circulating angiogenic cells
Recently Published Documents


TOTAL DOCUMENTS

113
(FIVE YEARS 18)

H-INDEX

21
(FIVE YEARS 2)

2021 ◽  
Vol 42 (12) ◽  
pp. 1139-1139
Author(s):  
William S. Evans ◽  
Ryan M. Sapp ◽  
Katherine Kim ◽  
James M. Heilman ◽  
James M. Hagberg ◽  
...  

Biomedicines ◽  
2021 ◽  
Vol 9 (9) ◽  
pp. 1147
Author(s):  
Lucía Beltrán-Camacho ◽  
Margarita Jiménez-Palomares ◽  
Ismael Sanchez-Gomar ◽  
Antonio Rosal-Vela ◽  
Marta Rojas-Torres ◽  
...  

Critical limb ischemia (CLI), the most severe form of peripheral artery disease, results from the blockade of peripheral vessels, usually correlated to atherosclerosis. Currently, endovascular and surgical revascularization strategies cannot be applied to all patients due to related comorbidities, and even so, most patients require re-intervention or amputation within a year. Circulating angiogenic cells (CACs) constitute a good alternative as CLI cell therapy due to their vascular regenerative potential, although the mechanisms of action of these cells, as well as their response to pathological conditions, remain unclear. Previously, we have shown that CACs enhance angiogenesis/arteriogenesis from the first days of administration in CLI mice. Also, the incubation ex vivo of these cells with factors secreted by atherosclerotic plaques promotes their activation and mobilization. Herein, we have evaluated the long-term effect of CACs administration in CLI mice, whether pre-stimulated or not with atherosclerotic factors. Remarkably, mice receiving CACs and moreover, pre-stimulated CACs, presented the highest blood flow recovery, lower progression of ischemic symptoms, and decrease of immune cells recruitment. In addition, many proteins potentially involved, like CD44 or matrix metalloproteinase 9 (MMP9), up-regulated in response to ischemia and decreased after CACs administration, were identified by a quantitative proteomics approach. Overall, our data suggest that pre-stimulation of CACs with atherosclerotic factors might potentiate the regenerative properties of these cells in vivo.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Guy S. Taylor ◽  
Andy Shaw ◽  
Kieran Smith ◽  
Tess E. Capper ◽  
Jadine H. Scragg ◽  
...  

AbstractExercise mobilizes angiogenic cells, which stimulate vascular repair. However, limited research suggests exercise-induced increase of endothelial progenitor cell (EPCs) is completely lacking in type 1 diabetes (T1D). Clarification, along with investigating how T1D influences exercise-induced increases of other angiogenic cells (hematopoietic progenitor cells; HPCs) and cell surface expression of chemokine receptor 4 (CXCR4) and 7 (CXCR7), is needed. Thirty T1D patients and 30 matched non-diabetes controls completed 45 min of incline walking. Circulating HPCs (CD34+, CD34+CD45dim) and EPCs (CD34+VEGFR2+, CD34+CD45dimVEGFR2+), and subsequent expression of CXCR4 and CXCR7, were enumerated by flow cytometry at rest and post-exercise. Counts of HPCs, EPCs and expression of CXCR4 and CXCR7 were significantly lower at rest in the T1D group. In both groups, exercise increased circulating angiogenic cells. However, increases was largely attenuated in the T1D group, up to 55% lower, with CD34+ (331 ± 437 Δcells/mL vs. 734 ± 876 Δcells/mL p = 0.048), CD34+VEGFR2+ (171 ± 342 Δcells/mL vs. 303 ± 267 Δcells/mL, p = 0.006) and CD34+VEGFR2+CXCR4+ (126 ± 242 Δcells/mL vs. 218 ± 217 Δcells/mL, p = 0.040) significantly lower. Exercise-induced increases of angiogenic cells is possible in T1D patients, albeit attenuated compared to controls. Decreased mobilization likely results in reduced migration to, and repair of, vascular damage, potentially limiting the cardiovascular benefits of exercise.Trial registration: ISRCTN63739203.


