

Bone marrow-derived endothelial progenitor cells (EPCs) contribute to endothelial repair and angiogenesis. Reduced number of circulating EPCs is associated with future cardiovascular events. We tested whether dysregulated glucose and/or triglyceride (TG) metabolism has an impact on EPC homeostasis. The analysis of metabolic factors associated with circulating EPC number in humans revealed that postprandial hyperglycemia is negatively correlated with circulating EPC number, and this correlation appears to be further enhanced in the presence of postprandial hypertriglyceridemia (hTG). We therefore examined the effect of glucose/TG spikes on bone marrow lineage-sca-1+ c-kit+ (LSK) cells in mice, because primitive EPCs reside in bone marrow LSK fraction. Repetitive glucose + lipid (GL) spikes, but not glucose (G) or lipid (L) spikes alone, induced senescence-like phenotypes of LSK cells, and this phenomenon was reversible after cessation of GL spikes. G spikes and GL spikes differentially affected transcriptional program of LSK cell metabolism and differentiation. GL spikes upregulated a histone H3K27 demethylase JMJD3, and inhibition of JMJD3 eliminated GL spikes-induced LSK cell senescence-like phenotypes. These observations suggest that postprandial glucose/TG dysmetabolism modulate transcriptional regulation in LSK cells through H3K27 demethylase-mediated epigenetic regulation, leading to senescence-like phenotypes of LSK cells, reduced number of circulating EPCs, and development of atherosclerotic cardiovascular disease.NEW & NOTEWORTHY Combination of hyperglycemia and hypertriglyceridemia is associated with increased risk of atherosclerotic cardiovascular disease. We found that 1) hypertriglyceridemia may enhance the negative impact of hyperglycemia on circulating EPC number in humans and 2) metabolic stress induced by glucose + triglyceride spikes in mice results in senescence-like phenotypes of bone marrow stem/progenitor cells via H3K27me3 demethylase-mediated epigenetic regulation. These findings have important implications for understanding the pathogenesis of atherosclerotic cardiovascular disease in patients with T2DM.


Subject(s)
Blood Glucose/metabolism , Bone Marrow Cells/enzymology , Cellular Senescence , DNA Methylation , Diabetes Mellitus, Type 2/blood , Endothelial Progenitor Cells/enzymology , Epigenesis, Genetic , Hyperglycemia/blood , Hypertriglyceridemia/blood , Jumonji Domain-Containing Histone Demethylases/metabolism , Triglycerides/blood , Adult , Aged , Animals , Bone Marrow Cells/pathology , Case-Control Studies , Cell Lineage , Cells, Cultured , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Endothelial Progenitor Cells/pathology , Female , Glycated Hemoglobin , Humans , Hyperglycemia/enzymology , Hyperglycemia/genetics , Hyperglycemia/pathology , Hypertriglyceridemia/enzymology , Hypertriglyceridemia/genetics , Hypertriglyceridemia/pathology , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice, Inbred C57BL , Middle Aged , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...