Circulation ◽  
2020 ◽  
Vol 142 (Suppl_3) ◽  
Author(s):  
Lexiao Jin ◽  
Jordan Lynch ◽  
Andre Richardson ◽  
Daniel J Conklin

Introduction: After a decade of electronic cigarettes (E-cig) in the U.S., 3 elements remain: 1) E-cigs evolve; 2) E-cigs use propylene glycol (PG) and vegetable glycerin (VG); and, 3) uncertainty about E-cig use and CVD risk. Because all E-cigs use PG:VG, we tested for vascular effects of PG:VG-derived aerosol alone. Methods: Female C57BL/6 mice were exposed by inhalation (6h/d*4d) to filtered air (n=10) or to PG:VG-derived aerosol (n=10). After exposures, blood and aortas were examined for changes in: circulating immune and stem cells and platelets by flow cytometry, and vascular function by isometric myography. Results: PG:VG exposure significantly decreased WBC (-47±7%); increased RBC (+6±1%) and hemoglobin (+4±1%), but had no effect on circulating angiogenic cells or platelet-leukocyte aggregates. In aorta, vascular smooth muscle (VSM) function measured as aortic contractility to phenylephrine (PE) was unaltered. However, acetylcholine (ACh)-induced, endothelium-dependent relaxation was significantly decreased (air: -75.5±4.2%; PG:VG: -61.8±4.2%; P<0.05), whereas sensitivity of ACh-induced relaxation was variably depressed (air EC 50 : 166±54nM; PG:VG EC 50 : 1020±435nM; 0.05<P<0.10). Because loss of ACh-induced relaxation may reflect VSM desensitization to NO, we measured relaxation of NO donor, sodium nitroprusside (SNP). Both sensitivity and efficacy of SNP-induced relaxations were unchanged in the PG:VG group (EC 50 : 8.8±0.8nM; % relaxation: -99.0±0.3%) vs air group (EC 50 : 11.1±1.0nM; % relaxation: -99.1±0.3%) indicating a loss of ACh responsiveness was independent of VSM. To test whether eNOS was altered by PG:VG, PE response was done in presence of eNOS inhibitor (LNAME). The ‘PE contraction ratio’ (i.e., Tension PE+LNAME / Tension PE alone ) was significantly decreased in PG:VG group (1.28±0.03) vs air group (1.39±0.20; P<0.05) re-enforcing that PG:VG depressed eNOS function and/or NO bioavailability. Conclusions: Acute exposure to E-cig solvent aerosol alone led to endothelial dysfunction and immune cell changes suggesting that E-cig use may increase CVD risk independently of nicotine and flavor constituents. Perhaps thermal degradation of PG and VG into aldehydes may contribute to endothelial dysfunction.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii215-ii215
Author(s):  
Karin Huizer ◽  
Andrea Sacchetti ◽  
Sigrid Swagemakers ◽  
Peter van der Spek ◽  
Dana Mustafa ◽  
...  

Abstract In order to identify suitable therapeutic targets for glioma anti-angiogenic therapy, the process of neovascularization mediated by circulating angiogenic cells (CACs) needs to be scrutinized. In the present study we compared the expression of neovascularization-related genes by three circulating CAC subsets (HPCs, CD34+ and KDR+ cells; internal controls: PBMCs and circulating endothelial cells) of treatment-naïve patients with glioblastoma (GBM) to those of patients undergoing reactive neovascularization (myocardial infarction (MI). CACs from umbilical cord (representing developmental neovascularization) and healthy subjects served as controls. Fluorescent activated cell sorting was used to isolate CACs, RT-PCR to determine the expression levels of a panel of 48 neovascularization-related genes, Luminex assays to measure plasma levels of 21 CAC-related circulating molecules. We found essential differences in gene expression between GBM and MI CACs. GBM CACs had a higher expression of pro-angiogenic factors (esp. KITL, CXCL12 and JAG1); growth factor and chemotactic receptors (IGF1R, TGFbR2, CXCR4 and CCR2); adhesion receptor monomers (ITGA5 and ITGA6) and matricellular factor POSTN. In addition, we found major differences in the levels of neovascularization-related plasma factors. A strong positive correlation between plasma MMP9 levels and expression of CXCR4 in the CAC subset of hematopoietic progenitor cells (HPCs) was found in GBM patients. Our findings indicate that CAC-mediated neovascularization in GBM is characterized by more efficient CAC homing to target tissue and a more potent pro-angiogenic response than in physiologic tissue repair in MI. Our findings can aid in selecting targets for therapeutic strategies acting against GBM-specific CACs.


Author(s):  
William S. Evans ◽  
Ryan M. Sapp ◽  
Katherine I. Kim ◽  
James M. Heilman ◽  
James Hagberg ◽  
...  

AbstractExercise training has various benefits on cardiovascular health, and circulating angiogenic cells have been proposed as executing these changes. Work from the late 1990s supported an important role of these circulating post-natal cells in contributing to the maintenance and repair of the endothelium and vasculature. It was later found that circulating angiogenic cells were a heterogenous population of cells and primarily functioned in a paracrine manner by adhering to damaged endothelium and releasing growth factors. Many studies have discovered novel circulating angiogenic cell secreted proteins, microRNA and extracellular vesicles that mediate their angiogenic potential, and some studies have shown that both acute and chronic aerobic exercise training have distinct benefits. This review highlights work establishing an essential role of secreted factors from circulating angiogenic cells and summarizes studies regarding the effects of exercise training on these factors. Finally, we highlight the various gaps in the literature in hopes of guiding future work.


2020 ◽  
Vol 21 (15) ◽  
pp. 5256
Author(s):  
Sara Eslava-Alcon ◽  
Mª Jesús Extremera-García ◽  
Ismael Sanchez-Gomar ◽  
Lucía Beltrán-Camacho ◽  
Antonio Rosal-Vela ◽  
...  

In atherosclerosis, circulating angiogenic cells (CAC), also known as early endothelial progenitor cells (eEPC), are thought to participate mainly in a paracrine fashion by promoting the recruitment of other cell populations such as late EPC, or endothelial colony-forming cells (ECFC), to the injured areas. There, ECFC replace the damaged endothelium, promoting neovascularization. However, despite their regenerative role, the number and function of EPC are severely affected under pathological conditions, being essential to further understand how these cells react to such environments in order to implement their use in regenerative cell therapies. Herein, we evaluated the effect of direct incubation ex vivo of healthy CAC with the secretome of atherosclerotic arteries. By using a quantitative proteomics approach, 194 altered proteins were identified in the secretome of pre-conditioned CAC, many of them related to inhibition of angiogenesis (e.g., endostatin, thrombospondin-1, fibulins) and cell migration. Functional assays corroborated that healthy CAC released factors enhanced ECFC angiogenesis, but, after atherosclerotic pre-conditioning, the secretome of pre-stimulated CAC negatively affected ECFC migration, as well as their ability to form tubules on a basement membrane matrix assay. Overall, we have shown here, for the first time, the effect of atherosclerotic factors over the paracrine role of CAC ex vivo. The increased release of angiogenic inhibitors by CAC in response to atherosclerotic factors induced an angiogenic switch, by blocking ECFC ability to form tubules in response to pre-conditioned CAC. Thus, we confirmed here that the angiogenic role of CAC is highly affected by the atherosclerotic environment.


2020 ◽  
Vol 105 (8) ◽  
pp. 1408-1419
Author(s):  
William S. Evans ◽  
Erik D. Hanson ◽  
Daniel D. Shill ◽  
Rian Q. Landers‐Ramos ◽  
Lee Stoner ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document